US20160235758A1 - Combination of mtor inhibitors and pi3-kinase inhibitors, and uses thereof - Google Patents

Combination of mtor inhibitors and pi3-kinase inhibitors, and uses thereof Download PDF

Info

Publication number
US20160235758A1
US20160235758A1 US15/041,172 US201615041172A US2016235758A1 US 20160235758 A1 US20160235758 A1 US 20160235758A1 US 201615041172 A US201615041172 A US 201615041172A US 2016235758 A1 US2016235758 A1 US 2016235758A1
Authority
US
United States
Prior art keywords
alkyl
aryl
kinase
alkynyl
alkenyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/041,172
Inventor
Pingda Ren
Yi Liu
Katayoun Jessen
Xin Guo
Christian Rommel
Troy Edward Wilson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Intellikine LLC
Original Assignee
Intellikine LLC
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 Intellikine LLC filed Critical Intellikine LLC
Priority to US15/041,172 priority Critical patent/US20160235758A1/en
Assigned to INTELLIKINE LLC reassignment INTELLIKINE LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: WILSON, TROY EDWARD, ROMMEL, CHRISTIAN, JESSEN, Katayoun, LIU, YI, REN, PINGDA, GUO, XIN
Publication of US20160235758A1 publication Critical patent/US20160235758A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • 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
    • A61K31/498Pyrazines or piperazines ortho- and peri-condensed with carbocyclic ring systems, e.g. quinoxaline, phenazine
    • 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines 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
    • A61K31/395Heterocyclic 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
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • 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
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with 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
    • A61K31/395Heterocyclic 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
    • A61K31/527Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim spiro-condensed
    • 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
    • A61K31/53Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with three nitrogens as the only ring hetero atoms, e.g. chlorazanil, melamine
    • 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
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/541Non-condensed thiazines 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
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • kinases signaling pathways play a central role in numerous biological processes. Defects in various components of signal transduction pathways have been found to account for a vast number of diseases, including numerous forms of cancer, inflammatory disorders, metabolic disorders, vascular and neuronal diseases (Gaestel et al. Current Medicinal Chemistry (2007) 14:2214-2234). In recent years, kinases that are associated with oncogenic signaling pathways have emerged as important drug targets in the treatment of various diseases including many types of cancers.
  • rapamycin also known as mechanistic target of rapamycin
  • mTOR is a serine/threonine protein kinase that regulates cell growth, translational control, angiogenesis and/or cell survival.
  • mTOR is encoded by the FK506 binding protein 12-rapamycin associated protein 1 (FRAP1) gene.
  • FRAP1 FK506 binding protein 12-rapamycin associated protein 1
  • mTOR is the catalytic subunit of two complexes, mTORC1 and mTORC2.
  • mTORC1 is composed of mTOR, regulatory associated protein of mTOR (Raptor), mammalian LST8/G-protein ⁇ -subunit like protein (mLST8/G ⁇ L), PRAS40, and DEPTOR.
  • mTOR Complex 2 (mTORC2) is composed of mTOR, rapamycin-insensitive companion of mTOR (Rictor), G ⁇ L, and mammalian stress-activated protein kinase interacting protein
  • mTORC1 and mTORC2 are distinguished by their differential sensitivities to rapamycin and its analogs (also known as rapalogs). Rapamycin binds to and allosterically inhibits mTORC1, but mTORC2 is generally rapamycin-insensitive. As a result of this rapamycin-insensitive mTOR signaling mediated by mTORC2, cancer cells treated with rapamycin analogs usually display only partial inhibition of mTOR signaling, which can lead to enhanced survival and resistance to rapamycin treatment.
  • PI 3-kinases Phosphatidylinositol 3-kinases
  • PI3Ks Phosphatidylinositol 3-kinases
  • These lipid kinases phosphorylate the 3-position hydroxyl group of the inositol ring of phosphatidylinositol (PtdIns), activating signaling cascades associated with such processes as cell growth, proliferation, differentiation, motility, survival and intracellular trafficking.
  • Disruption of these processes involving PI3K leads to many diseases including cancer, allergic contact dermatitis, rheumatoid arthritis, osteoarthritis, inflammatory bowel diseases, chronic obstructive pulmonary disorder, psoriasis, multiple sclerosis, asthma, disorders related to diabetic complications, and inflammatory complications of the cardiovascular system such as acute coronary syndrome.
  • the PI3K family comprises 15 kinases with distinct substrate specificities, expression patterns, and modes of regulation.
  • the class I PI3Ks (p110 ⁇ , p110 ⁇ , p110 ⁇ , and p110 ⁇ ) are typically activated by tyrosine kinases or G-protein coupled receptors to generate phosphatidylinositol-3,4,5-trisphosphate (PIP 3 ), which engages downstream effectors such as those in the Akt/PDK1 pathway, mTOR, the Tec family kinases, and the Rho family GTPases.
  • the alpha ( ⁇ ) isoform of type I PI3K has been implicated in a variety of human cancers.
  • Angiogenesis has been shown to selectively require the ⁇ isoform of PI3K in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453; 662-6). Mutations in the gene coding for PI3K ⁇ or mutations which lead to upregulation of PI3K a are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers.
  • mutations in the gene coding for PI3K ⁇ are point mutations clustered within several hotspots in helical and kinase domains, such as E542K, E545K, and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. While other PI3K isoforms such as PI3K ⁇ or PI3K ⁇ are expressed primarily in hematopoietic cells, PI3K ⁇ , along with PI3K ⁇ , is expressed constitutively.
  • the delta ( ⁇ ) isoform of class I PI3K has been implicated, in particular, in a number of diseases and biological processes.
  • PI3K ⁇ is expressed primarily in hematopoietic cells including leukocytes such as T-cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages.
  • PI3K ⁇ is integrally involved in mammalian immune system functions such as T-cell function, B-cell activation, mast cell activation, dendritic cell function, and neutrophil activity.
  • PI3K ⁇ Due to its integral role in immune system function, PI3K ⁇ is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases.
  • Other class I PI3K involved in immune system function includes PI3K ⁇ , which plays a role in leukocyte signaling and has been implicated in inflammation, rheumatoid arthritis, and autoimmune diseases such as lupus.
  • PI3K ⁇ has been implicated primarily in various types of cancer including PTEN-negative cancer (Edgar et al. Cancer Research (2010) 70(3): 1164-1172), and HER 2 -overexpressing cancer such as breast cancer and ovarian cancer.
  • drugs that bind to and inhibit a broad range of kinase isoforms and complexes with low specificity can lead to deleterious side effects.
  • excessive inhibition of PI3K ⁇ may lead to undesirable effects on metabolic pathways and disruption of insulin signaling.
  • excessive inhibition of PI3K ⁇ and/or PI3K ⁇ may disrupt or reduce immune function.
  • the present disclosure provides an alternative approach that effectively targets disease-related pathways, while limiting undesirable side effects.
  • the invention provides a method for treating a disease condition associated with PI3-kinase ⁇ and/or mTOR in a subject, comprising administering to said subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor, wherein the PI3-kinase ⁇ inhibitor exhibits selective inhibition of PI3-kinase ⁇ relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) ascertained by an in vitro kinase assay, wherein the one or more type I PI3-kinase is selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • PI3-kinase type I phosphatidylinositol 3-kinases
  • the combination comprises a therapeutic effective amount of a PI3-kinase ⁇ inhibitor and a therapeutic effective amount of an mTOR inhibitor.
  • the combination comprises a synergistically effective therapeutic amount of PI3-kinase ⁇ inhibitor and an mTOR inhibitor, wherein the PI3-kinase ⁇ inhibitor and/or the mTOR inhibitor is present in a sub-therapeutic amount.
  • the disease condition associated with PI3-kinase ⁇ and/or mTOR can include but are not limited to a neoplastic condition, autoimmune disease, inflammatory disease, fibrotic disease and kidney disease.
  • the neoplastic condition can be NSCLC, head and neck squamous cell carcinoma, pancreatic, breast and ovarian cancers, renal cell carcinoma, prostate cancer, neuroendocrine cancer, endometrial cancers, and other forms of cancer.
  • the invention further provides a method of inhibiting phosphorylation of both Akt (S473) and Akt (T308) in a cell, comprising contacting a cell with an effective amount of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor that selectively inhibits both mTORC1 and mTORC2 activity relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) as ascertained by a cell-based assay or an in vitro kinase assay, wherein the PI3-kinase ⁇ inhibitor exhibits selective inhibition of PI3-kinase ⁇ relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) ascertained by an in vitro kinase assay, wherein the one or more type I PI3-kinase is selected from the group consisting of PI3-kinase ⁇ , PI3-
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ relative to all other type I phosphatidylinositol 3-kinases (PI3-kinase) consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • PI3-kinase all other type I phosphatidylinositol 3-kinases
  • the PI3-kinase ⁇ inhibitor utilized in the subject methods inhibits PI3-kinase ⁇ with an IC50 value of about 500 nM or less, 400 nM or less, 300 nM or less, 200 nM or less, 100 nM or less, 10 nM or less, 1 nM or less as ascertained in an in vitro kinase assay.
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is at least 2, 5, 10, 50, 100, 1000 times less than its IC50 value against one, two, three or all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is less than about 200 nM, and said IC50 value is at least 2, 5 or 10 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ and/or PI3-kinase ⁇ with an IC50 value that is at least 5 times less than its IC50 value against PI3-kinase ⁇ or PI3-kinase ⁇ . In yet other embodiments, the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ and/or PI3-kinase ⁇ with an IC50 value that is at least 50 times less than its IC50 value against PI3-kinase ⁇ or PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is at least 50 times less than its IC50 value against PI3-kinase ⁇ or PI3-kinase ⁇ .
  • the mTOR inhibitor binds to and directly inhibits both mTORC1 and mTORC2.
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 500 nM or less, 400 nM or less, 300 nM or less, 200 nM or less, 100 nM or less, 50 nM or less, 10 nM or less, or 1 nM or less, as ascertained in an in vitro kinase assay.
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 10 nM or less as ascertained in an in vitro kinase assay, and the mTOR inhibitor is substantially inactive against one or more types I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay, and the IC50 value is at least 2, 5 or 10 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the mTor inhibitor inhibits mTORC1 selectively.
  • the mTor inhibitor inhibits mTORC1 with an IC50 value of about 1000 nM or less, 500 nM or less, 100 nM or less, 50 nM or less, 10 nM or less, as ascertained in an in vitro kinase.
  • the mTor inhibitor is rapamycin or an analogue of rapamycin.
  • the mTor inhibitor is sirolimus (rapamycin), deforolimus (AP23573, MK-8669), everolimus (RAD-001), temsirolimus (CCI-779), zotarolimus (ABT-578), or biolimus A9 (umirolimus).
  • the mTOR inhibitor is a compound of Formula I:
  • X 1 is N or C-E 1
  • X 2 is N or C
  • X 3 is N or C
  • X 4 is C—R 9 or N
  • X 5 is N or C-E 1
  • X 6 is C or N
  • X 7 is C or N; and wherein no more than two nitrogen ring atoms are adjacent;
  • R 1 is H, -L-C 1-10 alkyl, -L-C 3-8 cycloalkyl, —C 3-8 cycloalkyl, -L-aryl, -L-heteroaryl, -L-C 1-10 alkylaryl, -L-C 1-10 alkylhetaryl, -L-C 1-10 alkylheterocylyl, -L-C 2-10 alkenyl, -L-C 2-10 alkynyl, -L-C 2-10 alkenyl-C 3-8 cycloalkyl, -L-C 2-10 alkynyl-C 3-8 cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocylyl, -L-heteroalkyl-C 3-8 cycloalkyl,
  • L is absent, —(C ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O) N(R 31 )—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 N(R 31 )—, or —N(R 31 )—;
  • E 1 and E 2 are independently —(W 1 ) j —R 4 ;
  • M 1 is a 5, 6, 7, 8, 9, or -10 membered ring system, wherein the ring system is monocyclic or bicyclic, substituted with R 5 and additionally optionally substituted with one or more —(W 2 ) k —R 2 ;
  • each k is 0 or 1;
  • j in E 1 or j in E 2 is independently 0 or 1;
  • W 1 is —O—, —NR 7 —, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(R 7 )N(R 8 )S(O
  • W 2 is —O—, —NR 7 —, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )C(O)N(R 8 )—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )S(O)——, —CH(R 7 )N(R 8 )S(O
  • R 2 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • R 3 and R 4 are independently hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 C( ⁇ O) NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 , —
  • R 5 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • each of R 31 , R 32 , and R 33 is independently H or C 1-10 alkyl, wherein the C 1-10 alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or hetaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, hetaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • each of R 7′ and R 8′ is independently hydrogen, C 1-10 alkyl, C 2-10 alkenyl, aryl, heteroaryl, heterocyclyl or C 3-10 cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R 6 ;
  • R 6 is halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, hetaryl-C 1-10 alkyl, hetaryl-C 2-10 alkenyl, hetaryl-C 2-10 alkynyl, wherein each of said alkyl, alkeny
  • R 9 is H, halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, hetaryl-C 1-10 alkyl, hetaryl-C 2-10 alkenyl, hetaryl-C 2-10 alkynyl, wherein each of said alkyl, al
  • the PI3-kinase ⁇ inhibitor is a compound of formula:
  • W 1′ is N, NR 3′ , or CR 3′ ;
  • W 2 is N, NR 4′ , CR 4′ , or C ⁇ O;
  • W 3′ is N, NR 5 or CR 5 ;
  • W 4′ is N, wherein no more than two N atoms and no more than two C ⁇ O groups are adjacent;
  • W 5′ is N
  • W 6 is N or CR 8′ ;
  • W a′ and W b′ are independently N or CR 9′ ;
  • one of W e′ and W d′ is N, and the other is O, NR 10′ , or S;
  • R 1′ and R 2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R′′ wherein R′ and R′′ are taken together with nitrogen to form a cyclic moiety;
  • R 3′ and R 4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R′′ wherein R′ and R′′ are taken together with nitrogen to form a cyclic moiety;
  • R 5′ , R 6′ , R 7′ and R 8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R′′ wherein R′ and R′′ are taken together with nitrogen to form a cyclic moiety;
  • R 9′ is alkyl or halo
  • R 10′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R′′ wherein R′ and R′′ are taken together with nitrogen to form a cyclic moiety.
  • the PI3-kinase ⁇ inhibitor is a compound of formula:
  • X is O or S or N
  • W 1′ is S, N, NR 3′ or CR 3′
  • W 2′ is N or CR 4′
  • W 3′ is S, N or CR 5′
  • W 4′ is N or C
  • W 7′ is N or C, wherein no more than two N atoms and no more than two C ⁇ O groups are adjacent;
  • W 5′ is N or CR 7′ ;
  • W 6′ is N or CR 8′ ;
  • R 1′ and R 2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R′′ wherein R′ and R′′ are taken together with nitrogen to form a cyclic moiety;
  • R 3′ and R 4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R′′ wherein R′ and R′′ are taken together with nitrogen to form a cyclic moiety;
  • R 5′ , R 7′ and R 8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R′′ wherein R′ and R′′ are taken together with nitrogen to form a cyclic moiety.
  • the PI3-kinase ⁇ inhibitor and/or the mTOR inhibitor can be administered parenterally, orally, intraperitoneally, intravenously, intraarterially, transdermally, intramuscularly, liposomally, via local delivery by catheter or stent, subcutaneously, intraadiposally, or intrathecally.
  • the PI3-kinase ⁇ inhibitor and/or the mTOR inhibitor are co-administered to the subject in the same formulation.
  • the PI3-kinase ⁇ inhibitor and/or the mTOR inhibitor are co-administered to the subject in different formulations.
  • the invention also provides a pharmaceutical composition comprising a combination of an amount of PI3-kinase ⁇ inhibitor and an amount of mTOR inhibitor, wherein said combination provides a synergistic therapeutic effect in a subject in need thereof.
  • the pharmaceutical composition is formulated in an oral dosage.
  • at least one of the amounts is administered as a sub-therapeutic amount.
  • the pharmaceutical composition is formulated a tablet or a capsule.
  • the PI3-kinase ⁇ inhibitor and the mTOR inhibitor are packaged as separate tablets.
  • the PI3-kinase ⁇ inhibitor and the mTOR inhibitor are formulated as a single oral dosage form.
  • the invention also provides a method comprising: (a) determining the presence in a subject of a mutation in PI3-kinase ⁇ that is associated with a disease condition mediated by PI3-kinase ⁇ ; and (b) administering to said subject a pharmaceutical composition of the invention.
  • the mutation is in the nucleotide sequence coding for PI3-kinase ⁇ .
  • Exemplary mutations can include without limitation, deletion, insertion, translation, which can result in point mutations, frame shifts, and/or translation of the nucleic acid sequence coding for PI3-kinase ⁇ .
  • the mutation is in the amino acid sequence of PI3-kinase ⁇ .
  • the subject or cell comprises a mutation in PI3-kinase ⁇ which is associated with a disease condition mediated by PI3-kinase ⁇ .
  • FIG. 1 is a schematic illustration of multiple and distinct signaling pathways that are activated in human cancer.
  • FIG. 2 is a graph showing the synergistic effect of combined treatment with a PI3-kinase ⁇ inhibitor (Compound A) and an mTor inhibitor (Compound B) on tumor weight in a preclinical breast cancer model.
  • FIG. 3 is a western blot depicting the synergistic effect of combined treatment with Compound A and Compound B in terms of downregulating Akt and S6 phosphorylation.
  • FIG. 4 is a graph showing the synergistic effect of combined treatment with Compound A and rapamycin in terms of reducing tumor volume of a preclinical breast cancer model.
  • FIG. 5 shows A) an illustration of the distinct signaling pathways mediated by mTORC1 and mTORC2 and B) a western blot depicting sensitivity of mTORC1-dependent NRDG1 phosphorylation to Compound B, but not rapamycin.
  • FIG. 6 shows A) a graph depicting the selectivity of Compound B over Compound A in a PTEN-mutant negative control tumor model, B) a western blot depicting selective inhibition of Akt, S6, and 4EBP phosphorylation by Compound B and not Compound A in PTEN mutant cells, and C) a chart depicting the specificity of Compound A in inhibiting PI3K a over mTOR and other PI3K isoforms, and the specificity of Compound B in inhibiting mTOR over PI3K isoforms.
  • FIG. 7 is a western blot depicting differential inhibition of Akt phosphorylation at serine 473 over threonine 308 by Compound B (top panel). Also shown is the comparison of Akt phosphorylation inhibition for Pan-PI3K inhibitor versus Compound B.
  • FIG. 8 is a graph showing that Pan-PI3K inhibitor, but not Compound A, blocks B cell function in vivo. Mice were immunized with TNP-Ficoll and treated with 1) vehicle; 2) 70 mg/kg GDC0941; 3) 30 mg/kg Compound A; 4) 60 mg/kg Compound A; or 5) 120 mg/kg Compound A for 7 days. Antibody production was measured as a percentage of control group that were treated with vehicle.
  • FIG. 9 shows, left panel, a graph showing reduction in tumor weight of a breast cancer model using 70 mg/kg Pan-PI3K inhibitor and 60 mg/kg compound A and, right panel, reduced presence of MZB cells in mouse spleen for 70 mg/kg Pan-PI3K inhibitor compared to 60 mg/kg Compound A.
  • FIG. 10 illustrates the frequency of PI3K a mutation in various human cancers.
  • FIG. 11 is a western blot depicting inhibition of the PI3K pathway by Compound A in cell lines with elevated PI3K a activity.
  • the left column shows data from MDA-MB-361 breast cancer cells harboring PIK3CA mutation.
  • the middle column shows data from MDA-MB-453 breast cancer cells harboring PIK3CA mutation.
  • the right column shows data from SKBr3breast cancer cells harboring HER 2 mutation.
  • FIG. 12 shows A) a western blot showing inhibition of Akt phosphorylation at serine 473 by Compound A; and B) reduced inhibition of Akt phosphorylation at serine 473 by Compound A in a PTEN-mutant cell line.
  • FIG. 13 is a chart showing that Compound A preferentially inhibits proliferation of tumor cells harboring PI3K a mutations.
  • the term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term “about” meaning within an acceptable error range for the particular value should be assumed.
  • Treatment “Treatment”, “treating”, “palliating” and “ameliorating”, as used herein, are used interchangeably. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit.
  • therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated.
  • a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder.
  • the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • the term “neoplastic condition” refers to the presence of cells possessing abnormal growth characteristics, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, perturbed oncogenic signaling, and certain characteristic morphological features. This includes the abnormal growth of: (1) tumor cells (tumors) that proliferate by expressing a mutated tyrosine kinase or overexpression of a receptor tyrosine kinase; (2) benign and malignant cells of other proliferative diseases in which aberrant tyrosine kinase activation occurs; (3) any tumors that proliferate by receptor tyrosine kinases; (4) any tumors that proliferate by aberrant serine/threonine kinase activation; and (5) benign and malignant cells of other proliferative diseases in which aberrant serine/threonine kinase activation occurs.
  • the term “effective amount” or “therapeutically effective amount” refers to that amount of an inhibitor described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below.
  • the therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or downregulation of activity of a target protein.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • a “sub-therapeutic amount” of an agent or therapy is an amount less than the effective amount for that agent or therapy, but when combined with an effective or sub-therapeutic amount of another agent or therapy can produce a result desired by the physician, due to, for example, synergy in the resulting efficacious effects, or reduced side effects.
  • a “synergistically effective therapeutic amount” of an agent or therapy is an amount which, when combined with an effective or sub-therapeutic amount of another agent or therapy, produces a greater effect than when either of the two agents are therapies are used alone.
  • a syngergistically effective therapeutic amount of an agent or therapy produces a greater effect when used in combination than the additive effects of each of the two agents or therapies when used alone.
  • agent refers to a biological, pharmaceutical, or chemical compound or other moiety.
  • Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound.
  • Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures.
  • various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • agonist refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term “agonist” is defined in the context of the biological role of the target polypeptide. While preferred agonists herein specifically interact with (e.g., bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition.
  • antagonists are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the terms “antagonist” and “inhibitors” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition.
  • a preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor, or an undesired immune response as manifested in autoimmune disease.
  • mTOR inhibitor that binds to and directly inhibits both mTORC1 and mTORC2 kinases refers to an mTOR inhibitor that interacts with and reduces the kinase activity of both mTORC1 and mTORC2 complexes.
  • an “anti-cancer agent”, “anti-tumor agent” or “chemotherapeutic agent” refers to any agent useful in the treatment of a neoplastic condition.
  • One class of anti-cancer agents comprises chemotherapeutic agents.
  • “Chemotherapy” means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
  • cell proliferation refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
  • co-administration encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time.
  • Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present.
  • Co-administered agents may be in the same formulation.
  • Co-administered agents may also be in different formulations.
  • a “therapeutic effect,” as used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above.
  • a prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • pharmaceutically acceptable salt refers to salts derived from a variety of organic and inorganic counter ions well known in the art.
  • Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids.
  • Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like.
  • Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.
  • Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases.
  • Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like.
  • Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine.
  • the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
  • “Pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions of the invention is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • Signal transduction is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response.
  • a modulator of a signal transduction pathway refers to a compound which modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway.
  • a modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
  • selective inhibition or “selectively inhibit” as applied to a biologically active agent refers to the agent's ability to selectively reduce the target signaling activity as compared to off-target signaling activity, via direct or interact interaction with the target.
  • Subject refers to an animal, such as a mammal, for example a human.
  • the methods described herein can be useful in both human therapeutics, pre-clinical, and veterinary applications.
  • the subject is a mammal, and in some embodiments, the subject is human.
  • in vivo refers to an event that takes place in a subject's body.
  • in vitro refers to an event that takes places outside of a subject's body.
  • an in vitro assay encompasses any assay run outside of a subject assay.
  • in vitro assays encompass cell-based assays in which cells alive or dead are employed.
  • In vitro assays also encompass a cell-free assay in which no intact cells are employed.
  • PI3K Phosphoinositide 3-kinase
  • PI phosphatidylinositol
  • connection of compound name moieties are at the rightmost recited moiety. That is, the substituent name starts with a terminal moiety, continues with any linking moieties, and ends with the linking moiety.
  • heteroarylthio C 1-4 alkyl has a heteroaryl group connected through a thio sulfur to a C 1-4 alkyl radical that connects to the chemical species bearing the substituent.
  • the terminal group is a C 3-8 cycloalkyl group attached to a linking C 1-10 alkyl moiety which is attached to an element L, which is itself connected to the chemical species bearing the substituent.
  • Alkyl refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to ten carbon atoms (e.g., C 1 -C 10 alkyl). Whenever it appears herein, a numerical range such as “1 to 10” refers to each integer in the given range; e.g., “1 to 10 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated. In some embodiments, it is a C 1 -C 4 alkyl group.
  • Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl isobutyl, tertiary butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, decyl, and the like.
  • the alkyl is attached to the rest of the molecule by a single bond, for example, methyl (Me), ethyl (Et), n-propyl, 1-methylethyl (iso-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, and the like.
  • an alkyl group is optionally substituted by one or more of substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —OR a , —SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —OC(O)N(R a ) 2 , —C(O)N(R a ) 2 , —N(R a )C(O)OR a , —N(R a )C(O)OR a , —N(R a )C(O)R
  • halo or halogen refers to fluoro, chloro, bromo, or iodo.
  • haloalkyl refers to an alkyl group substituted with one or more halo groups, for example chloromethyl, 2-bromoethyl, 3-iodopropyl, trifluoromethyl, perfluoropropyl, 8-chlorononyl, and the like.
  • “Acyl” refers to the groups (alkyl)-C(O)—, (aryl)-C(O)—, (heteroaryl)-C(O)—, (heteroalkyl)-C(O)—, and (heterocycloalkyl)-C(O)—, wherein the group is attached to the parent structure through the carbonyl functionality.
  • it is a C 1 -C 10 acyl radical which refers to the total number of chain or ring atoms of the alkyl, aryl, heteroaryl or heterocycloalkyl portion of the acyloxy group plus the carbonyl carbon of acyl, i.e three other ring or chain atoms plus carbonyl.
  • R radical is heteroaryl or heterocycloalkyl
  • the hetero ring or chain atoms contribute to the total number of chain or ring atoms.
  • the “R” of an acyloxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —OR a , SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —OC(O)N(R a ) 2 , —C(O)N(R a ) 2 , —C(O)
  • Cycloalkyl refers to a monocyclic or polycyclic radical that contains only carbon and hydrogen, and may be saturated, or partially unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms (i.e., C 2 -C 10 cycloalkyl). Whenever it appears herein, a numerical range such as “3 to 10” refers to each integer in the given range; e.g., “3 to 10 carbon atoms” means that the cycloalkyl group may consist of 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C 3 -C 8 cycloalkyl radical.
  • cycloalkyl groups include, but are not limited to the following moieties: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloseptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl, and the like.
  • a cycloalkyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —OR a , —SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —OC(O)N(R a ) 2 , —C(O)N(R a ) 2 , —N(R a )C(O)OR a , —N(R a )C(O)OR a , —N(R a )C(
  • C 1-10 alkyl C 3-8 cycloalkyl is used to describe an alkyl group, branched or straight chain and containing 1 to 10 carbon atoms, attached to a linking cycloalkyl group which contains 3 to 8 carbons, such as for example, 2-methyl cyclopropyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • bicycloalkyl refers to a structure consisting of two cycloalkyl moieties, unsubstituted or substituted, that have two or more atoms in common. If the cycloalkyl moieties have exactly two atoms in common they are said to be “fused”. Examples include, but are not limited to, bicyclo[3.1.0]hexyl, perhydronaphthyl, and the like. If the cycloalkyl moieties have more than two atoms in common they are said to be “bridged”. Examples include, but are not limited to, bicyclo[3.2.1]heptyl (“norbornyl”), bicyclo[2.2.2]octyl, and the like.
  • heteroatom or “ring heteroatom” is meant to include oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
  • Heteroalkyl “heteroalkenyl” and “heteroalkynyl” include optionally substituted alkyl, alkenyl and alkynyl radicals and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof.
  • a numerical range may be given, e.g., C 1 -C 4 heteroalkyl which refers to the chain length in total, which in this example is 4 atoms long.
  • a —CH 2 OCH 2 CH 3 radical is referred to as a “C 4 ” heteroalkyl, which includes the heteroatom center in the atom chain length description.
  • a heteroalkyl group may be substituted with one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, —OR a , —SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —C(O)N(R a ) 2 , —N(R a )C(O)OR a , —N(R a )C(O)OR a , —N(R a )C(O)R a , —N(R a )C(O)OR
  • heteroalkylaryl refers to a heteroalkyl group as defined above which is attached to an aryl group, and may be attached at a terminal point or through a branched portion of the heteroalkyl, for example, an benzyloxymethyl moiety. Either portion of the moiety is unsubstituted or substituted.
  • heteroalkylheteroaryl refers likewise to a heteroalkyl group which is attached to a heteroaryl moiety, for example, an ethoxymethylpyridyl group. Either portion of the moiety is unsubstituted or substituted.
  • heteroalkyl-heterocyclyl refers to a heteroalkyl group as defined above, which is attached to a heterocyclic group, for example, 4(3-aminopropyl)-N-piperazinyl. Either portion of the moiety is unsubstituted or substituted.
  • heteroalkyl-C 3-8 cycloalkyl refers to a heteroalkyl group as defined above, which is attached to a cyclic alkyl containing 3 to 8 carbons, for example, 1-aminobutyl-4-cyclohexyl. Either portion of the moiety is unsubstituted or substituted.
  • heterocycloalkyl refers to a bicycloalkyl structure, which is unsubstituted or substituted, in which at least one carbon atom is replaced with a heteroatom independently selected from oxygen, nitrogen, and sulfur.
  • heterospiroalkyl refers to a spiroalkyl structure, which is unsubstituted or substituted, in which at least one carbon atom is replaced with a heteroatom independently selected from oxygen, nitrogen, and sulfur.
  • alkene refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond
  • an “alkyne” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond.
  • the alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
  • Alkenyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to ten carbon atoms (i.e., C 2 -C 10 alkenyl). Whenever it appears herein, a numerical range such as “2 to 10” refers to each integer in the given range; e.g., “2 to 10 carbon atoms” means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkenyl comprises two to eight carbon atoms.
  • an alkenyl comprises two to five carbon atoms (e.g., C 2 -C 5 alkenyl).
  • the alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-1-enyl (i.e., allyl), but-1-enyl, pent-1-enyl, penta-1,4-dienyl, and the like.
  • an alkenyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —OR a , —SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —OC(O)N(R a ) 2 , —C(O)N(R a ) 2 , —N(R a )C(O)OR a , —N(R a )C(O)OR a , —N(R a )C(O)R
  • C 2-10 alkenyl-heteroalkyl refers to a group having an alkenyl moiety, containing 2 to 10 carbon atoms and is branched or straight chain, which is attached to a linking heteroalkyl group, such as, for example, allyloxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkynyl-heteroalkyl refers to a group having an alkynyl moiety, which is unsubstituted or substituted, containing 2 to 10 carbon atoms and is branched or straight chain, which is attached to a linking heteroalkyl group, such as, for example, 4-but-1-ynoxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • haloalkenyl refers to an alkenyl group substituted with one or more halo groups.
  • cycloalkenyl refers to a cyclic aliphatic 3 to 8 membered ring structure, optionally substituted with alkyl, hydroxy and halo, having 1 or 2 ethylenic bonds such as methylcyclopropenyl, trifluoromethylcyclopropenyl, cyclopentenyl, cyclohexenyl, 1,4-cyclohexadienyl, and the like.
  • Alkynyl refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to ten carbon atoms (i.e., C 2 -C 10 alkynyl). Whenever it appears herein, a numerical range such as “2 to 10” refers to each integer in the given range; e.g., “2 to 10 carbon atoms” means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkynyl comprises two to eight carbon atoms.
  • an alkynyl has two to five carbon atoms (e.g., C 2 -C 5 alkynyl).
  • the alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like.
  • an alkynyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —OR a , SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —OC(O)N(R a ) 2 , —C(O)N(R a ) 2 , —N(R a )C(O)OR a , —N(R a )C(O)R a , —N(R a )C(O)OR
  • C 2-10 alkynyl-C 3-8 cycloalkyl refers to a group containing an alkynyl group, containing 2 to 10 carbons and branched or straight chain, which is attached to a linking cycloalkyl group containing 3 to 8 carbons, such as, for example 3-prop-3-ynyl-cyclopent-1yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • haloalkynyl refers to an alkynyl group substituted with one or more independent halo groups.
  • Amino or “amine” refers to a —N(R a ) 2 radical group, where each R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification.
  • R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification.
  • R a is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl
  • —N(R a ) 2 is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl.
  • an amino group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —OR a , —SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —OC(O)N(R a ) 2 , —C(O)N(R a ) 2 , —N(R a ) 2 , —N(
  • “Amide” or “amido” refers to a chemical moiety with formula —C(O)N(R) 2 or —NHC(O)R, where R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), each of which moiety may itself be optionally substituted. In some embodiments it is a C 1 -C 4 amido or amide radical, which includes the amide carbonyl in the total number of carbons in the radical.
  • the R 2′ of —N(R) 2 of the amide may optionally be taken together with the nitrogen to which it is attached to form a 4-, 5-, 6-, or 7-membered ring.
  • an amido group is optionally substituted independently by one or more of the substituents as described herein for alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl.
  • An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be amidified.
  • “Aromatic” or “aryl” refers to an aromatic radical with six to ten ring atoms (e.g., C 6 -C 10 aromatic or C 6 -C 10 aryl) which has at least one ring having a conjugated pi electron system which is carbocyclic (e.g., phenyl, fluorenyl, and naphthyl).
  • Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals.
  • Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in “-yl” by removal of one hydrogen atom from the carbon atom with the free valence are named by adding “-idene” to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene.
  • a numerical range such as “6 to 10” refers to each integer in the given range; e.g., “6 to 10 ring atoms” means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 10 ring atoms.
  • an aryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —OR a , —SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —OC(O)N(W) 2 , —C(O)N(R a ) 2 , —N(R a )C(
  • Heteroaryl or, alternatively, “heteroaromatic” refers to a 5- to 18-membered aromatic radical (e.g., C 5 -C 13 heteroaryl) that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system.
  • a numerical range such as “5 to 18” refers to each integer in the given range; e.g., “5 to 18 ring atoms” means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms.
  • Bivalent radicals derived from univalent heteroaryl radicals whose names end in “-yl” by removal of one hydrogen atom from the atom with the free valence are named by adding “-idene” to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene.
  • An N-containing “heteroaromatic” or “heteroaryl” moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom.
  • the polycyclic heteroaryl group may be fused or non-fused.
  • the heteroatom(s) in the heteroaryl radical is optionally oxidized.
  • heteroaryl is attached to the rest of the molecule through any atom of the ring(s).
  • heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl, benzo[d]thiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, benzo[b][1,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzof
  • a heteraryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, —OR a , —SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —C(O)N(R a ) 2 , —N(R a )C(O)OR a , —N(R a )C(O)OR a , —N(R a )C(O)R a , —N(R a )S(
  • aryl-alkyl arylalkyl
  • arylalkyl arylalkyl
  • aralkyl a group wherein the alkyl chain can be branched or straight chain forming a linking portion with the terminal aryl, as defined above, of the aryl-alkyl moiety.
  • aryl-alkyl groups include, but are not limited to, optionally substituted benzyl, phenethyl, phenpropyl and phenbutyl such as 4-chlorobenzyl, 2,4-dibromobenzyl, 2-methylbenzyl, 2-(3-fluorophenyl)ethyl, 2-(4-methylphenyl)ethyl, 2-(4-(trifluoromethyl)phenyl)ethyl, 2-(2-methoxyphenyl)ethyl, 2-(3-nitrophenyl)ethyl, 2-(2,4-dichlorophenyl)ethyl, 2-(3,5-dimethoxyphenyl)ethyl, 3-phenylpropyl, 3-(3-chlorophenyl)propyl, 3-(2-methylphenyl)propyl, 3-(4-methoxyphenyl)propyl, 3-(4-(trifluoromethyl)phenyl)propyl
  • C 1-10 alkylaryl refers to an alkyl group, as defined above, containing 1 to 10 carbon atoms, branched or unbranched, wherein the aryl group replaces one hydrogen on the alkyl group, for example, 3-phenylpropyl. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkyl monocycloaryl refers to a group containing a terminal alkyl group, branched or straight chain and containing 2 to 10 atoms attached to a linking aryl group which has only one ring, such as for example, 2-phenyl ethyl. Either portion of the moiety is unsubstituted or substituted.
  • C 1-10 alkyl bicycloaryl refers to a group containing a terminal alkyl group, branched or straight chain and containing 2 to 10 atoms attached to a linking aryl group which is bicyclic, such as for example, 2-(1-naphthyl)-ethyl. Either portion of the moiety is unsubstituted or substituted.
  • aryl-cycloalkyl and “arylcycloalkyl” are used to describe a group wherein the terminal aryl group is attached to a cycloalkyl group, for example phenylcyclopentyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • heteroaryl-C 3-8 cycloalkyl and “heteroaryl-C 3-8 cycloalkyl” are used to describe a group wherein the terminal heteroaryl group is attached to a cycloalkyl group, which contains 3 to 8 carbons, for example pyrid-2-yl-cyclopentyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • heteroaryl-heteroalkyl refers to a group wherein the terminal heteroaryl group is attached to a linking heteroalkyl group, such as for example, pyrid-2-yl methylenoxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • aryl-alkenyl arylalkenyl
  • arylalkenyl arylalkenyl
  • aralkenyl are used to describe a group wherein the alkenyl chain can be branched or straight chain forming a linking portion of the aralkenyl moiety with the terminal aryl portion, as defined above, for example styryl (2-phenylvinyl), phenpropenyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • aryl-C 2-10 alkenyl means an arylalkenyl as described above wherein the alkenyl moiety contains 2 to 10 carbon atoms such as for example, styryl (2-phenylvinyl), and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkenyl-aryl is used to describe a group wherein the terminal alkenyl group, which contains 2 to 10 carbon atoms and can be branched or straight chain, is attached to the aryl moiety which forms the linking portion of the alkenyl-aryl moiety, such as for example, 3-propenyl-naphth-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • aryl-alkynyl arylalkynyl
  • arylalkynyl arylalkynyl
  • aralkynyl are used to describe a group wherein the alkynyl chain can be branched or straight chain forming a linking portion of the aryl-alkynyl moiety with the terminal aryl portion, as defined above, for example 3-phenyl-1-propynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • aryl-C 2-10 alkynyl means an arylalkynyl as described above wherein the alkynyl moiety contains two to ten carbons, such as, for example 3-phenyl-1-propynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkynyl-aryl means a group containing an alkynyl moiety attached to an aryl linking group, both as defined above, wherein the alkynyl moiety contains two to ten carbons, such as, for example 3-propynyl-naphth-1-yl. Either portion of the moiety is unsubstituted or substituted.
  • aryl-oxy aryloxy
  • aryloxy and “aroxy” are used to describe a terminal aryl group attached to a linking oxygen atom.
  • Typical aryl-oxy groups include phenoxy, 3,4-dichlorophenoxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • aryl-oxyalkyl refers to a group wherein an alkyl group is substituted with a terminal aryl-oxy group, for example pentafluorophenoxymethyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 1-10 alkoxy-C 1-10 alkyl refers to a group wherein an alkoxy group, containing 1 to 10 carbon atoms and an oxygen atom within the branching or straight chain, is attached to a linking alkyl group, branched or straight chain which contains 1 to 10 carbon atoms, such as, for example methoxypropyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 1-10 alkoxy-C 2-10 alkenyl refers to a group wherein an alkoxy group, containing 1 to 10 carbon atoms and an oxygen atom within the branching or straight chain, is attached to a linking alkenyl group, branched or straight chain which contains 1 to 10 carbon atoms, such as, for example 3-methoxybut-2-en-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 1-10 alkoxy-C 2-10 alkynyl refers to a group wherein an alkoxy group, containing 1 to 10 carbon atoms and an oxygen atom within the branching or straight chain, is attached to a linking alkynyl group, branched or straight chain which contains 1 to 10 carbon atoms, such as, for example 3-methoxybut-2-in-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocycloalkenyl refers to a cycloalkenyl structure, which is unsubstituted or substituted in which at least one carbon atom is replaced with a heteroatom selected from oxygen, nitrogen, and sulfur.
  • heteroaryl-oxy refers to a terminal heteroaryl group, which is unsubstituted or substituted, attached to a linking oxygen atom.
  • Typical heteroaryl-oxy groups include 4,6-dimethoxypyrimidin-2-yloxy and the like.
  • heteroarylalkyl refers to a group wherein the alkyl chain can be branched or straight chain forming a linking portion of the heteroaralkyl moiety with the terminal heteroaryl portion, as defined above, for example 3-furylmethyl, thenyl, furfuryl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heteroaryl-C 1-10 alkyl is used to describe a heteroaryl alkyl group as described above where the alkyl group contains 1 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • C 1-10 alkyl-heteroaryl is used to describe a alkyl attached to a hetary group as described above where the alkyl group contains 1 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • heteroarylalkenyl refers to a heteroarylalkenyl group wherein the alkenyl chain can be branched or straight chain forming a linking portion of the heteroaralkenyl moiety with the terminal heteroaryl portion, as defined above, for example 3-(4-pyridyl)-1-propenyl. Either portion of the moiety is unsubstituted or substituted.
  • heteroaryl-C 2-10 alkenyl group is used to describe a group as described above wherein the alkenyl group contains 2 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkenyl-heteroaryl is used to describe a group containing an alkenyl group, which is branched or straight chain and contains 2 to 10 carbon atoms, and is attached to a linking heteroaryl group, such as, for example 2-styryl-4-pyridyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heteroarylalkynyl refers to a group wherein the alkynyl chain can be branched or straight chain forming a linking portion of the heteroaralkynyl moiety with the heteroaryl portion, as defined above, for example 4-(2-thienyl)-1-butynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heteroaryl-C 2-10 alkynyl is used to describe a heteroarylalkynyl group as described above wherein the alkynyl group contains 2 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkynyl-heteroaryl is used to describe a group containing an alkynyl group which contains 2 to 10 carbon atoms and is branched or straight chain, which is attached to a linking heteroaryl group such as, for example, 4(but-1-ynyl) thien-2-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclyl refers to a four-, five-, six-, or seven-membered ring containing one, two, three or four heteroaroms independently selected from nitrogen, oxygen and sulfur.
  • the four-membered ring has zero double bonds
  • the five-membered ring has zero to two double bonds
  • the six- and seven-membered rings have zero to three double bonds.
  • heterocyclyl also includes bicyclic groups in which the heterocyclyl ring is fused to another monocyclic heterocyclyl group, or a four- to se-membered aromatic or nonaromatic carbocyclic ring.
  • the heterocyclyl group can be attached to the parent molecular moiety through any carbon atom or nitrogen atom in the group.
  • Heterocycloalkyl refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Whenever it appears herein, a numerical range such as “3 to 18” refers to each integer in the given range; e.g., “3 to 18 ring atoms” means that the heterocycloalkyl group may consist of 3 ring atoms, 4 ring atoms, etc., up to and including 18 ring atoms. In some embodiments, it is a C 5 -C 10 heterocycloalkyl. In some embodiments, it is a C 4 -C 10 heterocycloalkyl.
  • the heterocycloalkyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems.
  • the heteroatoms in the heterocycloalkyl radical may be optionally oxidized.
  • One or more nitrogen atoms, if present, are optionally quaternized.
  • the heterocycloalkyl radical is partially or fully saturated.
  • the heterocycloalkyl may be attached to the rest of the molecule through any atom of the ring(s).
  • heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-o-
  • a heterocycloalkyl moiety is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, —OR′, SR a , —OC(O)—R a , —N(R a ) 2 , —C(O)R a , —C(O)OR a , —C(O)N(R a ) 2 , —N(R a )C(O)OR a , —N(R a )C(O)OR a , —N(R a )C(O)R a , —N(R a )S(O)
  • Heterocycloalkyl also includes bicyclic ring systems wherein one non-aromatic ring, usually with 3 to 7 ring atoms, contains at least 2 carbon atoms in addition to 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of the foregoing heteroatoms; and the other ring, usually with 3 to 7 ring atoms, optionally contains 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen and is not aromatic.
  • heterocyclylalkyl refers to the divalent derivative of heterocycloalkyl.
  • C 1-10 alkyl-heterocycyl refers to a group as defined above where the alkyl moiety contains 1 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclic group refers to a group containing a terminal heterocyclic group attached to a linking alkyl group which contains 1 to 10 carbons and is branched or straight chain, such as, for example, 4-morpholinyl ethyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclylalkenyl refers to the divalent derivative of heterocyclylalkenyl. Either portion of the moiety is unsubstituted or substituted.
  • heterocycyl-C 2-10 alkenyl refers to a group as defined above where the alkenyl group contains 2 to 10 carbon atoms and is branched or straight chain, such as, for example, 4-(N-piperazinyl)-but-2-en-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclylalkynyl refers to a group wherein the alkynyl chain can be branched or straight chain forming a linking portion of the heterocyclylalkynyl moiety with the terminal heterocyclyl portion, as defined above, for example 2-pyrrolidinyl-1-butynyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocycyl-C 2-10 alkynyl refers to a group as defined above where the alkynyl group contains 2 to 10 carbon atoms and is branched or straight chain, such as, for example, 4-(N-piperazinyl)-but-2-yn-1-yl, and the like.
  • aryl-heterocycyl refers to a group containing a terminal aryl group attached to a linking heterocyclic group, such as for example, N4-(4-phenyl)-piperazinyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heteroaryl-heterocycyl refers to a group containing a terminal heteroaryl group attached to a linking heterocyclic group, such as for example, N4-(4-pyridyl)-piperazinyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • carboxylalkyl refers to a terminal carboxyl (—COOH) group attached to branched or straight chain alkyl groups as defined above.
  • carboxylalkenyl refers to a terminal carboxyl (—COOH) group attached to branched or straight chain alkenyl groups as defined above.
  • carboxylalkynyl refers to a terminal carboxyl (—COOH) group attached to branched or straight chain alkynyl groups as defined above.
  • carboxylcycloalkyl refers to a terminal carboxyl (—COOH) group attached to a cyclic aliphatic ring structure as defined above.
  • carboxylcycloalkenyl refers to a terminal carboxyl (—COOH) group attached to a cyclic aliphatic ring structure having ethylenic bonds as defined above.
  • cycloalkylalkyl and “cycloalkyl-alkyl” refer to a terminal cycloalkyl group as defined above attached to an alkyl group, for example cyclopropylmethyl, cyclohexylethyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • cycloalkylalkenyl and “cycloalkyl-alkenyl” refer to a terminal cycloalkyl group as defined above attached to an alkenyl group, for example cyclohexylvinyl, cycloheptylallyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • cycloalkylalkynyl and “cycloalkyl-alkynyl” refer to a terminal cycloalkyl group as defined above attached to an alkynyl group, for example cyclopropylpropargyl, 4-cyclopentyl-2-butynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • cycloalkenylalkyl and “cycloalkenyl-alkyl” refer to a terminal cycloalkenyl group as defined above attached to an alkyl group, for example 2-(cyclopenten-1-yl)ethyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • cycloalkenylalkenyl and “cycloalkenyl-alkenyl” refer to terminal a cycloalkenyl group as defined above attached to an alkenyl group, for example 1-(cyclohexen-3-yl)allyl and the like.
  • cycloalkenylalkynyl and “cycloalkenyl-alkynyl” refer to terminal a cycloalkenyl group as defined above attached to an alkynyl group, for example 1-(cyclohexen-3-yl)propargyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • alkoxy refers to the group —O-alkyl, including from 1 to 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. “Lower alkoxy” refers to alkoxy groups containing one to six carbons. In some embodiments, C 1 -C 4 alkyl, is an alkyl group which encompasses both straight and branched chain alkyls of from 1 to 4 carbon atoms.
  • haloalkoxy refers to an alkoxy group substituted with one or more halo groups, for example chloromethoxy, trifluoromethoxy, difluoromethoxy, perfluoroisobutoxy, and the like.
  • alkoxyalkoxyalkyl refers to an alkyl group substituted with an alkoxy moiety which is in turn is substituted with a second alkoxy moiety, for example methoxymethoxymethyl, isopropoxymethoxyethyl, and the like. This moiety is substituted with further substituents or not substituted with other substituents.
  • alkylthio includes both branched and straight chain alkyl groups attached to a linking sulfur atom, for example methylthio and the like.
  • alkoxyalkyl refers to an alkyl group substituted with an alkoxy group, for example isopropoxymethyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • alkoxyalkenyl refers to an alkenyl group substituted with an alkoxy group, for example 3-methoxyallyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • alkoxyalkynyl refers to an alkynyl group substituted with an alkoxy group, for example 3-methoxypropargyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkenylC 3-8 cycloalkyl refers to an alkenyl group as defined above substituted with a three to eight membered cycloalkyl group, for example, 4-(cyclopropyl)-2-butenyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • C 2-10 alkynylC 3-8 cycloalkyl refers to an alkynyl group as defined above substituted with a three to eight membered cycloalkyl group, for example, 4-(cyclopropyl)-2-butynyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclyl-C 1-10 alkyl refers to a heterocyclic group as defined above substituted with an alkyl group as defined above having 1 to 10 carbons, for example, 4-(N-methyl)-piperazinyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclyl-C 2-10 alkenyl refers to a heterocyclic group as defined above, substituted with an alkenyl group as defined above, having 2 to 10 carbons, for example, 4-(N-allyl) piperazinyl, and the like. Moieties wherein the heterocyclic group is substituted on a carbon atom with an alkenyl group are also included. Either portion of the moiety is unsubstituted or substituted.
  • heterocyclyl-C 2-10 alkynyl refers to a heterocyclic group as defined above, substituted with an alkynyl group as defined above, having 2 to 10 carbons, for example, 4-(N-propargyl) piperazinyl, and the like. Moieties wherein the heterocyclic group is substituted on a carbon atom with an alkenyl group are also included. Either portion of the moiety is unsubstituted or substituted.
  • oligomer refers to a low-molecular weight polymer, whose number average molecular weight is typically less than about 5000 g/mol, and whose degree of polymerization (average number of monomer units per chain) is greater than one and typically equal to or less than about 50.
  • “Sulfonamidyl” or “sulfonamido” refers to a —S( ⁇ O) 2 —NR′R′ radical, where each R′ is selected independently from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon).
  • the R′ groups in NR′R′ of the —S( ⁇ O) 2 —NR′R′ radical may be taken together with the nitrogen to which it is attached to form a 4-, 5-, 6-, or 7-membered ring.
  • a sulfonamido group is optionally substituted by one or more of the substituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively.
  • Compounds described can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers.
  • the present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Compounds may be shown without a definitive stereochemistry at certain positions.
  • the present invention includes all stereoisomers of the disclosed compounds and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
  • the present invention includes all manner of rotamers and conformationally restricted states of an inhibitor of the invention.
  • Substituents for alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl monovalent and divalent derivative radicals can be one or more of a variety of groups selected from, but not limited to: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, —OR′, ⁇ O, ⁇ NR 7 —, ⁇ N—OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′,
  • R′, R′′, R′′′ and R′′′ each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1-3 halogens), substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups.
  • each of the R groups is independently selected as are each R′, R′′, R′′′ and R′′′ groups when more than one of these groups is present.
  • R′ and R′′ or R′′ and R′′′ are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring.
  • —NR′R′′ is meant to include, but not be limited to, 1-pyrrolidinyl, 4 piperazinyl, and 4-morpholinyl.
  • alkyl is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., —CF 3 and CH 2 CF 3 ) and acyl (e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like).
  • haloalkyl e.g., —CF 3 and CH 2 CF 3
  • acyl e.g., —C(O)CH 3 , —C(O)CF 3 , —C(O)CH 2 OCH 3 , and the like.
  • exemplary substituents for aryl and heteroaryl groups are varied and are selected from, for example: halogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, —OR′, —NR′R′′, —SR′, -halogen, —SiR′R′′R′′′, —OC(O)R′, —C(O)R′, —CO 2 R′, —C(O)NR′R′′, —OC(O)NR′R′′, —NR′′C(O)R′, —NR 7 —C(O)NR′′R′′′, —NR′′C(O)OR′, —NR—C(NR′R′′R′′′) ⁇ NR′′′, —NR—C(NR—C(NR)OR′, —NR—C(NR′R′′R′′′) ⁇ NR′′
  • 0-2 in the context of —S(O) (0-2) — are integers of 0, 1, and 2.
  • Two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally form a ring of the formula -T-C(O)—(CRR′) q —U—, wherein T and U are independently —NR—, —O—, —CRR′— or a single bond, and q is an integer of from 0 to 3.
  • two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH 2 ), —B—, wherein A and B are independently CRR′—, —O—, —NR—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 NR′— or a single bond, and r is an integer of from 1 to 4.
  • One of the single bonds of the new ring so formed may optionally be replaced with a double bond.
  • two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula —(CRR′) s —X′—(C′′R′′′) d —, where s and d are independently integers of from 0 to 3, and X′ is —O—, —NR 7 —, —S—, —S(O)—, —S(O) 2 —, or —S(O) 2 NR′—.
  • the substituents R, R′, R′′ and R′′′ are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • the compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds.
  • the compounds may be radiolabeled with radioactive isotopes, such as for example tritium ( 3 H), iodine-125 ( 125 I) or carbon-14 ( 14 C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
  • the present invention provides a method for treating a disease condition associated with PI3-kinase ⁇ and/or mTOR in a subject.
  • the method typically comprises administering to a subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor.
  • a therapeutically effective amount of a combination of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor refers to a combination of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor, wherein the combination is sufficient to effect the intended application including but not limited to disease treatment, as defined herein.
  • a therapeutically effective amount of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor in combination to effect such treatment.
  • Also contemplated in the subject methods is the use of a sub-therapeutic amount of a PI3-kinase ⁇ inhibitor and/or an mTOR inhibitor in the combination for treating an intended disease condition.
  • the present invention provides for a method for treating a disease condition associated with PI3-kinase ⁇ and/or mTOR in a subject, comprising administering to the subject simultaneously or sequentially a synergistically effective therapeutic amount of a combination of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor.
  • the therapeutically effective amount of the subject combination of compounds may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art.
  • the term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or downregulation of activity of a target protein.
  • the specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • the PI3-kinase ⁇ inhibitor utilized in the subject methods typically exhibits selective inhibition of PI3-kinase ⁇ relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) including, e.g., PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • PI3-kinase type I phosphatidylinositol 3-kinases
  • Selective inhibition of PI3-kinase ⁇ can be ascertained by an in vitro or an in vivo method. Any assay known in the art may be used, including without limitation, immunoassays, immunoprecipitation, fluorescence or cell-based assays. In some embodiments, an in vitro assay is used to determine selective inhibition of PI3-kinase ⁇ by an assay which measures the activity of the PI3K ⁇ protein relative to the activity of another PI3-kinase such as PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • an in vitro assay is used to determine selective inhibition of PI3-kinase ⁇ by an assay which measures the activity of the PI3K ⁇ protein relative to the activity of another PI3-kinase such as PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇
  • a time resolved FRET assay that indirectly measures PIP3 product formed by the activity of a PI3-K may be used to determine an IC50 value for a test compound for PI3-kinase ⁇ and/or any of the other PI3-kinases.
  • IC50 refers to the half maximal inhibitory concentration of an inhibitor in inhibiting biological or biochemical function. This quantitative measure indicates how much of a particular inhibitor is needed to inhibit a given biological process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half. In other words, it is the half maximal (50%) inhibitory concentration (IC) of a substance (50% IC, or IC50).
  • IC50 refers to the plasma concentration required for obtaining 50% of a maximum effect in vivo.
  • Determination of IC50 can be made by determining and constructing a dose-response curve and examining the effect of different concentrations of an inhibitor on reversing agonist activity. In vitro assays that are useful in making these determinations are referred to as “in vitro kinase assays.”
  • an in vitro kinase assay includes the use of labeled ATP as phosphodonor, and following the kinase reaction the substrate peptide is captured on an appropriate filter. Unreacted labeled ATP and metabolites are resolved from the radioactive peptide substrate by various techniques, involving trichloroacetic acid precipitation and extensive washing. Addition of several positively charged residues allows capture on phosphocellulose paper followed by washing. Radioactivity incorporated into the substrate peptide is detected by scintillation counting.
  • This assay is relatively simple, reasonably sensitive, and the peptide substrate can be adjusted both in terms of sequence and concentration to meet the assay requirements.
  • a cell-based assay is used to ascertain selective inhibition of PI3-kinase ⁇ .
  • an inhibitor can be shown to be selective for PI3-kinase ⁇ if it selectively downregulates PI3-kinase signal transduction in cells that express PI3-kinase ⁇ , preferably in cells that exhibit abnormally high level or activity of PI3-kinase ⁇ .
  • a variety of cells having PI3-kinase ⁇ mutations and hence exhibiting such PI3-kinase ⁇ abnormalities are known in the art.
  • Non-limiting examples of cell lines harboring such mutations include those that carry point mutations, deletions, substitutions, or translation of nucleic acid sequence of the PI3-kinase ⁇ gene.
  • Examples of such cell lines include but are not limited to BT20 (H1047R mutation), MCF-7 (E545K mutation), MDA-MB-361 (E545K mutation), MDA-MB-453 (H1047R mutation), T47D (H1047R mutation), Hec-1A (G1049R mutation) and HCT-116 (H1047R mutation).
  • Other cell lines having mutations in the PI3K ⁇ protein may be used, such as cells harboring mutations in the p85, C2, helical or kinase domains.
  • inhibition of PI3-kinase ⁇ activity can be determined by a reduction in signal transduction of the PI3-kinase ⁇ pathway.
  • a wide variety of readouts can be utilized to establish a reduction of the output of such signaling pathway.
  • Some non-limiting exemplary readouts include (1) a decrease in phosphorylation of Akt at residues, including but not limited to S473 and T308; (2) a decrease in activation of Akt as evidenced by a reduction of phosphorylation of Akt substrates including but not limited to FoxO1/O3a T24/32, GSK3 ⁇ / ⁇ S21/9, and TSC2 T1462; (3) a decrease in phosphorylation of signaling molecules downstream of PI3-kinase ⁇ , including but not limited to ribosomal S6 S240/244, 70S6K T389, and 4EBP1 T37/46; (4) inhibition of proliferation of cells including but not limited to normal or neoplastic cells, mouse embryonic fibroblasts, leukemic blast cells, cancer stem cells, and cells that mediate autoimmune reactions; (5) induction of apoptosis of cells or cell cycle arrest; (6) reduction of cell chemotaxis; and (7) an increase in binding of 4EBP1 to
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ relative to one, two or three other type I phosphatidylinositol 3-kinases (PI3-kinases) consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • PI3-kinases type I phosphatidylinositol 3-kinases
  • some of the subject inhibitors selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases.
  • some of the subject inhibitors selectively inhibit PI3-kinase ⁇ and PI3-kinase ⁇ as compared to the rest of the type I PI3-kinases. In still yet other embodiments, some of the subject inhibitors selectively inhibit PI3-kinase ⁇ and PI3-kinase 6 as compared to the rest of the type I PI3-kinases.
  • the subject methods utilizes a PI3-kinase ⁇ inhibitor with an IC50 value of about or less than a predetermined value, as ascertained in an in vitro kinase assay.
  • the PI3-kinase ⁇ inhibitor inhibits PI3-kinase ⁇ with an IC50 value of about 1 nM or less, 2 nM or less, 5 nM or less, 7 nM or less, 10 nM or less, 20 nM or less, 30 nM or less, 40 nM or less, 50 nM or less, 60 nM or less, 70 nM or less, 80 nM or less, 90 nM or less, 100 nM or less, 120 nM or less, 140 nM or less, 150 nM or less, 160 nM or less, 170 nM or less, 180 nM or less, 190 nM or less, 200 nM or less, 225 nM
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, or 1000 times less than its IC50 value against one, two, or three other type I PI3-kinase(s) selected from the group consisting of PI3-kinase PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is less than about 1 nM, 2 nM, 5 nM, 7 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 120 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 225 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM,
  • the PI3-kinase ⁇ inhibitor inhibits PI3-kinase ⁇ with an IC50 value of about 200 nM or less as ascertained in an in vitro kinase assay and the IC50 value is at least 5, 10, 15, 20, 25, 50, 100, or 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is less than about 100 nM, and said IC50 value is at 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor inhibits PI3-kinase ⁇ with an IC50 value of about 50 nM or less as ascertained in an in vitro kinase assay and the IC50 value is at least 5, 10, 15, 20, 25, 50, 100, or 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is less than about 20 nM, and said IC50 value is at 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an IC50 value that is less than about 20 nM, and said IC50 value is at 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor inhibits PI3-kinase ⁇ with an IC50 value of about 20 nM or less as ascertained in an in vitro kinase assay and the IC50 value is at least 100 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor inhibits PI3-kinase ⁇ with an EC50 value of about 10 ⁇ M or less, 5 ⁇ M or less, 2.5 ⁇ M or less, 1 ⁇ M or less, 500 nM or less, 100 nM or less, 75 nM or less, 50 nM or less, 25 nM or less, 10 nM or less, 5 nM or less, 1 nM or less, 500 ⁇ M or less, or 100 ⁇ M or less as ascertained in an in vitro kinase assay.
  • the PI3-kinase ⁇ inhibitor selectively inhibits PI3-kinase ⁇ with an EC50 value that is at least 5, 10, 15, 20, 25, 50, 100, or 1000 times less than its EC50 value against one, two or there other type I PI3-kinases selected from the group consisting of PI3-kinase PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the PI3-kinase ⁇ inhibitor inhibits PI3-kinase ⁇ with an EC50 value of about 10 ⁇ M or less, 5 ⁇ M or less, 2.5 ⁇ M or less, 1 ⁇ M or less, 500 nM or less, 100 nM or less, 75 nM or less, 50 nM or less, 25 nM or less, 10 nM or less, 5 nM or less, 1 nM or less, 500 pM or less, or 100 pM or less as ascertained in an in vitro kinase assay, and such EC50 value is at least 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its EC50 value against one, two or three other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the mTOR inhibitor utilized in the subject methods is typically highly selective for the target molecule.
  • the mTOR inhibitor binds to and directly inhibits both mTORC1 and mTORC2.
  • Such ability can be ascertained using any method known in the art or described herein.
  • inhibition of mTorC1 and/or mTorC2 activity can be determined by a reduction in signal transduction of the PI3K/Akt/mTor pathway.
  • a wide variety of readouts can be utilized to establish a reduction of the output of such signaling pathway.
  • Some non-limiting exemplary readouts include (1) a decrease in phosphorylation of Akt at residues, including but not limited to S473 and T308; (2) a decrease in activation of Akt as evidenced by a reduction of phosphorylation of Akt substrates including but not limited to FoxO1/O3a T24/32, GSK3 ⁇ / ⁇ S21/9, and TSC2 T1462; (3) a decrease in phosphorylation of signaling molecules downstream of mTor, including but not limited to ribosomal S6 S240/244, 70S6K T389, and 4EBP1 T37/46; (4) inhibition of proliferation of cells including but not limited to normal or neoplastic cells, mouse embryonic fibroblasts, leukemic blast cells, cancer stem cells, and cells that mediate autoimmune reactions; (5) induction of apoptosis of cells or cell cycle arrest; (6) reduction of cell chemotaxis; and (7) an increase in binding of 4EBP1 to eIF4E.
  • mTor exists in two types of complexes, mTorC1 containing the raptor subunit and mTorC2 containing rictor.
  • rictor refers to a cell growth regulatory protein having human gene locus 5p13.1. These complexes are regulated differently and have a different spectrum of substrates. For instance, mTorC1 phosphorylates S6 kinase (S6K) and 4EBP1, promoting increased translation and ribosome biogenesis to facilitate cell growth and cell cycle progression. S6K also acts in a feedback pathway to attenuate PI3K/Akt activation.
  • S6K S6 kinase
  • 4EBP1 S6 kinase
  • inhibition of mTorC1 results in activation of 4EBP1, resulting in inhibition of (e.g. a decrease in) RNA translation.
  • mTorC2 is generally insensitive to rapamycin and selective inhibitors and is thought to modulate growth factor signaling by phosphorylating the C-terminal hydrophobic motif of some AGC kinases such as Akt.
  • Akt AGC kinases
  • mTorC2 is required for phosphorylation of the S473 site of Akt.
  • mTorC1 activity is partly controlled by Akt whereas Akt itself is partly controlled by mTorC2.
  • Akt Growth factor stimulation of PI3K causes activation of Akt by phosphorylation at the two key sites, S473 and T308. It has been reported that full activation of Akt requires phosphorylation of both S473 and T308Active. Akt promotes cell survival and proliferation in many ways including suppressing apoptosis, promoting glucose uptake, and modifying cellular metabolism. Of the two phosphorylation sites on Akt, activation loop phosphorylation at T308, mediated by PDK1, is believed to be indispensable for kinase activity, while hydrophobic motif phosphorylation at S473 enhances Akt kinase activity.
  • Selective mTor inhibition may also be determined by expression levels of the mTor genes, its downstream signaling genes (for example by RT-PCR), or expression levels of the proteins (for example by immunocytochemistry, immunohistochemistry, Western blots) as compared to other PI3-kinases or protein kinases.
  • Cell-based assays for establishing selective inhibition of mTorC1 and/or mTorC2 can take a variety of formats. This generally will depend on the biological activity and/or the signal transduction readout that is under investigation. For example, the ability of the agent to inhibit mTorC1 and/or mTorC2 to phosphorylate the downstream substrate(s) can be determined by various types of kinase assays known in the art. Representative assays include but are not limited to immunoblotting and immunoprecipitation with antibodies such as anti-phosphotyrosine, anti-phosphoserine or anti-phosphothreonine antibodies that recognize phosphorylated proteins.
  • kinase activity can be detected by high throughput chemiluminescent assays such as AlphaScreenTM (available from Perkin Elmer) and eTagTM assay (Chan-Hui, et al. (2003) Clinical Immunology 111: 162-174).
  • single cell assays such as flow cytometry as described in the phosflow experiment can be used to measure phosphorylation of multiple downstream mTOR substrates in mixed cell populations.
  • One advantage of the immunoblotting and phosflow methods is that the phosphorylation of multiple kinase substrates can be measured simultaneously. This provides the advantage that efficacy and selectivity can be measured at the same time.
  • cells may be contacted with an mTOR inhibitor at various concentrations and the phosphorylation levels of substrates of both mTOR and other kinases can be measured.
  • a large number of kinase substrates are assayed in what is termed a “comprehensive kinase survey.”
  • Selective mTOR inhibitors are expected to inhibit phosphorylation of mTOR substrates without inhibiting phosphorylation of the substrates of other kinases.
  • selective mTOR inhibitors may inhibit phosphorylation of substrates of other kinases through anticipated or unanticipated mechanisms such as feedback loops or redundancy.
  • Non-limiting examples of cell proliferation assays include testing for tritiated thymidine uptake assays, BrdU (5′-bromo-2′-deoxyuridine) uptake (kit marketed by Calibochem), MTS uptake (kit marketed by Promega), MTT uptake (kit marketed by Cayman Chemical), CyQUANT® dye uptake (marketed by Invitrogen).
  • Apoptosis and cell cycle arrest analysis can be performed with any methods exemplified herein as well other methods known in the art. Many different methods have been devised to detect apoptosis. Exemplary assays include but are not limited to the TUNEL (TdT-mediated dUTP Nick-End Labeling) analysis, ISEL (in situ end labeling), and DNA laddering analysis for the detection of fragmentation of DNA in populations of cells or in individual cells, Annexin-V analysis that measures alterations in plasma membranes, detection of apoptosis related proteins such p53 and Fas.
  • TUNEL TdT-mediated dUTP Nick-End Labeling
  • ISEL in situ end labeling
  • DNA laddering analysis for the detection of fragmentation of DNA in populations of cells or in individual cells
  • Annexin-V analysis that measures alterations in plasma membranes
  • detection of apoptosis related proteins such p53 and Fas.
  • a cell-based assay typically proceeds with exposing the target cells (e.g., in a culture medium) to a test compound which is a potential mTorC1 and/or mTorC2 selective inhibitor, or a PI3-kinase inhibitor and then assaying for readout under investigation.
  • a test compound which is a potential mTorC1 and/or mTorC2 selective inhibitor, or a PI3-kinase inhibitor and then assaying for readout under investigation.
  • a test compound which is a potential mTorC1 and/or mTorC2 selective inhibitor, or a PI3-kinase inhibitor and then assaying for readout under investigation.
  • a test compound which is a potential mTorC1 and/or mTorC2 selective inhibitor, or a PI3-kinase inhibitor
  • they can directly be added to the cells or in conjunction with carriers.
  • the agent when the agent is nucleic acid, it can be
  • the nucleic acid can be incorporated into an expression or insertion vector for incorporation into the cells.
  • Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are well known in the art. Such vectors are capable of transcribing RNA in vitro or in vitro, and are commercially available from sources such as Stratagene (La Jolla, Calif.) and Promega Biotech (Madison, Wis.).
  • vectors are viruses, such as baculovirus and retrovirus, bacteriophage, adenovirus, adeno-associated virus, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
  • viruses such as baculovirus and retrovirus, bacteriophage, adenovirus, adeno-associated virus, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression.
  • non-viral vectors including DNA/liposome complexes, and targeted viral protein DNA complexes.
  • the nucleic acid or proteins of this invention can be conjugated to antibodies or binding fragments thereof which bind cell surface anti
  • Liposomes that also comprise a targeting antibody or fragment thereof can be used in the methods of this invention.
  • Other biologically acceptable carriers can be utilized, including those described in, for example, REMINGTON'S PHARMACEUTICAL SCIENCES, 19th Ed. (2000), in conjunction with the subject compounds.
  • the subject agents can also be utilized to inhibit phosphorylation of both Akt (S473) and Akt (T308) in a cell.
  • the present invention provides a method comprises the step of contacting a cell with an effective amount of such biologically active agent such that Akt phosphorylation at residues S473 and T308 is simultaneously inhibited.
  • the biologically active agent inhibits phosphorylation of S473 of Akt more effectively than phosphorylation of T308 of Akt when tested at a comparable molar concentration, preferably at an identical molar concentration.
  • Inhibition of Akt phosphorylation can be determined using any methods known in the art or described herein. Representative assays include but are not limited to immunoblotting and immunoprecipitation with antibodies such as anti-phosphotyrosine antibodies that recognize the specific phosphorylated proteins. Cell-based ELISA kit quantifies the amount of activated (phosphorylated at S473) Akt relative to total Akt protein is also available (SuperArray Biosciences).
  • any cells that express PI3-kinase ⁇ , mTorC1, mTorC2 and/or Akt can be utilized.
  • specific cell types whose proliferation can be inhibited include fibroblast, cells of skeletal tissue (bone and cartilage), cells of epithelial tissues (e.g. liver, lung, breast, skin, bladder and kidney), cardiac and smooth muscle cells, neural cells (glia and neurones), endocrine cells (adrenal, pituitary, pancreatic islet cells), melanocytes, and many different types of haemopoietic cells (e.g., cells of B-cell or T-cell lineage, and their corresponding stem cells, lymphoblasts).
  • haemopoietic cells e.g., cells of B-cell or T-cell lineage, and their corresponding stem cells, lymphoblasts.
  • cells exhibiting a neoplastic propensity or phenotype are also of interest.
  • diseases involving abnormal functioning of genes include but are not limited to autoimmune diseases, cancer, obesity, hypertension, diabetes, neuronal and/or muscular degenerative diseases, cardiac diseases, endocrine disorders, and any combinations thereof.
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1 nM, 2 nM, 5 nM, 7 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 120 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 225 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 200, 100, 75, 50, 25, 10, 5, 1 or 0.5 nM or less as ascertained in an in vitro kinase assay. In one instance, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay. Alternatively, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 10 nM or less as ascertained in an in vitro kinase assay.
  • the present invention provides the use of an mTOR inhibitor, wherein the mTOR inhibitor directly binds to and inhibits both mTORC1 and mTORC2 with an IC50 value of about or less than a predetermined value, as ascertained in an in vitro kinase assay.
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1 nM or less, 2 nM or less, 5 nM or less, 7 nM or less, 10 nM or less, 20 nM or less, 30 nM or less, 40 nM or less, 50 nM or less, 60 nM or less, 70 nM or less, 80 nM or less, 90 nM or less, 100 nM or less, 120 nM or less, 140 nM or less, 150 nM or less, 160 nM or less, 170 nM or less, 180 nM or less, 190 nM or less, 200 nM or less, 225 nM or less, 250 nM or less, 275 nM or less, 300 nM or less, 325 nM or less, 350 nM or less, 375 nM or less, 400 nM or less, 425 nM or less, 425
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1 nM or less, 2 nM or less, 5 nM or less, 7 nM or less, 10 nM or less, 20 nM or less, 30 nM or less, 40 nM or less, 50 nM or less, 60 nM or less, 70 nM or less, 80 nM or less, 90 nM or less, 100 nM or less, 120 nM or less, 140 nM or less, 150 nM or less, 160 nM or less, 170 nM or less, 180 nM or less, 190 nM or less, 200 nM or less, 225 nM or less, 250 nM or less, 275 nM or less, 300 nM or less, 325 nM or less, 350 nM or less, 375 nM or less, 400 nM or less, 425 nM or less, 425
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 10 nM or less as ascertained in an in vitro kinase assay, and the mTOR inhibitor is substantially inactive against one or more types I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • substantially inactive refers to an inhibitor that inhibits the activity of its target by less than approximately 1%, 5%, 10%, 15% or 20% of its maximal activity in the absence of the inhibitor, as determined by an in vitro enzymatic assay (e.g. in vitro kinase assay).
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1000, 500, 100, 75, 50, 25, 10, 5, 1, or 0.5 nM or less as ascertained in an in vitro kinase assay, and said IC50 value is at least 2, 5, 10, 15, 20, 50, 100 or 100 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay, and said IC50 value is at least 5 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay, and said IC50 value is at least 5 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase ⁇ , PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • the mTOR inhibitor utilized in the subject methods inhibits one of mTORC1 and mTORC2 selectively with an IC50 value of about 1000, 500, 100, 75, 50, 25, 10, 5, 1, or 0.5 nM or less as ascertained in an in vitro kinase.
  • the mTOR inhibitor utilized in the subject methods inhibits mTORC1 selectively with an IC50 value of about 1000, 500, 100, 75, 50, 25, 10, 5, 1, or 0.5 nM or less as ascertained in an in vitro kinase.
  • rapamycin and rapamycin derivatives or analogues have been shown to primarily inhibit mTORC1 and not mTORC2.
  • Suitable mTORC1 inhibitors compounds include, for example, sirolimus (rapamycin), deforolimus (AP23573, MK-8669), everolimus (RAD-001), temsirolimus (CCI-779), zotarolimus (ABT-578), and biolimus A9 (umirolimus).
  • PI3-kinase ⁇ inhibitors or mTOR inhibitors suitable for use in the subject methods can be selected from a variety types of molecules.
  • an inhibitor can be biological or chemical compound such as a simple or complex organic or inorganic molecule, peptide, peptide mimetic, protein (e.g. antibody), liposome, or a polynucleotide (e.g. small interfering RNA, microRNA, anti-sense, aptamer, ribozyme, or triple helix).
  • a polynucleotide e.g. small interfering RNA, microRNA, anti-sense, aptamer, ribozyme, or triple helix.
  • cancer cells may depend on overactive signaling for their survival (known as the oncogene addiction hypothesis). In this way, cancer cells are frequently observed to adapt to drug inhibition of an aberrant signaling component by selecting for mutations in the same pathway that overcome the effect of the drug. Therefore, cancer therapies may be more successful in overcoming the problem of drug resistance if they target a signaling pathway as a whole, or target more than one component within a signaling pathway.
  • selective inhibition of PI3-kinase ⁇ provides a more targeted treatment to a disease condition mediated by PI3-kinase without disrupting one or more pathways that are implicated by one or more other type I phosphatidylinositol 3-kinases, namely PI3-kinase ⁇ , PI3-kinase ⁇ , and PI3-kinase ⁇ .
  • FIG. 1 Some signaling pathways that contain PI3K and mTOR are illustrated in FIG. 1 .
  • Akt serine/threonine kinase.
  • Akt possesses a protein domain known as a PH domain, or Pleckstrin Homology domain, which binds to phosphoinositides with high affinity.
  • PH domain a protein domain known as a PH domain, or Pleckstrin Homology domain, which binds to phosphoinositides with high affinity.
  • PIP3 phosphatidylinositol (3,4,5)-trisphosphate, PtdIns(3,4,5)P3
  • PIP2 phosphatidylinositol (3,4)-bisphosphate, PtdIns(3,4)P2).
  • PI3K phosphorylates PIP2 in response to signals from chemical messengers, such as ligand binding to G protein-coupled receptors or receptor tyrosine kinases. Phosphorylation by PI3K converts PIP2 to PIP3, recruiting Akt to the cell membrane where it is phosphorylated at serine 473 (S473) by mTORC2. Phosphorylation of Akt at another site, threonine 308 (T308), is not directly dependent on mTORC2, but requires PI3K activity. Therefore, PI3K activity towards Akt can be isolated from mTOR activity by examining Akt threonine 308 phosphorylation status in cells lacking mTORC2 activity.
  • the subject methods are useful for treating a disease condition associated with PI3-kinase ⁇ and/or mTOR.
  • Any disease condition that results directly or indirectly from an abnormal activity or expression level of PI3-kinase ⁇ and/or mTOR can be an intended disease condition.
  • PI3-kinase ⁇ has been implicated, for example, in a variety of human cancers.
  • Angiogenesis has been shown to selectively require the ⁇ isoform of PI3K in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453; 662-6). Mutations in the gene coding for PI3K a or mutations which lead to upregulation of PI3K a are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers.
  • mutations in the gene coding for PI3K a are point mutations clustered within several hotspots in helical and kinase domains, such as E542K, E545K, and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. Because of the high rate of PI3K a mutations, targeting of this pathway provides valuable therapeutic opportunities. While other PI3K isoforms such as PI3K ⁇ or PI3K ⁇ are expressed primarily in hematopoietic cells, PI3K a, along with PI3K (3, is expressed constitutively.
  • Disease conditions associated with PI3-kinase ⁇ and/or mTOR can also be characterized by abnormally high level of activity and/or expression of downstream messengers of PI3-kinase ⁇ .
  • proteins or messengers such as PIP2, PIP3, PDK, Akt, PTEN, PRAS40, GSK-313, p21, p27 may be present in abnormal amounts which can be identified by any assays known in the art.
  • Deregulation of the mTOR pathway is emerging as a common theme in diverse human diseases and as a consequence drugs that target mTOR have therapeutic value.
  • the diseases associated with deregulation of mTORC1 include, but are not limited to, tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM), both of which are caused by mutations in TSC1 or TSC2 tumor suppressors.
  • TSC tuberous sclerosis complex
  • LAM lymphangioleiomyomatosis
  • Patients with TSC develop benign tumors that when present in brain, however, can cause seizures, mental retardation and death.
  • LAM is a serious lung disease
  • Inhibition of mTORC1 may help patients with Peutz-Jeghers cancer-prone syndrome caused by the LKB 1 mutation.
  • mTORC1 may also have role in the genesis of sporadic cancers.
  • Akt Activated Akt regulates cell survival, cell proliferation and metabolism by phosphorylating proteins such as BAD, FOXO, NF-KB, p21Cip1, p27Kip1, GSK3 ⁇ and others. Akt might also promote cell growth by phosphorylating TSC2. Akt activation probably promotes cellular transformation and resistance to apoptosis by collectively promoting growth, proliferation and survival, while inhibiting apoptotic pathways.
  • the combination of an inhibitor of mTORC1 and mTORC2 and a PI3-kinase ⁇ inhibitor is beneficial for treatment of tumors with elevated Akt phosphorylation, and should down-regulate cell growth, cell survival and cell proliferation.
  • the subject to be treated is tested prior to treatment using a diagnostic assay to determine the sensitivity of tumor cells to a PI3K ⁇ kinase inhibitor.
  • a diagnostic assay to determine the sensitivity of tumor cells to a PI3K ⁇ kinase inhibitor. Any method known in the art that can determine the sensitivity of the tumor cells of a subject to a PI3K ⁇ kinase inhibitor can be employed.
  • the subject is tested prior to treatment using a diagnostic assay to determine the sensitivity of tumor cells to an PI3K ⁇ kinase inhibitor
  • a diagnostic assay to determine the sensitivity of tumor cells to an PI3K ⁇ kinase inhibitor when the subject is identified as one whose tumor cells are predicted to have low sensitivity to an PI3K ⁇ kinase inhibitor as a single agent, are likely to display enhanced sensitivity in the presence of an mTOR inhibitor, or vice versa, when the subject is administered, simultaneously or sequentially, a therapeutically effective amount of a combination of an PI3K ⁇ kinase inhibitor and an mTOR inhibitor.
  • the subject when the subject is identified as one whose tumor cells are predicted to have high sensitivity to an PI3K ⁇ kinase inhibitor as a single agent, but may also display enhanced sensitivity in the presence of an mTOR inhibitor based on the results described herein, the subject is administered, simultaneously or sequentially, a therapeutically effective amount of a combination of an PI3K ⁇ kinase inhibitor and an mTOR inhibitor.
  • one or more additional anti-cancer agents or treatments can be co-administered simultaneously or sequentially with the PI3K ⁇ kinase inhibitor and mTOR inhibitor, as judged to be appropriate by the administering physician given the prediction of the likely responsiveness of the subject to the combination of PI3K ⁇ kinase inhibitor and mTOR inhibitor, in combination with any additional circumstances pertaining to the individual subject.
  • the present invention provides for a method comprising: (a) determining the presence in a subject of a mutation in PI3-kinase ⁇ that is associated with a disease condition mediated by PI3-kinase ⁇ ; and (b) administering to said subject a pharmaceutical composition of the invention.
  • the present invention provides for a method of inhibiting phosphorylation of both Akt (S473) and Akt (T308) in a cell, comprising contacting a cell with an effective amount of a PI3-kinase ⁇ inhibitor and an mTOR inhibitor, biologically active agent that selectively inhibits both mTORC1 and mTORC2 activity relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) as ascertained by a cell-based assay or an in vitro kinase assay, wherein the PI3-kinase ⁇ inhibitor exhibits selective inhibition of PI3-kinase ⁇ relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) ascertained by an in vitro kinase assay, wherein the one or more type I PI3-kinase is selected from the group consisting of PI3-kinase
  • Non-limiting examples of such conditions include but are not limited to Acanthoma, Acinic cell carcinoma, Acoustic neuroma, Acral lentiginous melanoma, Acrospiroma, Acute eosinophilic leukemia, Acute lymphoblastic leukemia, Acute megakaryoblastic leukemia, Acute monocytic leukemia, Acute myeloblastic leukemia with maturation, Acute myeloid dendritic cell leukemia, Acute myeloid leukemia, Acute promyelocytic leukemia, Adamantinoma, Adenocarcinoma, Adenoid cystic carcinoma, Adenoma, Adenomatoid odontogenic tumor, Adrenocortical carcinoma, Adult T-cell leukemia, Aggressive NK-cell leukemia, AIDS-Related Cancers, AIDS-related lymphoma, Alveolar soft part sarcoma, Ameloblastic fibroma, Anal cancer, Anaplastic large cell lymph
  • the methods of using a PI3K ⁇ inhibitor and an mTOR inhibitor described herein are applied to the treatment of heart conditions including atherosclerosis, heart hypertrophy, cardiac myocyte dysfunction, elevated blood pressure and vasoconstriction.
  • the invention also relates to a method of treating diseases related to vasculogenesis or angiogenesis in a mammal that comprises administering to said mammal a therapeutically effective amount of a PI3K ⁇ inhibitor and an mTOR inhibitor of the present invention, or any pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma
  • diabetes diabetic retinopathy, retinopathy of prematurity
  • age-related macular degeneration hemangio
  • the invention provides for the use of a PI3K ⁇ inhibitor and an mTOR inhibitor for treating a disease condition associated with PI3-kinase ⁇ and/or mTOR, including, but not limited to, conditions related to an undesirable, over-active, harmful or deleterious immune response in a mammal, collectively termed “autoimmune disease.”
  • Autoimmune disorders include, but are not limited to, Crohn's disease, ulcerative colitis, psoriasis, psoriatic arthritis, juvenile arthritis and ankylosing spondilitis.
  • Other non-limiting examples of autoimmune disorders include autoimmune diabetes, multiple sclerosis, systemic lupus erythematosus (SLE), rheumatoid spondylitis, gouty arthritis, allergy, autoimmune uveitis, nephrotic syndrome, multisystem autoimmune diseases, autoimmune hearing loss, adult respiratory distress syndrome, shock lung, chronic pulmonary inflammatory disease, pulmonary sarcoidosis, pulmonary fibros
  • Undesirable immune response can also be associated with or result in, e.g., asthma, emphysema, bronchitis, psoriasis, allergy, anaphylaxsis, auto-immune diseases, rhuematoid arthritis, graft versus host disease, transplantation rejection, lung injuries, and lupus erythematosus.
  • the pharmaceutical compositions of the present invention can be used to treat other respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing.
  • the compositions of the invention can be further used to treat multiorgan failure.
  • the invention also provides methods of using a PI3K ⁇ inhibitor and an mTOR inhibitor for the treatment of liver diseases (including diabetes), pancreatitis or kidney disease (including proliferative glomerulonephritis and diabetes-induced renal disease) or pain in a mammal.
  • liver diseases including diabetes
  • pancreatitis or kidney disease including proliferative glomerulonephritis and diabetes-induced renal disease
  • the invention also provides a method of using a PI3K ⁇ inhibitor and an mTOR inhibitor for the treatment of sperm motility.
  • the invention further provides a method of using a PI3K ⁇ inhibitor and an mTOR inhibitor for the treatment of neurological or neurodegenerative diseases including, but not limited to, Alzheimer's disease, Huntington's disease, CNS trauma, and stroke.
  • the invention further provides a method of using a PI3K ⁇ inhibitor and an mTOR inhibitor for the prevention of blastocyte implantation in a mammal.
  • the invention also relates to a method of using a PI3K ⁇ inhibitor and an mTOR inhibitor for treating a disease related to vasculogenesis or angiogenesis in a mammal which can manifest as tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma
  • diabetes diabetic retinopathy,
  • the invention further provides a method of using a PI3K ⁇ inhibitor and an mTOR inhibitor for the treatment of disorders involving platelet aggregation or platelet adhesion, including but not limited to Bernard-Soulier syndrome, Glanzmann's thrombasthenia, Scott's syndrome, von Willebrand disease, Hermansky-Pudlak Syndrome, and Gray platelet syndrome.
  • methods of using a PI3K ⁇ inhibitor and an mTOR inhibitor are provided for treating a disease which is skeletal muscle atrophy, skeletal muscle hypertrophy, leukocyte recruitment in cancer tissue, invasion metastasis, melanoma, Kaposi's sarcoma, acute and chronic bacterial and viral infections, sepsis, glomerulo sclerosis, glomerulo, nephritis, or progressive renal fibrosis.
  • Certain embodiments contemplate a human subject such as a subject that has been diagnosed as having or being at risk for developing or acquiring a disease condition associated with PI3-kinase ⁇ and/or mTOR.
  • a non-human subject for example a non-human primate such as a macaque, chimpanzee, gorilla, vervet, orangutan, baboon or other non-human primate, including such non-human subjects that can be known to the art as preclinical models, including preclinical models for inflammatory disorders.
  • non-human subject that is a mammal, for example, a mouse, rat, rabbit, pig, sheep, horse, bovine, goat, gerbil, hamster, guinea pig or other mammal.
  • subject or biological source can be a non-mammalian vertebrate, for example, another higher vertebrate, or an avian, amphibian or reptilian species, or another subject or biological source.
  • a transgenic animal is utilized.
  • a transgenic animal is a non-human animal in which one or more of the cells of the animal includes a nucleic acid that is non-endogenous (i.e., heterologous) and is present as an extrachromosomal element in a portion of its cell or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells).
  • a nucleic acid that is non-endogenous (i.e., heterologous) and is present as an extrachromosomal element in a portion of its cell or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells).
  • the invention provides a compound which is an inhibitor of mTor of the Formula I:
  • X 1 is N or C-E 1
  • X 2 is N or C
  • X 3 is N or C
  • X 4 is C—R 9 or N
  • X 5 is N or C-E 1
  • X 6 is C or N
  • X 7 is C or N; and wherein no more than two nitrogen ring atoms are adjacent;
  • R 1 is H, -L-C 3-8 cycloalkyl, —C 3-8 cycloalkyl, -L-aryl, -L-heteroaryl, -L-C 1-10 alkylaryl, -L-C 1-10 alkylhetaryl, -L-C 1-10 alkylheterocylyl, -L-C 2-10 alkenyl, -L-C 2-10 alkynyl, -L-C 2-10 alkenyl-C 3-8 cycloalkyl, -L-C 2-10 alkynyl-C 3-8 cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocylyl, -L-heteroalkyl-C 3-8 cycloalkyl, -L-aralkyl, -L
  • L is absent, —(C ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O) N(R 31 )—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 N(R 31 )—, or —N(R 31 )—;
  • E 1 and E 2 are independently —(W 1 ) j —R 4 ;
  • M 1 is a 5, 6, 7, 8, 9, or -10 membered ring system, wherein the ring system is monocyclic or bicyclic, substituted with R 5 and additionally optionally substituted with one or more —(W 2 ) k —R 2 ;
  • each k is 0 or 1;
  • j in E 1 or j in E 2 is independently 0 or 1;
  • W 1 is —O—, —NR 7 —, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(R 7 )N(R 8 )S(O
  • W 2 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )C(O)N(R 8 )—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(
  • R 2 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • R 3 and R 4 are independently hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 , —NR 31 C( ⁇ NR 3 )SR 33 , —OC( ⁇
  • R 5 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • each of R 31 , R 32 , and R 33 is independently H or C 1-10 alkyl, wherein the C 1-10 alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or hetaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, hetaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • each of R 7′ and R 8′ is independently hydrogen, C 1-10 alkyl, C 2-10 alkenyl, aryl, heteroaryl, heterocyclyl or C 3-10 cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R 6 ;
  • R 6 is halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, hetaryl-C 1-10 alkyl, hetaryl-C 2-10 alkenyl, hetaryl-C 2-10 alkynyl, wherein each of said alkyl, alkeny
  • M 1 is a 5, 6, 7, 8, 9, or -10 membered ring system, wherein the ring system is monocyclic or bicyclic.
  • the monocyclic M 1 ring is unsubstituted or substituted with one or more R 5 substituents (including 0, 1, 2, 3, 4, or 5 R 5 substituents).
  • the monocyclic M 1 ring is aromatic (including phenyl) or heteroaromatic (including but not limited to pyridinyl, pyrrolyl, imidazolyl, thiazolyl, or pyrimidinyl).
  • the monocyclic M 1 ring may be a 5 or 6 membered ring (including but not limited to pyridinyl, pyrrolyl, imidazolyl, thiazolyl, or pyrimidinyl).
  • M 2 is a five membered heteroaromatic group with one heteroatom, wherein the heteroatom is N, S, or O. In another embodiment, M 2 is a five membered heteroaromatic group with two heteroatoms, wherein the heteroatoms are nitrogen and oxygen or nitrogen and sulfur.
  • the bicyclic M 1 ring is unsubstituted or substituted with one or more R 5 substituents (including 0, 1, 2, 3, 4, 5, 6 or 7 R 5 substituents).
  • Bicyclic M 1 ring is a 7, 8, 9, or 10 membered aromatic or heteroaromatic.
  • Examples of an aromatic bicyclic M 1 ring include naphthyl.
  • the bicyclic M 1 ring is heteroaromatic and includes but is not limited to benzothiazolyl, quinolinyl, quinazolinyl, benzoxazolyl, and benzoimidazolyl.
  • the invention also provides compounds wherein M 1 is a moiety having a structure of Formula M1-A or Formula M1-B:
  • W 1 , W 2 , and W 7 are independently N or C—R 5 ;
  • W 4 and W 10 are independently N—R 5 , O, or S;
  • W 6 and W 8 are independently N or C—R 5 ;
  • W 5 and W 9 are independently N or C—R 2 ;
  • W 3 is C or N, provided no more than two N and/or N—R 5 are adjacent and no two 0 or S are adjacent.
  • W 4 is N—R 5 , O, or S; W 6 is N or C—R 5 and W 5 is N or C—R 2 .
  • M 1 moiety of Formula M1-A include:
  • R 5 is —(W 1 ) k —R 53 or R 55 ; each k is independently 0 or 1, n is 0, 1, 2, or 3, and —(W 1 ) k —R 53 and R 55 are as defined above.
  • the M 1 moiety of Formula M1-B is a moiety of Formula M1-B1, Formula M1-B2, Formula M1-B3, or Formula M1-B4:
  • W 10 is N—R 5 , O, or S
  • W g is N or C—R 5
  • W 5 is N or C—R 2 .
  • M 1 moiety of Formula M1-B include:
  • R′ 5 is —(W 1 ) k —R 53 or R 55 ; k is 0 or 1, n is 0, 1, 2, or 3, and —(W 1 ) k —R 53 and R 55 are as defined above.
  • the invention also provides compounds wherein M 1 is a moiety having a structure of Formula M1-C or Formula M1-D:
  • W 12 , W 13 , W 14 , and W 15 are independently N or C—R 5 ;
  • W 11 and W 18 are independently N—R 5 , O, or S;
  • W 16 and W 17 are independently N or C—R 5 ; provided no more than two N are adjacent.
  • the M 1 moiety of Formula M1-C or Formula M1-D is a moiety of Formula M1-C1 or Formula M1-D1:
  • W 11 and W 18 are N—R 5 , O, or S; and W 16 and W 17 are N or C—R 5 .
  • M 1 moiety of Formula M1-C and Formula M1-D include:
  • R′ 5 is —(W 1 ) k —R 53 or R 55 ; k is 0 or 1, and —(W 1 ) k —R 53 and R 55 are as defined above.
  • the invention also provides compounds wherein M 1 is a moiety having a structure of Formula M1-E:
  • X 11 , X 12 , X 13 , X 14 , X 15 , X 16 , and X 17 are independently N, or C—R 5 ; provided that no more than two N are adjacent.
  • the M 1 moiety having a structure of Formula M1-E is a moiety having a structure of Formula M1-E1, M1-E2, M1-E3, M1-E4, M1-E5, M1-E6, M1-E7, or M1-E8:
  • the M 1 moiety having a structure of Formula M1-E is a moiety having a structure:
  • M 1 moiety of Formula M1-E include:
  • R′ 5 is —(W 1 ) k —R 53 or R 55 ; k is 0 or 1, n is 0, 1, 2, or 3, and —(W 1 ) k —R 53 or R 55 are as defined above. In some embodiments, k is 0, and R 5 is R 53 .
  • R 53 is hydrogen, unsubstituted or substituted C 1 -C 10 alkyl (which includes but is not limited to —CH 3 , —CH 2 CH 3 , n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or unsubstituted or substituted C 3 -C 8 cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl).
  • C 1 -C 10 alkyl which includes but is not limited to —CH 3 , —CH 2 CH 3 , n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl
  • C 3 -C 8 cycloalkyl
  • R 53 is monocyclic or bicyclic aryl, wherein the R 53 aryl is unsubstituted or substituted. Some examples of aryl include but are not limited to phenyl, naphthyl or fluorenyl. In some other embodiments, R 53 is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R 53 includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl.
  • Bicyclic heteroaryl R 53 includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, and purinyl.
  • R 53 may be alkylcycloalkyl (including but not limited to cyclopropylethyl, cyclopentylethyl, and cyclobutylpropyl), -alkylaryl (including but not limited to benzyl, phenylethyl, and phenylnaphthyl), alkylhetaryl (including but not limited to pyridinylmethyl, pyrrolylethyl, and imidazolylpropyl), or alkylheterocyclyl (non-limiting examples are morpholinylmethyl, 1-piperazinylmethyl, and azetidinylpropyl).
  • R 53 is unsubstituted or substituted C 2 -C 10 alkenyl (including but not limited to alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C 2 -C 10 alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl).
  • alkenyl including but not limited to alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl
  • unsubstituted or substituted alkynyl including but not limited to unsubstituted or substituted C 2 -C 10 alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl.
  • R 53 wherein R 53 is alkenylaryl, alkenylheteroaryl, alkenylheteroalkyl, or alkenylheterocycicyl, wherein each of alkenyl, aryl, heteroaryl, heteroalkyl, and heterocyclyl is as described herein and wherein the alkenylaryl, alkenylhetaryl, alkenylheteroalkyl, or alkenylheterocycicyl moiety is attached to M 1 through the alkenyl.
  • R 53 is -alkynylaryl, -alkynylhetaryl, -alkynylheteroalkyl, -alkynylheterocylyl, -alkynylcycloalkyl, or -alkynylC 3-8 cycloalkenyl, wherein each of alkynyl, aryl, heteroaryl, heteroalkyl, and heterocyclyl is as described herein and wherein the alkynylaryl, alkynylhetaryl, alkynylheteroalkyl, or alkynylheterocycicyl moiety is attached to M 1 through the alkynyl.
  • R 53 is -alkoxyalkyl, -alkoxyalkenyl, or -alkoxyalkynyl, wherein each of alkoxy, alkyl, alkenyl, and alkynyl is as described herein and wherein the -alkoxyalkyl, -alkoxyalkenyl, or -alkoxyalkynyl moiety is attached to M 1 through the alkoxy.
  • R 53 is -heterocyclylalkyl, -heterocyclylalkenyl, or -heterocyclylalkynyl, wherein the heterocyclyl, alkyl, alkenyl, or alkynyl is as described herein and wherein the -heterocyclylalkyl, -heterocyclylalkenyl, or -heterocyclylalkynyl is attached to M 1 through the heterocyclyl portion of the moiety.
  • R 53 may be aryl-alkenyl, aryl-alkynyl, or aryl-heterocyclyl, wherein the aryl, alkenyl, alkynyl, or heterocyclyl is as described herein and wherein the aryl-alkenyl, aryl-alkynyl, or aryl-heterocyclyl moiety is attached to M 1 through the aryl portion of the moiety.
  • R 53 is heteroaryl-alkyl, heteroaryl-alkenyl, heteroaryl -alkynyl, heteroaryl-cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of heteroaryl, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, and heterocyclyl is as described herein and wherein the heteroaryl-alkyl, heteroaryl-alkenyl, heteroaryl-alkynyl, heteroaryl-cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl moiety is attached to M 1 through the heteroaryl portion of the moiety.
  • the aryl or heteroaryl is unsubstituted or is substituted with one or more independent halo, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —NR 31 R 32 , —C( ⁇ O)NNR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)OR 31 , —C( ⁇ O)
  • each of the alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moieties forming part of all of R 53 is unsubstituted or substituted with one or more halo, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —O-aryl, —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NNR 34 R 35 , or —C( ⁇ O)NR 31 R 32 substituents.
  • R 5 is —W 1 —R 53 .
  • R 5 is —OR 53 , including but not limited to Oalkyl (including but not limited to methoxy or ethoxy), -Oaryl (including but not limited to phenoxy), —O-heteroaryl (including but not limited to pyridinoxy) and —O-heterocycloxy(including but not limited to 4-N-piperidinoxy).
  • R 5 is —NR 6 R 53 including but not limited to anilinyl, diethylamino, and 4-N-piperidinylamino.
  • R 5 is —S(O) 0-2 R 53 , including but not limited to phenylsulfonyl and pyridinylsulfonyl.
  • the invention also provides compounds wherein R 5 is —C(O) (including but not limited to acetyl, benzoyl, and pyridinoyl) or —C(O)O R 53 (including but not limited to carboxyethyl, and carboxybenzyl).
  • R 5 is —C(O)N(R 6 )R 53 (including but not limited to C(O)NH(cyclopropyl) and C(O)N(Me)(phenyl)) or —CH(R 6 )N(R 7 )R 53 (including but not limited to —CH 2 —NH-pyrrolidinyl, CH 2 —NHcyclopropyl, and CH 2 -anilinyl).
  • R 5 is —N(R 6 )C(O)R 53 (including but not limited to —NHC(O)phenyl, —NHC(O)cyclopentyl, and to —NHC(O)piperidinyl) or —N(R 6 )S(O) 2 R 53 (including but not limited to —NHS(O) 2 phenyl, —NHS(O) 2 piperazinyl, and —NHS(O) 2 methyl.
  • R 5 is —N(R 6 )S(O) R 53 , —CH(R 6 )N(C(O)OR 7 ) R 53 , —CH(R 7 )N(C(O)R 7 ) R 53 , —CH(R 6 )N(SO 2 R 7 ) R 53 , —CH(R 6 )N(R 7 ) R 53 , —CH(R 6 )C(O)N(R 7 ) R 53 , —CH(R 6 )N(R 7 )C(O) R 53 , —CH(R 6 )N(R 7 )S(O) R 53 , or —CH(R 6 )N(R 7 )S(O) 2 R 53 .
  • R 5 is R 55 .
  • R 55 is halo, —OH, —NO 2 , —CF 3 , —OCF 3 , or —CN.
  • R 55 is —R 31 , —OR 31 (including but not limited to methoxy, ethoxy, and butoxy) —C(O)R 31 (non-limiting examples include acetyl, propionyl, and pentanoyl), or —CO 2 R 31 (including but not limited to carboxymethyl, carboxyethyl and carboxypropyl).
  • R 55 is —NR 31 R 32 , —C( ⁇ O)NR 31 R 32 , —SO 2 NR 31 R 32 , or —S(O) 0-2 R 31 .
  • R 55 is —NR 34 R 35 or —SO 2 NR 34 R 35 , wherein R 34 R 35 are taken together with the nitrogen to which R 34 R 35 are attached to form a cyclic moiety.
  • the cyclic moiety so formed may be unsubstituted or substituted, wherein the substituents are selected from the group consisting of alkyl, —C(O)alkyl, —S(O) 2 alkyl, and —S(O) 2 aryl.
  • R 55 is —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 , —NR 31 C( ⁇ NR 32 )SR 33 , —OC( ⁇ O)OR 33 , —OC( ⁇ O)NR 31 R 32 , —C( ⁇ O)NNR 34 R 35 , —OC( ⁇ O)SR 31 , —SC( ⁇ O)OR 31 , —P(O)OR 31 OR 32 , or —SC( ⁇ O)NR 31 R 32
  • the invention further provides a compound which is an mTor inhibitor, wherein the compound has the Formula I-A:
  • X 1 is N or C-E 1 , X 2 is N, X 3 is C, and X 4 is C—R 9 or N; or X 1 is N or C-E 1 , X 2 is C, X 3 is N, and X 4 is C—R 9 or N;
  • R 1 is —H, -L-C 1-10 alkyl, -L-C 3-8 cycloalkyl, -L-C 1-10 alkyl-C 3-8 cycloalkyl, -L-aryl, -L-heteroaryl, -L-C 1-10 alkylaryl, -L-C 1-10 alkylheteroaryl, -L-C 1-10 alkylheterocyclyl, -L-C 2-10 alkenyl, -L-C 2-10 alkynyl, -L-C 2-10 alkenyl-C 3-8 cycloalkyl, -L-C 2-10 alkynyl-C 3-8 cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C
  • L is absent, —(C ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O) N(R 31 )—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 N(R 31 )—, or —N(R 31 )—;
  • M 1 is a moiety having the structure of Formula M1-F1 or M1-F2:
  • k 0 or 1
  • E 1 and E 2 are independently —(W 1 ) j —R 4 ;
  • j in each instance (i.e., in E 1 or j in E 2 ), is independently 0 or 1
  • W 1 is —O—, —NR 7 —, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(R 7 )N(R 8 )S(O
  • W 2 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )C(O)N(R 8 )—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(
  • R 2 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 , —NR 31 C( ⁇ O
  • R 3 and R 4 are independently hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR
  • R 5 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • R 31 , R 32 , and R 33 are independently H or C 1-10 alkyl, wherein the C 1-10 alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, heteroaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • R 7′ and R 8′ are each independently hydrogen, C 1-10 alkyl, C 2-10 alkenyl, aryl, heteroaryl, heterocyclyl or
  • R 6 is halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, heteroaryl-C 2-10 alkenyl, heteroaryl-C 2-10 alkynyl, wherein each of said alkyl, alkenyl, alkyny
  • R 9 is H, halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, heteroaryl-C 2-10 alkenyl, heteroaryl-C 2-10 alkynyl, wherein each of said alkyl, alkenyl, alky
  • X 4 is C—R 9 .
  • the invention also provides an inhibitor as defined above, wherein the compound is of Formula I:
  • the compound of Formula I-B or its pharmaceutically acceptable salt thereof is a compound having the structure of Formula I-B1 or Formula I-B2:
  • X 1 is N and X 2 is N. In other embodiments, X 1 is C-E 1 and X 2 is N. In yet other embodiments, X 1 is NH and X 2 is C. In further embodiments, X 1 is CH-E 1 and X 2 is C.
  • X 1 is N and X 2 is C. In further embodiments, X 1 is C-E 1 and X 2 is C.
  • X 1 is C—(W 1 ) j —R 4 , where j is 0.
  • X 1 is CH. In yet another embodiment, X 1 is C-halogen, where halogen is Cl, F, Br, or I.
  • X 1 it is C—(W 1 ), —R 4 .
  • W 1 is —O—.
  • W 1 is —NR 7 —.
  • W 1 is —NH—.
  • W 1 is —S(O) 0-2 -.
  • W 1 is —C(O)—.
  • W 1 is —C(O)N(R 7 )—.
  • X 1 , j is 1, and W 1 is —N(R 7 )C(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O) 2 —. In various embodiments of X 1 , j is 1, and W 1 is —C(O)O—. In various embodiments of X 1 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 )—.
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )C(O)—.
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • X 1 is CH 2 . In yet another embodiment, X 1 is CH-halogen, where halogen is Cl, F, Br, or I.
  • X 1 is N.
  • X 2 is N. In other embodiments, X 2 is C.
  • E 2 is —(W 1 ) j —R 4 , where j is 0.
  • E 2 is CH. In yet another embodiment, E 2 is C-halogen, where halogen is Cl, F, Br, or I.
  • E 2 it is —(W 1 ) j —R 4 .
  • j is 1, and W 1 is —O—.
  • W 1 is —NR 7 —.
  • W 1 is —NH—.
  • W 1 is —S(O) 0-2 -.
  • W 1 is —C(O)—.
  • j is 1, and W 1 is —C(O)N(R 7 )—.
  • E 2 j is 1, and W 1 is —N(R 7 )C(O)—. In various embodiments of E 2 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —N(R 7 )S(O) 2 —. In various embodiments of E 2 , j is 1, and W 1 is —C(O)O—. In various embodiments of E 2 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 )—.
  • E 2 j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )C(O)—.
  • E 2 j is 1, and W 1 is —CH(R)N(R 8 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • M 1 when M 1 is a moiety of Formula M1-F1, M 1 is benzoxazolyl substituted with —(W 2 ) k —R 2 . In some embodiments, M 1 is a benzoxazolyl substituted at the 2-position with (W 2 ) j —R 2 , In some embodiments, M 1 is either a 5-benzoxazolyl or a 6-benzoxazolyl moiety, optionally substituted at the 2-position with —(W 2 ) j —R 2 .
  • Exemplary Formula M1-F1 M 1 moieties include but are not limited to the following:
  • Formula M1-F2 is an aza-substituted benzoxazolyl moiety having a structure of one of the following formulae:
  • Exemplary Formula M1-F2 M 1 moieties include but are not limited to the following:
  • M 1 In various embodiments of M 1 , k is 0. In other embodiments of M 1 , k is 1, and W 2 is selected from one of the following: —O—, —NR 7 —, —S(O) 0-2 , —C(O)—, —C(O)N(R 7 ), —N(R 7 )C(O), or N(R 7 )C(O)N(R 8 ).
  • k is 1, and W 2 is —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 ), —CH(R 7 )N(C(O)R 8 ), or —CH(R 7 )N(SO 2 R 8 ).
  • k is 1, and W 2 is —CH(R 7 )N(R 8 ), —CH(R 7 )C(O)N(R 8 ), —CH(R 7 )N(R 8 )C(O), or —CH(R 7 )N(R 8 )S(O)—.
  • k is 1, and W 2 is CH(R 7 )N(R 8 )S(O) 2 .
  • the invention provides an inhibitor of mTor which is a compound of Formula I-C or Formula I-D:
  • X 1 is N or C-E 1 , X 2 is N, and X 3 is C; or X 1 is N or C-E 1 , X 2 is C, and X 3 is N;
  • R 1 is —H, -L-C 1-10 alkyl, -L-C 3-8 cycloalkyl, -L-C 1-10 alkyl-C 3-8 cycloalkyl, -L-aryl, -L-heteroaryl, -L-C 1-10 alkylaryl, -L-C 1-10 alkylheteroaryl, -L-C 1-10 alkylheterocyclyl, -L-C 2-10 alkenyl, -L-C 2-10 alkynyl, -L-C 2-10 alkenyl-C 3-8 cycloalkyl, -L-C 2-10 alkynyl-C 3-8 cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C
  • L is absent, —(C ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O) N(R 31 )—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 N(R 31 )—, or —N(R 31 )—;
  • E 1 and E 2 are independently —(W 1 ) j —R 4 ;
  • j in E 1 or j in E 2 is independently 0 or 1;
  • W 1 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(R 7 )N(R 8 )S(O) 2 —;
  • W 2 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )C(O)N(R 8 )—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(
  • k 0 or 1
  • R 2 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 , —NR 31 C( ⁇ O
  • R 3 and R 4 are independently hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33
  • R 5 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • R 31 , R 32 , and R 33 are independently H or C 1-10 alkyl, wherein the C 1-10 alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, heteroaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom; and
  • R 7′ and R 8′ are each independently hydrogen, C 1-10 alkyl, C 2-10 alkenyl, aryl, heteroaryl, heterocyclyl or C 3-10 cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R 6 ; and R 6 is halo, —OR 31 , —SH, NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O) NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, or C 2-10 alkynyl; or R 6 is aryl-C 1-10 al
  • the compound of Formula I-C has a structure of Formula I-C1 or Formula I-C2:
  • X 1 is N and X 2 is N. In other embodiments, X 1 is C-E 1 and X 2 is N. In yet other embodiments, X 1 is NH and X 2 is C. In further embodiments, X 1 is CH-E 1 and X 2 is C.
  • X 1 is N and X 2 is C. In yet other embodiments, X 1 is NH and X 2 is C. In further embodiments, X 1 is CH-E 1 and X 2 is C.
  • the compound of Formula I-D has a structure of Formula I-D1 or Formula I-D2:
  • X 1 is N and X 2 is N. In other embodiments, X 1 is C-E 1 and X 2 is N. In yet other embodiments, X 1 is NH and X 2 is C. In further embodiments, X 1 is CH-E 1 and X 2 is C.
  • X 1 is N and X 2 is C. In further embodiments, X 1 is C-E 1 and X 2 is C.
  • X 1 is C—(W 1 ) j —R 4 , where j is 0.
  • X 1 is CH. In yet another embodiment, X 1 is C-halogen, where halogen is Cl, F, Br, or I.
  • X 1 it is C—(W 1 ), —R 4 .
  • W 1 is —O—.
  • W 1 is —NR 7 —.
  • W 1 is —NH—.
  • W 1 is —S(O) 0-2 .
  • W 1 is —C(O)—.
  • W 1 is —C(O)N(R 7 ).
  • X 1 , j is 1, and W 1 is —N(R 7 )C(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O) 2 . In various embodiments of X 1 , j is 1, and W 1 is —C(O)O—. In various embodiments of X 1 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 ).
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )C(O)—.
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • X 1 is CH—(W 1 ) j —R 4 , where j is 0.
  • X 1 is CH 2 . In yet another embodiment, X 1 is CH-halogen, where halogen is Cl, F, Br, or I.
  • X 1 it is CH—(W 1 ) j —R 4 .
  • W 1 is —O—.
  • W 1 is NR 7 —.
  • W 1 is NH—.
  • W 1 is —S(O) 0-2 .
  • W 1 is —C(O)—.
  • W 1 is —C(O)N(R 7 ).
  • X 1 , j is 1, and W 1 is N(R 7 )C(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O) 2 . In various embodiments of X 1 , j is 1, and W 1 is —C(O)O—. In various embodiments of X 1 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 ).
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is CH(R 7 )N(R 8 )C(O)—.
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is CH(R 7 )N(R 8 )S(O) 2 —.
  • X 1 is N.
  • X 2 is N. In other embodiments, X 2 is C.
  • E 2 is (W 1 ) j —R 4 , where j is 0.
  • E 2 is CH. In yet another embodiment, E 2 is C-halogen, where halogen is Cl, F, Br, or I.
  • E 2 it is (W 1 ) j —R 4 .
  • j is 1, and W 1 is —O—.
  • W 1 is NR 7 —.
  • W 1 is NH—.
  • W 1 is —S(O) 0-2 .
  • W 1 is —C(O)—.
  • j is 1, and W 1 is —C(O)N(R 7 ).
  • E 2 j is 1, and W 1 is N(R 7 )C(O)—. In various embodiments of E 2 , j is 1, and W 1 is N(R 7 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —N(R 7 )S(O) 2 . In various embodiments of E 2 , j is 1, and W 1 is —C(O)O—. In various embodiments of E 2 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 ). In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—.
  • E 2 j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is CH(R 7 )N(R 8 )C(O)—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • k is 0. In other embodiments, k is 1 and W 2 is —O—. In another embodiment, k is 1 and W 2 is NR 7 . In yet another embodiment of, k is 1, and W 2 is —S(O) 0-2 . In another embodiment of, k is 1 and W 2 is —C(O)—. In a further embodiment, k is 1 and W 2 is —C(O)N(R 7 ). In another embodiment, k is 1, and W 2 is N(R 7 )C(O)—. In another embodiment, k is 1 and W 2 is N(R 7 )C(O)N(R 8 ).
  • k is 1 and W 2 is —N(R 7 )S(O)—. In still yet another embodiment, k is 1 and W 2 is —N(R 7 )S(O) 2 . In a further embodiment, k is 1 and W 2 is —C(O)O—. In another embodiment, k is 1 and W 2 is CH(R 7 )N(C(O)OR 8 ). In another embodiment, k is 1 and W 2 is —CH(R 7 )N(C(O)R 8 )—. In another embodiment, k is 1 and W 2 is —CH(R 7 )N(SO 2 R 8 )—.
  • k is 1 and W 2 is —CH(R 7 )N(R 8 )—. In another embodiment, k is 1 and W 2 is CH(R 7 )C(O)N(R 8 ). In yet another embodiment, k is 1 and W 2 is CH(R 7 )N(R 8 )C(O)—. In another embodiment, k is 1 and W 2 is —CH(R 7 )N(R 8 )S(O)—. In yet another embodiment, k is 1 and W 2 is CH(R 7 )N(R 8 )S(O) 2 —.
  • the invention also provides a compound which is an mTor inhibitor of Formula I-E:
  • X 1 is N or C-E 1 , X 2 is N, and X 3 is C; or X 1 is N or C-E 1 , X 2 is C, and X 3 is N;
  • R 1 is —H, L-C 1-10 alkyl, -L-C 3-8 cycloalkyl, -L-C 1-10 alkyl-C 3-8 cycloalkyl, -L-aryl, -L-heteroaryl, -L-C 1-10 alkylaryl, -L-C 1-10 alkylheteroaryl, -L-C 1-10 alkylheterocyclyl, -L-C 2-10 alkenyl, -L-C 2-10 alkynyl, -L-C 2-10 alkenyl-C 3-8 cycloalkyl, -L-C 2-10 alkynyl-C 3-8 cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C 3-8
  • L is absent, —(C ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O) N(R 31 )—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 N(R 31 )—, or —N(R 31 )—;
  • M 1 is a moiety having the structure of Formula M1-F1 or Formula M1-F2:
  • k 0 or 1
  • E 1 and E 2 are independently (W 1 ); —R 4 ;
  • j in E 1 or j in E 2 is independently 0 or 1;
  • W 1 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O), —N(R 7 )S(O)—, —N(R 7 )S(O) 2 , —C(O)O, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 ), —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(R 7 )N(R 8 )S(O) 2 —;
  • W 2 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )C(O)N(R 8 ), —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(R 7
  • R 2 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • R 5 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • R 31 , R 32 , and R 33 are independently H or C 1-10 alkyl, wherein the C 1-10 alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, heteroaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • R 7′ and R 8′ are each independently hydrogen, C 1-10 alkyl, C 2-10 alkenyl, aryl, heteroaryl, heterocyclyl or C 3-10 cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R 6 ;
  • R 6 is halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, heteroaryl-C 2-10 alkenyl, heteroaryl-C 2-10 alkynyl, wherein each of said alkyl, alkenyl, alkyny
  • R 9 is H, halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, heteroaryl-C 2-10 alkenyl, heteroaryl-C 2-10 alkynyl, wherein each of said alkyl, alkenyl, alky
  • the compound of Formula I-E has a structure of Formula I-E1 or Formula I-E2:
  • X 1 is N and X 2 is N. In other embodiments, X 1 is C-E 1 and X 2 is N. In yet other embodiments, X 1 is NH and X 2 is C. In further embodiments, X 1 is CH-E 1 and X 2 is C.
  • X 1 is N and X 2 is C. In further embodiments, X 1 is C-E 1 and X 2 is C.
  • X 1 is C—(W 1 ) j —R 4 , where j is 0.
  • X 1 is CH. In yet another embodiment, X 1 is C-halogen, where halogen is Cl, F, Br, or I.
  • X 1 it is C—(W 1 ), —R 4 .
  • W 1 is —O—.
  • W 1 is —NR 7 —.
  • W 1 is —NH—.
  • W 1 is —S(O) 0-2 -.
  • W 1 is —C(O)—.
  • W 1 is —C(O)N(R 7 )—.
  • X 1 , j is 1, and W 1 is —N(R 7 )C(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O) 2 —. In various embodiments of X 1 , j is 1, and W 1 is —C(O)O—. In various embodiments of X 1 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 )—.
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )C(O)—.
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • X 1 is N.
  • X 2 is N. In other embodiments, X 2 is C.
  • E 2 is —(W 1 ) j —R 4 , where j is 0.
  • E 2 is CH. In yet another embodiment, E 2 is C-halogen, where halogen is Cl, F, Br, or I.
  • E 2 it is —(W 1 ), —R 4 .
  • j is 1, and W 1 is —O—.
  • W 1 is —NR 7 —.
  • W 1 is —NH—.
  • W 1 is —S(O) 0-2 -.
  • W 1 is —C(O)—.
  • j is 1, and W 1 is —C(O)N(R 7 )—.
  • E 2 j is 1, and W 1 is —N(R 7 )C(O)—. In various embodiments of E 2 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —N(R 7 )S(O) 2 —. In various embodiments of E 2 , j is 1, and W 1 is —C(O)O—. In various embodiments of E 2 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 )—.
  • E 2 j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )C(O)—.
  • E 2 j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • M 1 when M 1 is a moiety of Formula I-E1, M 1 is benzoxazolyl substituted with (W 2 ) k —R 2 . In some embodiments, M 1 is a benzoxazolyl moiety, substituted at the 2-position with (W 2 ) k —R 2 . In some embodiments, M 1 is either a 5-benzoxazolyl or a 6-benzoxazolyl moiety, optionally substituted with (W 2 ) k —R 2 .
  • Exemplary Formula I-E1 M 1 moieties include but are not limited to the following:
  • Formula I-E2 is an aza-substituted benzoxazolyl moiety having a structure of one of the following formulae:
  • Exemplary Formula I-E2 M 1 moieties include but are not limited to the following:
  • M 1 In various embodiments of M 1 , k is 0. In other embodiments of M 1 , k is 1 and W 2 is —O—. In another embodiment of M 1 , k is 1 and W 2 is —NR 7 —. In yet another embodiment of M 1 , k is 1 and W 2 is S(O) 0-2 -. In another embodiment of M 1 , k is 1 and W 2 is —C(O)—. In a further embodiment of M 1 , k is 1 and W 2 is —C(O)N(R 7 )—. In another embodiment of M 1 , k is 1 and W 2 is N(R 7 )C(O)—.
  • k is 1 and W 2 is N(R 7 )C(O)N(R 8 ). In yet another embodiment of M 1 , k is 1 and W 2 is —N(R 7 )S(O)—. In still yet another embodiment of M 1 , k is 1 and W 2 is —N(R 7 )S(O) 2 . In a further embodiment of M 1 , k is 1 and W 2 is —C(O)O—. In another embodiment of M 1 , k is 1 and W 2 is CH(R 7 )N(C(O)OR 8 ). In another embodiment of M 1 , k is 1 and W 2 is —CH(R 7 )N(C(O)R 8 )—.
  • M 1 k is 1 and W 2 is —CH(R 7 )N(SO 2 R 8 )—. In a further embodiment of M 1 , k is 1 and W 2 is —CH(R 7 )N(R 8 )—. In another embodiment of M 1 , k is 1 and W 2 is —CH(R 7 )C(O)N(R 8 )—. In yet another embodiment of M 1 , k is 1 and W 2 is —CH(R 7 )N(R 8 )C(O)—. In another embodiment of M 1 , k is 1 and W 2 is —CH(R 7 )N(R 8 )S(O)—. In yet another embodiment of M 1 , k is 1 and W 2 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • L is absent.
  • L is —(C ⁇ O)—.
  • L is C( ⁇ O)O—.
  • L is —C( ⁇ O) NR 31 —.
  • L is —S—.
  • L is —S(O)—.
  • L is —S(O) 2 —.
  • L is —S(O) 2 NR 31 —.
  • L is —NR 31 —.
  • R 1 is L-C 1-10 alkyl, which is unsubstituted. In another embodiment, R 1 is L-C 1-10 alkyl, which is substituted by one or more independent R 3 . In yet another embodiment, R 1 is L-unsubstituted C 1-10 alkyl, where L is absent. In another embodiment, R 1 is L-C 1-10 alkyl, which is substituted by one or more independent R 3 , and L is absent.
  • R 1 is -L-C 3-8 cycloalkyl, which is unsubstituted.
  • R 1 is L-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 .
  • R 1 is -L-C 3-8 cycloalkyl, which is unsubstituted, and L is absent.
  • R 1 is -L-C 3-8 cycloalkyl which is substituted by one or more independent R 3 , and L is absent.
  • R 1 is H.
  • R 1 is -L-aryl, which is unsubstituted. In another embodiment, R 1 is -L-aryl, which is substituted by one or more independent R 3 . In another embodiment, R 1 is -L-aryl which is unsubstituted, and L is absent. In yet another embodiment, R 1 is -L-aryl, which is substituted by one or more independent R 3 , and L is absent.
  • R 1 is -L-heteroaryl, which is unsubstituted.
  • R 1 is -L-heteroaryl, which is substituted by one or more independent R 3 .
  • R 1 is -L-heteroaryl which is unsubstituted and L is absent.
  • R 1 is -L-heteroaryl, which is substituted by one or more independent R 3 , and L is absent.
  • R 1 is -L-C 1-10 alkyl-C 3-8 cycloalkyl, which is unsubstituted.
  • R 1 is -L-C 1-10 alkyl-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 .
  • R 1 is -L-C 1-10 alkyl-C 3-8 cycloalkyl which is unsubstituted and L is absent.
  • R 1 is -L-C 1-10 alkyl-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 , and L is absent.
  • R 1 is -L-C 1-10 alkylaryl, which is unsubstituted.
  • R 1 is -L-C 1-10 alkylaryl, which is substituted by one or more independent R 3 .
  • R 1 is -L-C 1-10 alkylaryl which is unsubstituted and L is absent.
  • R 1 is -L-C 1-10 alkylaryl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-C 1-10 alkylheteroaryl, which is unsubstituted.
  • R 1 is -L-C 1-10 alkylheteroaryl, which is substituted by one or more independent R 3 .
  • R 1 is -L-C 1-10 alkylheteroaryl which is unsubstituted and L is absent.
  • R 1 is -L-C 1-10 alkylheteroaryl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-C 1-10 alkylheterocyclyl, which is unsubstituted.
  • R 1 is -L-C 1-10 alkylheterocyclyl, which is substituted by one or more independent R 3 .
  • R 1 is -L-C 1-10 alkylheterocyclyl which is unsubstituted and L is absent.
  • R 1 is -L-C 1-10 alkylheterocyclyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-C 2-10 alkenyl, which is unsubstituted. In another embodiment, R 1 is -L-C 2-10 alkenyl which is substituted by one or more independent R 3 . In a further embodiment, R 1 is -L-C 2-10 alkenyl which is unsubstituted and L is absent. In yet another embodiment, R 1 is -L-C 2-10 alkenyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-C 2-10 alkynyl, which is unsubstituted. In another embodiment, R 1 is -L-C 2-10 alkynyl which is substituted by one or more independent R 3 . In a further embodiment, R 1 is -L-C 2-10 alkynyl which is unsubstituted and L is absent. In yet another embodiment, R 1 is -L-C 2-10 alkynyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-C 2-10 alkenyl-C 3-8 cycloalkyl, which is unsubstituted.
  • R 1 is -L-C 2-10 alkenyl-C 3-8 cycloalkyl which is substituted by one or more independent R 3 .
  • R 1 is -L-C 2-10 alkenyl-C 3-8 cycloalkyl which is unsubstituted and L is absent.
  • R 1 is -L-C 2-10 alkenyl-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl, which is unsubstituted.
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl which is substituted by one or more independent R 3 .
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl which is unsubstituted and L is absent.
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl, which is unsubstituted.
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl which is substituted by one or more independent R 3 .
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl which is unsubstituted and L is absent.
  • R 1 is -L-C 2-10 alkynyl-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-heteroalkyl, which is unsubstituted. In another embodiment, R 1 is -L-heteroalkyl which is substituted by one or more independent R 3 . In a further embodiment, R 1 is -L-heteroalkyl which is unsubstituted and L is absent. In yet another embodiment, R 1 is -L-heteroalkyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-heteroalkylaryl, which is unsubstituted.
  • R 1 is -L-heteroalkylaryl which is substituted by one or more independent R 3 .
  • R 1 is -L-heteroalkylaryl which is unsubstituted and L is absent.
  • R 1 is -L-heteroalkylaryl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-heteroalkylheteroaryl, which is unsubstituted.
  • R 1 is -L-heteroalkylheteroaryl, which is substituted by one or more independent R 3 .
  • R 1 is -L-heteroalkylheteroaryl which is unsubstituted and L is absent.
  • R 1 is -L-heteroalkylheteroaryl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-heteroalkyl-heterocyclyl, which is unsubstituted.
  • R 1 is -L-heteroalkyl-heterocyclyl, which is substituted by one or more independent R 3 .
  • R 1 is -L-heteroalkyl-heterocyclyl which is unsubstituted, and L is absent.
  • R 1 is -L-heteroalkyl-heterocyclyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-heteroalkyl-C 3-8 cycloalkyl, which is unsubstituted.
  • R 1 is -L-heteroalkyl-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 .
  • R 1 is -L-heteroalkyl-C 3-8 cycloalkyl which is unsubstituted and L is absent.
  • R 1 is -L-heteroalkyl-C 3-8 cycloalkyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-aralkyl, which is unsubstituted. In another embodiment, R 1 is -L-aralkyl, which is substituted by one or more independent R 3 . In a further embodiment, R 1 is -L-aralkyl which is unsubstituted. In yet another embodiment, R 1 is -L-aralkyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-heteroaralkyl, which is unsubstituted.
  • R 1 is -L-heteroaralkyl, which is substituted by one or more independent R 3 .
  • R 1 is -L-heteroaralkyl which is unsubstituted and L is absent.
  • R 1 is -L-heteroaralkyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is -L-heterocyclyl, which is unsubstituted.
  • R 1 is -L-heterocyclyl, which is substituted by one or more independent R 3 .
  • R 1 is -L-heterocyclyl which is unsubstituted and L is absent.
  • R 1 is -L-heterocyclyl, which is substituted by one or more independent R 3 , where L is absent.
  • R 1 is a substituent as shown below:
  • R 2 is hydrogen. In another embodiment, R 2 is halogen. In another embodiment, R 2 is OH. In another embodiment, R 2 is R 31 . In another embodiment, R 2 is CF 3 . In another embodiment, R 2 is OCF 3 . In another embodiment, R 2 is —OR 31 . In another embodiment, R 2 is NR 31 R 32 . In another embodiment, R 2 is NR 34 R 35 . In another embodiment, R 2 is —C(O)R 31 . In another embodiment, R 2 is CO 2 R 31 . In another embodiment, R 2 is —C( ⁇ O)NR 31 R′′. In another embodiment, R 2 is —C( ⁇ O)NR 34 R 35 .
  • R 2 is —NO 2 . In another embodiment, R 2 is —CN. In another embodiment, R 2 is —S(O) 0-2 R 3 . In another embodiment, R 2 is —SO 2 NR 31 R 32 . In another embodiment, R 2 is —SO 2 NR 34 R 35 . In another embodiment, R 2 is —NR 31 C( ⁇ O)R 32 . In another embodiment, R 2 is —NR 31 C( ⁇ O)OR 32 . In another embodiment, R 2 is —NR 31 C( ⁇ O)NR 32 R 33 . In another embodiment, R 2 is —NR 31 S(O) 0-2 R 32 . In another embodiment, R 2 is —C( ⁇ S)OR 31 .
  • R 2 is —C( ⁇ O)SR 31 .
  • R 2 is —NR 31 C( ⁇ NR 32 )NR 33 R 32 .
  • R 2 is —NR 31 C( ⁇ NR 32 )OR 33 .
  • R 2 is —NR 31 C( ⁇ NR 32 )SR 33 .
  • R 2 is —OC( ⁇ O)OR 33 .
  • R 2 is —OC( ⁇ O)NR 31 R 32 .
  • R 2 is —OC( ⁇ O)SR 31 .
  • R 2 is —SC( ⁇ O)OR 31 .
  • R 2 is —P(O)OR 31 OR 32 .
  • R 2 is —SC( ⁇ O)NR 31 R 32 .
  • R 2 is monocyclic aryl.
  • R 2 is bicyclic aryl.
  • R 2 is substituted monocyclic aryl.
  • R 2 is heteroaryl.
  • R 2 is C 1-4 alkyl.
  • R 2 is C 1-10 alkyl.
  • R 2 is C 3-8 cycloalkyl.
  • R 2 is C 3-8 cycloalkyl-C 1-10 alkyl.
  • R 2 is C 1-10 alkyl-C 3-8 cycloalkyl.
  • R 2 is C 1-10 alkyl-monocyclic aryl.
  • R 2 is C 2-10 alkyl-monocyclic aryl. In another embodiment, R 2 is monocyclic aryl-C 2-10 alkyl. In another embodiment, R 2 is C 1-10 alkyl-bicyclicaryl. In another embodiment, R 2 is bicyclicaryl-C 1-10 alkyl. In another embodiment, R 2 is —C 1-10 alkylheteroaryl. In another embodiment, R 2 is —C 1-10 alkylheterocyclyl. In another embodiment, R 2 is —C 2-10 alkenyl. In another embodiment, R 2 is —C 2-10 alkynyl. In another embodiment, R 2 is C 2-10 alkenylaryl.
  • R 2 is C 2-10 alkenylheteroaryl. In another embodiment, R 2 is C 2-10 alkenylheteroalkyl. In another embodiment, R 2 is C 2-10 alkenylheterocycicyl. In another embodiment, R 2 is —C 2-10 alkynylaryl. In another embodiment, R 2 is —C 2-10 alkynylheteroaryl. In another embodiment, R 2 is —C 2-10 alkynylheteroalkyl. In another embodiment, R 2 is —C 2-10 alkynylheterocyclyl. In another embodiment, R 2 is —C 2-10 alkynylC 3-8 cycloalkyl.
  • R 2 is —C 2-10 alkynylC 3-8 cycloalkenyl. In another embodiment, R 2 is —C 1-10 alkoxy-C 1-10 alkyl. In another embodiment, R 2 is —C 1-10 alkoxy-C 2-10 alkenyl. In another embodiment, R 2 is —C 1-10 alkoxy-C 2-10 alkynyl. In another embodiment, R 2 is -heterocyclyl C 1-10 alkyl. In another embodiment, R 2 is heterocyclylC 2-10 alkenyl. In another embodiment, R 2 is heterocyclylC 2-10 alkynyl. In another embodiment, R 2 is aryl-C 2-10 alkyl.
  • R 2 is aryl-C 1-10 alkyl. In another embodiment, R 2 is aryl-C 2-10 alkenyl. In another embodiment, R 2 is aryl-C 2-10 alkynyl. In another embodiment, R 2 is aryl-heterocyclyl. In another embodiment, R 2 is heteroaryl-C 1-10 alkyl. In another embodiment, R 2 is heteroaryl-C 2-10 alkenyl. In another embodiment, R 2 is heteroaryl-C 2-10 alkynyl. In another embodiment, R 2 is heteroaryl-C 3-8 cycloalkyl. In another embodiment, R 2 is heteroaryl-heteroalkyl. In another embodiment, R 2 is heteroaryl-heterocyclyl.
  • R 2 when R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is unsubstituted.
  • R 2 when R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent halo.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OH.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —R 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —CF 3 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OCF.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OR 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 34 R 35 .
  • R 4 when R 4 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C(O)R 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —CO 2 R 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C( ⁇ O)NR 31 R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C( ⁇ O)NR 34 R 35 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NO 2 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —CN.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —S(O) 0-2 R 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SO 2 NR 31 R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SO 2 NR 34 R 35 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent NR 31 C( ⁇ O)R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ O)OR 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ O)NR 32 R 33 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 S(O) 0-2 R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C( ⁇ S)OR 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C( ⁇ O)SR 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ NR 32 )NR 33 R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent, —NR 31 C( ⁇ NR 32 )OR 33 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ NR 32 )SR 33 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)OR 33 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)NR 31 R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)SR 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SC( ⁇ O)OR 31 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —P(O)OR 31 OR 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SC( ⁇ O)NR 31 R 32 .
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent alkyl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent heteroalkyl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent alkenyl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent alkynyl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent cycloalkyl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent heterocycloalkyl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent aryl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent arylalkyl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent heteroaryl.
  • R 2 is bicyclic aryl, monocyclic aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, monocyclic aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent heteroarylalkyl.
  • R 3 is hydrogen. In another embodiment, R 3 is halogen. In another embodiment, R 3 is OH. In another embodiment, R 3 is R 31 . In another embodiment, R 3 is CF 3 . In another embodiment, R 3 is OCF 3 . In another embodiment, R 3 is —OR 31 . In another embodiment, R 3 is NR 31 R 32 . In another embodiment, R 3 is NR 34 R 35 . In another embodiment, R 3 is —C(O)R 31 . In another embodiment, R 3 is CO 2 R 31 . In another embodiment, R 3 is —C( ⁇ O)NR 31 R 32 . In another embodiment, R 3 is —C( ⁇ O)NR 34 R 35 .
  • R 3 is —NO 2 . In another embodiment, R 3 is —CN. In another embodiment, R 3 is —S(O) 0-2 R 3 . In another embodiment, R 3 is SO 2 NR 31 R 32 . In another embodiment, R 3 is —SO 2 NR 34 R 35 . In another embodiment, R 3 is —NR 31 C( ⁇ O)R 32 . In another embodiment, R 3 is —NR 31 C( ⁇ O)OR 32 . In another embodiment, R 3 is —NR 31 C( ⁇ O)NR 32 R 33 . In another embodiment, R 3 is NR 31 S(O) 0-2 R 32 . In another embodiment, R 3 is —C( ⁇ S)OR 31 .
  • R 3 is —C( ⁇ O)SR 31 . In another embodiment, R 3 is —NR 31 C( ⁇ NR 32 )NR 33 R 32 . In another embodiment, R 3 is —NR 31 C( ⁇ NR 32 )OR 33 . In another embodiment, R 3 is —NR 31 C( ⁇ NR 32 )SR 33 . In another embodiment, R 3 is —OC( ⁇ O)OR 33 . In another embodiment, R 3 is —OC( ⁇ O)NR 31 R 32 . In another embodiment, R 3 is —OC( ⁇ O)SR 31 . In another embodiment, R 3 is —SC( ⁇ O)OR 31 . In another embodiment, R 3 is —P(O)OR 31 OR 32 .
  • R 3 is —SC( ⁇ O)NR 31 R 32 .
  • R 3 is aryl.
  • R 2 is heteroaryl.
  • R 3 is C 1-4 alkyl.
  • R 3 is C 1-10 alkyl.
  • R 3 is C 3-8 cycloalkyl.
  • R 3 is C 3-8 cycloalkyl-C 1-10 alkyl.
  • R 3 is —C 1-10 alkyl-C 3-8 cycloalkyl.
  • R 3 is C 2-10 alkyl-monocyclic aryl.
  • R 3 is monocyclic aryl-C 2-10 alkyl.
  • R 3 is C 1-10 alkyl-bicyclicaryl. In another embodiment, R 3 is bicyclicaryl-C 1-10 alkyl. In another embodiment, R 3 is C 1-10 alkylheteroaryl. In another embodiment, R 3 is C 1-10 alkylheterocyclyl. In another embodiment, R 3 is C 2-10 alkenyl. In another embodiment, R 3 is C 2-10 alkynyl. In another embodiment, R 3 is C 2-10 alkenylaryl. In another embodiment, R 3 is C 2-10 alkenylheteroaryl. In another embodiment, R 3 is C 2-10 alkenylheteroalkyl. In another embodiment, R 3 is C 2-10 alkenylheterocycicyl.
  • R 3 is —C 2-10 alkynylaryl. In another embodiment, R 3 is —C 2-10 alkynylheteroaryl. In another embodiment, R 3 is —C 2-10 alkynylheteroalkyl. In another embodiment, R 3 is C 2-10 alkynylheterocyclyl. In another embodiment, R 3 is —C 2-10 alkynylC 3-8 cycloalkyl. In another embodiment, R 3 is C 2-10 alkynylC 3-8 cycloalkenyl. In another embodiment, R 3 is —C 1-10 alkoxy C 1-10 alkyl. In another embodiment, R 3 is C 1-10 alkoxy-C 2-10 alkenyl.
  • R 3 is —C 1-10 alkoxy-C 2-10 alkynyl. In another embodiment, R 3 is heterocyclyl-C 1-10 alkyl. In another embodiment, R 3 is -heterocyclylC 2-10 alkenyl. In another embodiment, R 3 is heterocyclyl-C 2-10 alkynyl. In another embodiment, R 3 is aryl-C 1-10 alkyl. In another embodiment, R 3 is aryl-C 2-10 alkenyl. In another embodiment, R 3 is aryl-C 2-10 alkynyl. In another embodiment, R 3 is aryl-heterocyclyl. In another embodiment, R 3 is heteroaryl-C 1-10 alkyl.
  • R 3 is heteroaryl-C 2-10 alkenyl. In another embodiment, R 3 is heteroaryl-C 2-10 alkynyl. In another embodiment, R 3 is heteroaryl-C 3-8 cycloalkyl. In another embodiment, R 3 is heteroaryl-heteroalkyl. In another embodiment, R 3 is heteroaryl-heterocyclyl.
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is unsubstituted. In another embodiment, when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent halo.
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —OH. In another embodiment, when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —R 31 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —CF 3 .
  • R 3 when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —OCF.
  • R 3 when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent OR 31 . In another embodiment, when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent NR 31 R 32 .
  • R 3 when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent NR 34 R 35 . In another embodiment, when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent C(O)R 31 .
  • R 3 when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl or heteroalkyl, it is substituted with one or more independent CO 2 R 31 . In another embodiment, when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent C( ⁇ O)NR 31 R 32 .
  • R 3 when R 3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent C( ⁇ O)NR 34 R 35 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —NO 2 .
  • R 3 when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent CN. In another embodiment, when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent S(O) 0-2 R 31 .
  • R 3 when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent SO 2 NR 31 R 32 . In another embodiment, when R 3 is aryl, heteroaryl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent SO 2 NR 34 R 35 .
  • R 3 when R 3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl. C 2-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent NR 31 C( ⁇ O)R 32 .
  • R 3 when R 3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ O)OR 32 .
  • R 3 when R 3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ O)NR 32 R 33 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 S(O) 0-2 R 32 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent C( ⁇ S)OR 31 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent C( ⁇ O)SR 31 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —NR 31 C( ⁇ NR 32 )NR 33 R 32 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent, —NR 31 C( ⁇ NR 32 )OR 33 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent NR 31 C( ⁇ NR 32 )SR 33 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)OR 33 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent OC( ⁇ O)NR 31 R 32 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)SR 31 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SC( ⁇ O)OR 31 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —P(O)OR 31 OR 32 .
  • R 3 when R 3 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SC( ⁇ O)NR 31 R 32 .
  • R 4 is hydrogen. In another embodiment, R 4 is halogen. In another embodiment, R 4 is OH. In another embodiment, R 4 is R 31 . In another embodiment, R 4 is CF 3 . In another embodiment, R 4 is OCF 3 . In another embodiment, R 4 is —OR 31 . In another embodiment, R 4 is NR 31 R 32 . In another embodiment, R 4 is NR 34 R 35 . In another embodiment, R 4 is —C(O)R 31 . In another embodiment, R 4 is —CO 2 R 31 . In another embodiment, R 4 is —C( ⁇ O)NR 31 R 32 . In another embodiment, R 4 is —C( ⁇ O)NR 34 R 35 .
  • R 4 is —NO 2 . In another embodiment, R 4 is —CN. In another embodiment, R 4 is —S(O) 0-2 R 3 . In another embodiment, R 4 is —SO 2 NR 31 R 32 . In another embodiment, R 4 is —SO 2 NR 34 R 35 . In another embodiment, R 4 is —NR 31 C( ⁇ O)R 32 . In another embodiment, R 4 is —NR 31 C( ⁇ O)OR 32 . In another embodiment, R 4 is —NR 31 C( ⁇ O)NR 32 R 33 . In another embodiment, R 4 is —NR 31 S(O) 0-2 R 32 . In another embodiment, R 4 is —C( ⁇ S)OR 31 .
  • R 4 is —C( ⁇ O)SR 31 . In another embodiment, R 4 is —NR 31 C( ⁇ NR 32 )NR 33 R 32 . In another embodiment, R 4 is —NR 31 C( ⁇ NR 32 )OR 33 . In another embodiment, R 4 is —NR 31 C( ⁇ NR 32 )SR 33 . In another embodiment, R 4 is —OC( ⁇ O)OR 33 . In another embodiment, R 4 is —OC( ⁇ O)NR 31 R 32 . In another embodiment, R 4 is —OC( ⁇ O)SR 31 . In another embodiment, R 4 is —SC( ⁇ O)OR 31 . In another embodiment, R 4 is —P(O)OR 31 OR 32 .
  • R 4 is —SC( ⁇ O)NR 31 R 32 .
  • R 4 is aryl.
  • R 4 is heteroaryl.
  • R 4 is C 1-4 alkyl.
  • R 4 is C 1-10 alkyl.
  • R 4 is C 3-8 cycloalkyl.
  • R 4 is C 1-10 alkyl-C 3-8 cycloalkyl.
  • R 4 is C 1-10 alkylaryl.
  • R 4 is C 1-10 alkylheteroaryl.
  • R 4 is C 1-10 alkylheterocyclyl.
  • R 4 is C 2-10 alkenyl.
  • R 4 is C 2-10 alkynyl. In another embodiment, R 4 is C 2-10 alkynyl-C 3-8 cycloalkyl. R 4 is C 2-10 alkenyl-C 3-8 cycloalkyl. In another embodiment, R 4 is C 2-10 alkenylaryl. In another embodiment, R 4 is C 2-10 alkenyl-heteroaryl. In another embodiment, R 4 is C 2-10 alkenylheteroalkyl. In another embodiment, R 4 is C 2-10 alkenylheterocycicyl. In another embodiment, R 4 is —C 2-10 alkynylaryl. In another embodiment, R 4 is C 2-10 alkynylheteroaryl.
  • R 4 is C 2-10 alkynylheteroalkyl. In another embodiment, R 4 is C 2-10 alkynylheterocyclyl. In another embodiment, R 4 is C 2-10 alkynylC 3-8 cycloalkyl. In another embodiment, R 4 is heterocyclyl C 1-10 alkyl. In another embodiment, R 4 is heterocyclylC 2-10 alkenyl. In another embodiment, R 4 is heterocyclyl-C 2-10 alkynyl. In another embodiment, R 4 is aryl-C 1-10 alkyl. In another embodiment, R 4 is aryl-C 2-10 alkenyl. In another embodiment, R 4 is aryl-C 2-10 alkynyl.
  • R 4 is aryl-heterocyclyl. In another embodiment, R 4 is heteroaryl-C 1-10 alkyl. In another embodiment, R 4 is heteroaryl-C 2-10 alkenyl. In another embodiment, R 4 is heteroaryl-C 2-10 alkynyl. In another embodiment, R 4 is C 3-8 cycloalkyl-C 1-10 alkyl. In another embodiment, R 4 is C 3-8 cycloalkyl-C 2-10 alkenyl. In another embodiment, R 4 is C 3-8 cycloalkyl-C 2-10 alkynyl.
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is unsubstituted. In another embodiment, when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent halo.
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —OH. In another embodiment, when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —R 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent CF 3 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —OCF.
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent OR 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent NR 31 R 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent NR 34 R 35 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —C(O)R 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent CO 2 R 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —C( ⁇ O)NR 31 R 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C( ⁇ O)NR 34 R 35 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —NO 2 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —CN.
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —S(O) 0-2 R 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —SO 2 NR 31 R 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent SO 2 NR 34 R 35 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent NR 31 C( ⁇ O)R 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent NR 31 C( ⁇ O)OR 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ O)NR 32 R 33 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 S(O) 0-2 R 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C( ⁇ S)OR 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —C( ⁇ O)SR 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —NR 31 C( ⁇ NR 32 )NR 33 R 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl C 2-10 alkyl heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent, —NR 31 C( ⁇ NR 32 )OR 33 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —NR 31 C( ⁇ NR 32 )SR 33 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)OR 33 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)NR 31 R 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —OC( ⁇ O)SR 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SC( ⁇ O)OR 31 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, C 3-8 cycloalkyl, C 3-8 cycloalkyl-C 1-10 alkyl, heterocyclyl, heterocyclyl C 1-10 alkyl, or heteroalkyl, it is substituted with one or more independent —P(O)OR 31 OR 32 .
  • R 4 when R 4 is aryl, heteroaryl, C 1-10 alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C 2-10 alkenyl, C 2-10 alkynyl, aryl-C 2-10 alkyl, heterocyclyl C 1-10 alkyl, or C 3-8 cycloalkyl-C 1-10 alkyl, it is substituted with one or more independent —SC( ⁇ O)NR 31 R 32 .
  • R 5 is hydrogen. In another embodiment, R 5 is halogen. In another embodiment, R 5 is —OH. In another embodiment, R 5 is —R 31 . In another embodiment, R 5 is —CF 3 . In another embodiment, R 5 is —OCF 3 . In another embodiment, R 5 is —OR 31 . In another embodiment, R 5 is —NR 31 R 32 . In another embodiment, R 5 is —NR 34 R 35 . In another embodiment, R 5 is —C(O)R 31 . In another embodiment, R 5 is —CO 2 R 31 . In another embodiment, R 5 is —C( ⁇ O)NR 31 R 32 . In another embodiment, R 5 is —C( ⁇ O)NR 34 R 35 .
  • R 5 is —NO 2 . In another embodiment, R 5 is —CN. In another embodiment, R 5 is —S(O) 0-2 R 31 . In another embodiment, R 5 is —SO 2 NR 31 R 32 . In another embodiment, R 5 is —SO 2 NR 34 R 35 . In another embodiment, R 5 is —NR 31 C( ⁇ O)R 32 . In another embodiment, R 5 is —NR 31 C( ⁇ O)OR 32 . In another embodiment, R 5 is —NR 31 C( ⁇ O)NR 32 R 33 . In another embodiment, R 5 is —NR 31 S(O) 0-2 R 32 . In another embodiment, R 5 is —C( ⁇ S)OR 31 .
  • R 5 is —C( ⁇ O)SR 31 .
  • R 5 is —NR 31 C( ⁇ NR 32 )NR 33 R 32 .
  • R 5 is —NR 31 C( ⁇ NR 32 )OR 33 .
  • R 5 is —NR 31 C( ⁇ NR 32 )SR 33 .
  • R 5 is —OC( ⁇ O)OR 33 .
  • R 5 is —OC( ⁇ O)NR 31 R 32 .
  • R 5 is —OC( ⁇ O)SR 31 .
  • R 5 is —SC( ⁇ O)OR 31 .
  • R 5 is —P(O)OR 31 OR 32 .
  • R 5 is or —SC( ⁇ O)NR 31 R 32 .
  • R 7 is hydrogen. In another embodiment, R 7 is unsubstituted C 1-10 alkyl. In another embodiment, R 7 is unsubstituted C 2-10 alkenyl. In another embodiment, R 7 is unsubstituted aryl. In another embodiment, R 7 is unsubstituted heteroaryl. In another embodiment, R 7 is unsubstituted heterocyclyl. In another embodiment, R 7 is unsubstituted C 3-10 cycloalkyl. In another embodiment, R 7 is C 1-10 alkyl substituted by one or more independent R 6 . In another embodiment, R 7 is C 2-10 alkenyl substituted by one or more independent R 6 .
  • R 7 is aryl substituted by one or more independent R 6 . In another embodiment, R 7 is heteroaryl substituted by one or more independent R 6 . In another embodiment, R 7 is heterocycly substituted by one or more independent R 6 . In another embodiment, R 7 is C 3-10 cycloalkyl substituted by one or more independent R 6 .
  • R 8 is hydrogen. In another embodiment, R 8 is unsubstituted C 1-10 alkyl. In another embodiment, R 8 is unsubstituted C 2-10 alkenyl. In another embodiment, R 8 is unsubstituted aryl. In another embodiment, R 8 is unsubstituted heteroaryl. In another embodiment, R 8 is unsubstituted heterocyclyl. In another embodiment, R 8 is unsubstituted C 3-10 cycloalkyl. In another embodiment, R 8 is C 1-10 alkyl substituted by one or more independent R 6 . In another embodiment, R 8 is C 2-10 alkenyl substituted by one or more independent R 6 .
  • R 8 is aryl substituted by one or more independent R 6 . In another embodiment, R 8 is heteroaryl substituted by one or more independent R 6 . In another embodiment, R 8 is heterocyclyl substituted by one or more independent R 6 . In another embodiment, leis C 3-10 cycloalkyl substituted by one or more independent R 6 .
  • R 6 is halo, In another embodiment, R 6 is —OR 31 . In another embodiment, R 6 is —SH. In another embodiment, R 6 is NH 2 . In another embodiment, R 6 is —NR 34 R 35 . In another embodiment, R 6 is NR 31 R 32 . In another embodiment, R 6 is —CO 2 R 31 . In another embodiment, R 6 is CO 2 aryl. In another embodiment, R 6 is —C( ⁇ O)NR 31 R 32 . In another embodiment, R 6 is C( ⁇ O) NR 34 R 35 . In another embodiment, R 6 is NO 2 . In another embodiment, R 6 is —CN.
  • R 6 is —S(O) 0-2 C 1-10 alkyl. In another embodiment, R 6 is —S(O) 0-2 aryl. In another embodiment, R 6 is —SO 2 NR 34 R 35 . In another embodiment, R 6 is —SO 2 NR 31 R 32 . In another embodiment, R 6 is C 1-10 alkyl. In another embodiment, R 6 is C 2-10 alkenyl. In another embodiment, R 6 is C 2-10 alkynyl. In another embodiment, R 6 is unsubstituted aryl-C 1-10 alkyl. In another embodiment, R 6 is unsubstituted aryl-C 2-10 alkenyl.
  • R 6 is unsubstituted aryl-C 2-10 alkynyl. In another embodiment, R 6 is unsubstituted heteroaryl-C 1-10 alkyl. In another embodiment, R 6 is unsubstituted heteroaryl-C 2-10 alkenyl. In another embodiment, R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent halo.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent cyano.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent nitro.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —OC 1-10 alkyl.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C 1-10 alkyl.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C 2-10 alkenyl.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C 2-10 alkynyl.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent (halo)C 1-10 alkyl.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent (halo)C 2-10 alkenyl.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent (halo)C 2-10 alkynyl.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —COOH.
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C( ⁇ O)NR 31 R 32 .
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C( ⁇ O) NR 34 R 35 .
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent SO 2 NR 34 R 35 .
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —SO 2 NR 31 R 32 .
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —NR 31 R 32 .
  • R 6 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —NR 34 R 35 .
  • R 9 is H. In another embodiment, R 9 is halo. In another embodiment, R 9 is —OR 31 . In another embodiment, R 9 is —SH. In another embodiment, R 9 is NH 2 . In another embodiment, R 9 is —NR 34 R 35 . In another embodiment, R 9 is —NR 31 R 32 . In another embodiment, R 9 is —CO 2 R 31 . In another embodiment, R 9 is CO 2 aryl. In another embodiment, R 9 is —C( ⁇ O)NR 31 R 32 . In another embodiment, R 9 is C( ⁇ O) NR 34 R 35 . In another embodiment, R 9 is NO 2 . In another embodiment, R 9 is CN.
  • R 9 is —S(O) 0-2 C 1-10 alkyl. In another embodiment, R 9 is —S(O) 0-2 aryl. In another embodiment, R 9 is SO 2 NR 34 R 35 . In another embodiment, R 9 is —SO 2 NR 31 R 32 . In another embodiment, R 9 is C 1-10 alkyl. In another embodiment, R 9 is C 2-10 alkenyl. In another embodiment, R 9 is C 2-10 alkynyl. In another embodiment, R 9 is unsubstituted aryl-C 1-10 alkyl. In another embodiment, R 9 is unsubstituted aryl-C 2-10 alkenyl.
  • R 9 is unsubstituted aryl-C 2-10 alkynyl. In another embodiment, R 9 is unsubstituted heteroaryl-C 1-10 alkyl. In another embodiment, R 9 is unsubstituted heteroaryl-C 2-10 alkenyl. In another embodiment, R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent halo.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent cyano.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent nitro.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —OC 1-10 alkyl.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C 1-10 alkyl.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C 2-10 alkenyl.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C 2-10 alkynyl.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent -(halo)C 1-10 alkyl.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent -(halo)C 2-10 alkenyl.
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C( ⁇ O)NR 31 R 32 .
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —C( ⁇ O) NR 34 R 35 .
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —SO 2 NR 34 R 35 .
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —SO 2 NR 31 R 32 .
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —NR 31 R 32 .
  • R 9 is aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, or heteroaryl-C 2-10 alkenyl substituted by one or more independent —NR 34 R 35 .
  • R 31 is H. In some embodiments, R 31 is unsubstituted C 1-10 alkyl. In some embodiments, R 31 is substituted C 1-10 alkyl. In some embodiments, R 31 is C 1-10 alkyl substituted with one or more aryl. In some embodiments, R 31 is C 1-10 alkyl substituted with one or more heteroalkyl. In some embodiments, R 31 is C 1-10 alkyl substituted with one or more heterocyclyl. In some embodiments, R 31 is C 1-10 alkyl substituted with one or more heteroaryl.
  • each of said aryl substituents is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(—C( ⁇ O)NH(—C( ⁇ O)NR 34 R 35 , —C( ⁇ O)NH 2 , —OCF 3 , —O-aryl, —N(
  • each of said heterocyclyl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2-10 C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C
  • each of said heteroaryl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 —C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C 1
  • R 32 is H. In some embodiments, R 32 is unsubstituted C 1-10 alkyl. In some embodiments, R 32 is substituted C 1-10 alkyl. In some embodiments, R 32 is C 1-10 alkyl substituted with one or more aryl. In some embodiments, R 32 is C 1-10 alkyl substituted with one or more heteroalkyl. In some embodiments, R 32 is C 1-10 alkyl substituted with one or more heterocyclyl. In some embodiments, R 32 is C 1-10 alkyl substituted with one or more heteroaryl.
  • each of said aryl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 —C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C 1
  • each of said heteroalkyl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 —C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C 1-10 alkyl), —C( ⁇ O)NR 34 R 35
  • each of said heterocyclyl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 —C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(
  • each of said heteroaryl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2-10 C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C 1-10
  • R 33 is unsubstituted C 1-10 alkyl. In some embodiments, R 33 is substituted C 1-10 alkyl. In some embodiments, R 33 is C 1-10 alkyl substituted with one or more aryl. In some embodiments, R 33 is C 1-10 alkyl substituted with one or more heteroalkyl. In some embodiments, R 33 is C 1-10 alkyl substituted with one or more heterocyclyl. In some embodiments, R 33 is C 1-10 alkyl substituted with one or more heteroaryl.
  • each of said aryl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 —C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C 1
  • each of said heteroalkyl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 —C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C 1-10 alkyl), —C( ⁇ O)NR 34 R 35
  • each of said heterocyclyl group is unsubstituted or substituted with one or more halo, —OH, —C 10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2 —C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C
  • each of said heteroaryl group is unsubstituted or substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —C 1-10 alkyl, —CO 2-10 C 1-10 alkylaryl, —CO 2 -aryl, —C( ⁇ O)N(C 1-10 alkyl)(C 1-10 alkyl), —C( ⁇ O)NH(C 1-10
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, heteroaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen.
  • X 1 is C ⁇ NH 2 .
  • X 1 is C ⁇ NH—R 4 , where —NH—R 4 is:
  • the invention provides an inhibitor of Formula I-C1 where R 5 is H. In another embodiment, the invention provides an inhibitor of Formula I-C2 where R 5 is H.
  • R 3 is hydrogen, —OH, —OR 31 , —NR 31 R 32 , —C(O)R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , aryl, heteroaryl, C 1-4 alkyl, C 1-10 alkyl, C 3-8 cycloalkyl, or heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C(
  • an inhibitor of Formula I-C1 is a compound of Formula I-C1a:
  • R 1 is -L-C 1-10 alkyl, -L-C 3-8 cycloalkyl, -L-C 1-10 alkylheterocyclyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R 3 ;
  • L is absent, —(C ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O) N(R 31 )—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 N(R 31 )—, or —N(R 31 )—;
  • R 3 is hydrogen, —OH, —OR 31 , —NR 31 R 32 , —C(O)R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , aryl, heteroaryl, C 1-4 alkyl, C 1-10 alkyl, C 3-8 cycloalkyl, or heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —NR 31
  • —(W 2 ) k — is —NH—, —N(H)C(O)— or —N(H)S(O) 2 —;
  • R 31 , R 32 , and R 33 are independently H or C 1-10 alkyl, wherein the C 1-10 alkyl is unsubstituted;
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, heteroaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen.
  • the invention further provides a compound which is an mTor inhibitor, wherein the compound has the Formula I-A:
  • X 1 is N or C-E 1 , X 2 is N, X 3 is C, and X 4 is C—R 9 or N; or X 1 is N or C-E 1 , X 2 is C, X 3 is N, and X 4 is C—R 9 or N;
  • R 1 is —H, -L-C 1-10 alkyl, -L-C 3-8 cycloalkyl, -L-C 1-10 alkyl-C 3-8 cycloalkyl, -L-aryl, -L-heteroaryl, -L-C 1-10 alkylaryl, -L-C 1-10 alkylheteroaryl, -L-C 1-10 alkylheterocyclyl, -L-C 2-10 alkenyl, -L-C 2-10 alkynyl, -L-C 2-10 alkenyl-C 3-8 cycloalkyl, -L-C 2-10 alkynyl-C 3-8 cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C
  • L is absent, —(C ⁇ O)—, —C( ⁇ O)O—, —C( ⁇ O) N(R 31 )—, —S—, —S(O)—, —S(O) 2 —, —S(O) 2 N(R 31 )—, or —N(R 31 )—;
  • M 1 is benzothiazolyl substituted with —(W 2 ) k —R 2 ;
  • k 0 or 1
  • E 1 and E 2 are independently —(W 1 ) j —R 4 ;
  • j in each instance (i.e., in E 1 or j in E 2 ), is independently 0 or 1
  • W 1 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(R 7 )N(R 8 )S(O) 2 —;
  • W 2 is —O—, —S(O) 0-2 -, —C(O)—, —C(O)N(R 7 )—, —N(R 7 )C(O)—, —N(R 7 )C(O)N(R 8 )—, —N(R 7 )S(O)—, —N(R 7 )S(O) 2 —, —C(O)O—, —CH(R 7 )N(C(O)OR 8 )—, —CH(R 7 )N(C(O)R 8 )—, —CH(R 7 )N(SO 2 R 8 )—, —CH(R 7 )N(R 8 )—, —CH(R 7 )C(O)N(R 8 )—, —CH(R 7 )N(R 8 )C(O)—, —CH(R 7 )N(R 8 )S(O)—, or —CH(
  • R 2 is hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR 33 ,
  • R 3 and R 4 are independently hydrogen, halogen, —OH, —R 31 , —CF 3 , —OCF 3 , —OR 31 , —NR 31 R 32 , —NR 34 R 35 , —C(O)R 31 , —CO 2 R 31 , —C( ⁇ O)NR 31 R 32 , —C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 R 31 , —SO 2 NR 31 R 32 , —SO 2 NR 34 R 35 , —NR 31 C( ⁇ O)R 32 , —NR 31 C( ⁇ O)OR 32 , —NR 31 C( ⁇ O)NR 32 R 33 , —NR 31 S(O) 0-2 R 32 , —C( ⁇ S)OR 31 , —C( ⁇ O)SR 31 , —NR 31 C( ⁇ NR 32 )NR 33 R 32 , —NR 31 C( ⁇ NR 32 )OR
  • R 31 , R 32 , and R 33 are independently H or C 1-10 alkyl, wherein the C 1-10 alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C 1-10 alkyl, —CF 3 , —O-aryl, —OCF 3 , —OC 1-10 alkyl, —NH 2 , —N(C 1-10 alkyl)(C 1-10 alkyl), —NH(C 1-10 alkyl), —NH(aryl), —NR 34 R 35 , —C(O)(C 1-10 alkyl), —C(O)(C 1-10 alkyl-aryl), —C(O)(aryl), —CO 2 —
  • R 34 and R 35 in —NR 34 R 35 , —C( ⁇ O)NR 34 R 35 , or —SO 2 NR 34 R 35 are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR 31 R 32 , hydroxyl, halogen, oxo, aryl, heteroaryl, C 1-6 alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • R 7′ and R 8′ are each independently hydrogen, C 1-10 alkyl, C 2-10 alkenyl, aryl, heteroaryl, heterocyclyl or C 3-10 cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R 6 ;
  • R 6 is halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, heteroaryl-C 2-10 alkenyl, heteroaryl-C 2-10 alkynyl, wherein each of said alkyl, alkenyl, alkyny
  • R 9 is H, halo, —OR 31 , —SH, —NH 2 , —NR 34 R 35 , —NR 31 R 32 , —CO 2 R 31 , —CO 2 aryl, —C( ⁇ O)NR 31 R 32 , C( ⁇ O)NR 34 R 35 , —NO 2 , —CN, —S(O) 0-2 C 1-10 alkyl, —S(O) 0-2 aryl, —SO 2 NR 34 R 35 , —SO 2 NR 31 R 32 , C 1-10 alkyl, C 2-10 alkenyl, C 2-10 alkynyl; aryl-C 1-10 alkyl, aryl-C 2-10 alkenyl, aryl-C 2-10 alkynyl, heteroaryl-C 1-10 alkyl, heteroaryl-C 2-10 alkenyl, heteroaryl-C 2-10 alkynyl, wherein each of said alkyl, alkenyl, alky
  • X 4 is C—R 9 .
  • the invention also provides an inhibitor as defined above, wherein the compound is of Formula I-B:
  • the compound of Formula I-B or its pharmaceutically acceptable salt thereof is an inhibitor having the structure of Formula I-B1 or Formula I-B2:
  • X 1 is N and X 2 is N. In other embodiments, X 1 is C-E 1 and X 2 is N. In yet other embodiments, X 1 is NH and X 2 is C. In further embodiments, X 1 is CH-E 1 and X 2 is C.
  • X 1 is N and X 2 is C. In further embodiments, X 1 is C-E 1 and X 2 is C.
  • X 1 is C—(W 1 ) j —R 4 , where j is 0.
  • X 1 is CH. In yet another embodiment, X 1 is C-halogen, where halogen is Cl, F, Br, or I.
  • X 1 it is C—(W 1 ) j —R 4 .
  • W 1 is —O—.
  • W 1 is NR′—.
  • W 1 is —NH—.
  • W 1 is —S(O) 0-2 .
  • W 1 is —C(O)—.
  • W 1 is —C(O)N(R 7 ).
  • X 1 , j is 1, and W 1 is —N(R 7 )C(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —N(R 7 )S(O) 2 —. In various embodiments of X 1 , j is 1, and W 1 is —C(O)O—. In various embodiments of X 1 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 ).
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )C(O)—.
  • X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of X 1 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • X 1 is CH 2 . In yet another embodiment, X 1 is CH-halogen, where halogen is Cl, F, Br, or I.
  • X 1 is N.
  • E 2 is —(W 1 ) j —R 4 , where j is 0.
  • E 2 is CH. In yet another embodiment, E 2 is C-halogen, where halogen is Cl, F, Br, or I.
  • E 2 it is —(W 1 ), —R 4 .
  • j is 1, and W 1 is —O—.
  • W 1 is —NR′—.
  • W 1 is —NH—.
  • W 1 is —S(O) 0-2 -.
  • W 1 is —C(O)—.
  • j is 1, and W 1 is —C(O)N(R 7 )—.
  • E 2 j is 1, and W 1 is —N(R 7 )C(O)—. In various embodiments of E 2 , j is 1, and W 1 is —N(R 7 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —N(R 7 )S(O) 2 —. In various embodiments of E 2 , j is 1, and W 1 is —C(O)O—. In various embodiments of E 2 , j is 1, and W 1 is CH(R 7 )N(C(O)OR 8 )—.
  • E 2 j is 1, and W 1 is —CH(R 7 )N(C(O)R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(SO 2 R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )C(O)N(R 8 )—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )C(O)—.
  • E 2 j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O)—. In various embodiments of E 2 , j is 1, and W 1 is —CH(R 7 )N(R 8 )S(O) 2 —.
  • M 1 is:
  • M 1 is benzothiazolyl substituted with —(W 2 ) k —R 2 .
  • W 2 can be —O—, —S(O) 0-2 -(including but not limited to —S—, —S(O)—, and —S(O) 2 —), —C(O)—, or —C(O)O—.
  • W 1 is —NR 6 — or —CH(R 6 )N(R 7 )—, wherein R 6 and R 7 are each independently hydrogen, unsubstituted or substituted C 1 -C 10 alkyl (which includes but is not limited to —CH 3 , —CH 2 CH 3 , n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), unsubstituted or substituted C 2 -C 10 alkenyl (including but not limited to alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl).
  • C 1 -C 10 alkyl which includes but is not limited to —CH 3 , —CH 2 CH 3 , n-propyl, isopropyl, n-butyl, tert-butyl, sec-but
  • R 6 and R 7 are each independently unsubstituted or substituted aryl (including phenyl and naphthtyl).
  • R 6 and R 7 are each independently heteroaryl, wherein the heteroaryl is unsubstituted or substituted.
  • R 6 and R 7 heteroaryl is monocyclic heteroaryl, and includes but is not limited to imidazolyl, pyrrolyl, oxazolyl, thiazolyl, and pyridinyl.
  • R 6 and R 7 are each independently unsubstituted or substituted heterocyclyl (which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl) or unsubstituted or substituted C 3-8 cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl).
  • Non exemplary W 2 include NH—, —N(cyclopropyl), and N(4-N-piperidinyl).
  • exemplary mTor inhibitors of the invention have the Formulas:
  • mTor inhibitor compounds disclosed herein may be prepared by the routes described below. Materials used herein are either commercially available or prepared by synthetic methods generally known in the art. These schemes are not limited to the compounds listed or by any particular substituents employed for illustrative purposes. Numbering does not necessarily correspond to that of claims or other tables.
  • compounds are synthesized by condensing a functionalized heterocycle A-1 with formamide, to provide a pyrazolopyrimidine A-2.
  • the pyrazolopyrimidine is treated with N-iodosuccinimide, which introduces an iodo substituent in the pyrazole ring as in A-3.
  • the R 1 substituent is introduced by reacting the pyrazolopyrimidine A3 with a compound of Formula R 1 -Lg in the presence of a base such as potassium carbonate to produce a compound of Formula A-4.
  • bases that are suitable for use in this step include but are not limited to sodium hydride and potassium t-butoxide.
  • the compound of Formula R 1 -Lg has a moiety R 1 as defined for R 1 of a compound of Formula I-A, and wherein Lg is an appropriate leaving group such as halide (including bromo, iodo, and chloro), tosylate, or other suitable leaving group,
  • Mitsunobu chemistry can be used to obtain alkylated pyrazolopyrimidine A-4, as shown in Scheme A-1.
  • Iodopyrazolopyrimidine A-3 is reacted with a suitable alcohol, in the presence of triphenylphosphine and diisopropylazodicarboxylate (DIAD) to produce pyrazolopyrimidine A-4.
  • DIAD diisopropylazodicarboxylate
  • the compounds of the invention may be synthesized via a reaction scheme represented generally in Scheme B.
  • the synthesis proceeds via coupling a compound of Formula A with a compound of Formula B to yield a compound of Formula C.
  • the coupling step is typically catalyzed by using, e.g., a palladium catalyst, including but not limited to palladium tetrakis (triphenylphosphine).
  • the coupling is generally performed in the presence of a suitable base, a nonlimiting example being sodium carbonate.
  • a suitable solvent for the reaction is aqueous dioxane.
  • a compound of Formula A for use in Scheme B has a structure of Formula A, wherein T 1 is triflate or halo (including bromo, chloro, and iodo), and wherein R 1 , X 1 , X 2 , X 3 , R 31 and R 32 are defined as for a compound of Formula I-A.
  • M is either M 1 or M 2 .
  • M 1 is defined as for a compound of Formula I-A.
  • M 1 can be a 5-benzoxazolyl or a 6-benzoxazolyl moiety, including but not limited to those M 1 moieties disclosed herein.
  • M 2 is a moiety which is synthetically transformed to form M 1 , after the M 2 moiety has been coupled to the bicyclic core of the compound of Formula A.
  • G is hydrogen or R G1 , wherein R G1 is alkyl, alkenyl, or aryl.
  • B(OG) 2 is taken together to form a 5- or 6-membered cyclic moiety.
  • the compound of Formula B is a compound having a structure of Formula E:
  • G is H or R G1 ;
  • R G1 is alkyl, alkenyl, or aryl.
  • R 2 is a R G2 moiety, wherein the R G2 moiety is H, acyl, or an amino protecting group including but not limited to tert-butyl carbamate (Boc), carbobenzyloxy (Cbz), benzyl (Bz), fluorenylmethyloxycarbonyl (FMOC), p-methoxybenzyl (PMB), and the like.
  • R G2 moiety is H, acyl, or an amino protecting group including but not limited to tert-butyl carbamate (Boc), carbobenzyloxy (Cbz), benzyl (Bz), fluorenylmethyloxycarbonyl (FMOC), p-methoxybenzyl (PMB), and the like.
  • a compound of Formula B is a compound of Formula B′, wherein G is R G1 . or a compound of Formula B′′, wherein G is hydrogen.
  • Scheme C depicts an exemplary scheme for synthesizing a compound of Formula B′ or, optionally, Formula B′′ for use in Reaction Scheme C.
  • This reaction proceeds via reacting a compound of Formula D with a trialkyl borate or a boronic acid derivative to produce a compound of Formula B′.
  • the reaction is typically run a solvent such as dioxane or tetrahydrofuran.
  • the trialkyl borate includes but is not limited to triisopropyl borate and the boronic acid derivative includes but is not limited to bis(pinacolato)diboron.
  • a base such as n-butyllithium is first added to the compound of Formula D to generate an anion, prior to the addition of the borate.
  • a boronic acid derivative such as bis(pinacolato)diboron
  • a palladium catalyst and a base is used.
  • Typical palladium catalysts include but is not limited to palladium chloride (diphenylphosphino)ferrocene).
  • a suitable base includes but is not limited to potassium acetate.
  • a compound of Formula D for use in Scheme C is a compound wherein T 2 is halo or another leaving group, and M is as defined above in Scheme B.
  • the compound of Formula B′ may further be converted to a compound of Formula B′′ by treatment with an acid such as hydrochloric acid.
  • the G groups are hydrogen. In another of a compound of Formula B, B′, B′′, or E, the G groups are R G1 .
  • no further synthetic transformation of M 1 moiety is performed after the coupling reaction when, e.g. M 1 is 2-N-acetyl-benzoxazol-5-yl.
  • a compound of Formula E is synthesized from a compound of Formula F, as shown in Scheme C-1:
  • Scheme C-1 depicts an exemplary scheme for synthesizing a compound of Formula E. This reaction proceeds via reacting a compound of Formula F with a trialkyl borate or a boronic acid derivative to produce a compound of Formula E. The conditions of the reaction are as described above in Scheme C.
  • a compound of Formula F for use in Scheme C-1 is a compound wherein T 2 is halo (including Br, Cl, and I) or another leaving group (including but not limited to triflate, tosylate, and mesylate), and the G p moiety is H, acyl, or an amino protecting group including but not limited to tert-butyl carbamate (Boc), carbobenzyloxy (Cbz), benzyl (Bz), fluorenylmethyloxycarbonyl (FMOC), p-methoxybenzyl (PMB), and the like.
  • T 2 is halo (including Br, Cl, and I) or another leaving group (including but not limited to triflate, tosylate, and mesylate)
  • the G p moiety is H, acyl, or an amino protecting group including but not limited to tert-butyl carbamate (Boc), carbobenzyloxy (Cbz), benzyl (Bz), fluorenylmethyloxy
  • the compound of Formula E, wherein G is alkyl may further be converted to a compound of Formula E, wherein G is hydrogen, by treatment with an acid such as hydrochloric acid
  • deprotection of a substituent e.g., removal of Boc protection from an amino substituent
  • M 1 of Formula C benzoxazolyl moiety
  • Some exemplary compounds with such protecting groups include but are not limited to compounds of the following formulae:
  • Step 1 a compound of Formula 3-1 is reacted with boronic acid 3-2, in the presence of palladium tetrakis (triphenylphosphine) and a suitable base, such as sodium carbonate in an aqueous/organic solvent mixture to produce a compound of Formula 3-3.
  • Step 2 the compound of Formula 3-3 is reacted with about 2 equivalents of nitric acid in acetic acid as solvent to produce a compound of Formula 3-4.
  • Two alternative transformations may be used to effect the next transformation of Step 3.
  • the compound of Formula 3-4 is treated with sodium dithionite and sodium hydroxide in water to produce a compound of Formula 3-5.
  • the compound of Formula 3-4 is reduced using palladium on carbon in a suitable solvent under a hydrogen atmosphere to yield a compound of Formula 3-5.
  • Step 4 compound 3-5 is reacted with about 1.2 equivalents of cyanogen bromide in a solvent such as methanol/tetrahydrofuran mixture to produce a compound of Formula 3-6.
  • a solvent such as methanol/tetrahydrofuran mixture
  • the compound of Formula 3-6 may be further transformed by other substitution or derivatization.
  • a compound of Formula 3-1 useful in the method of Scheme D is a compound having a structure of Formula 3-1, wherein T 1 is triflate or halo (including bromo, chloro, and iodo), and wherein R 1 , X 1 , X 2 , X 3 , R 31 and R 32 are defined as for a compound of Formula I-A.
  • Exemplary compounds having a pyrazolopyrimidine core can be synthesized via Scheme E.
  • Step 1 of Scheme E compound A-2 in dimethylformamide (DMF), is reacted with an N-halosuccinimide (NT 1 S) at about 80° C., to provide compound 4-1, where T 1 is iodo or bromo.
  • NT 1 S N-halosuccinimide
  • Step 2 compound 4-1 in DMF is reacted with a compound R 1 T x , in the presence of potassium carbonate, to provide compound 4-2.
  • compound 4-2 is coupled with a compound of Formula B using palladium catalysis such as palladium tetrakis (triphenylphosphine), and in the presence of sodium carbonate, to yield a pyrazolopyrimidine compound as shown.
  • a compound of Formula R 1 T x suitable for use in Reaction Scheme E is the compound wherein R 1 is cycloalkyl or alkyl and T x is halo (including bromo, iodo, or chloro) or a leaving group, including but not limited to mesylate or tosylate.
  • Reaction Schemes F-M illustrate methods of synthesis of borane reagents useful in preparing intermediates of use in synthesis of the compounds of the invention as described in Reaction Schemes A, B, and E above, to introduce M 1 substituents.
  • a compound of Formula N-1 and a compound of N-2 are coupled to produce a compound of Formula C.
  • the coupling step is typically catalyzed by using, e.g., a palladium catalyst, including but not limited to palladium tetrakis (triphenylphosphine).
  • the coupling is generally performed in the presence of a suitable base, a nonlimiting example being sodium carbonate.
  • a suitable base a nonlimiting example being sodium carbonate.
  • a suitable solvent for the reaction is aqueous dioxane.
  • a compound of Formula N-1 for use in Scheme N has a structure of Formula N-1, wherein G is hydrogen or R G1 , wherein R G1 is alkyl, alkenyl, or aryl.
  • B(OG) 2 of the compound of Formula N-1 is taken together to form a 5- or 6-membered cyclic moiety.
  • R 1 , X 1 , X 2 , X 3 , R 31 and R 32 of the compound of Formula N-1 are defined as for a compound of Formula I-A.
  • a compound of Formula N-2 for use in Scheme N has a structure of Formula N-2 wherein T 1 is triflate or halo (including bromo, chloro, and iodo).
  • M of the compound of Formula N-2 is either M 1 or M 2 .
  • M 1 is defined as for a compound of Formula I
  • M 1 can be a 5-benzoxazolyl or a 6-benzoxazolyl moiety, including but not limited to those M 1 moieties disclosed herein.
  • M 2 is a moiety which is synthetically transformed to form M 1 , after the M 2 moiety has been coupled to the bicyclic core of the compound of Formula N-1.
  • a compound of Formula N-1 may be synthesized as shown in Scheme N-1.
  • a compound of Formula N-1 is reacted with a trialkyl borate or a boronic acid derivative to produce a compound of Formula N-1.
  • the reaction is typically run a solvent such as dioxane or tetrahydrofuran.
  • the trialkyl borate includes but is not limited to triisopropyl borate and the boronic acid derivative includes but is not limited to bis(pinacolato)diboron.
  • a base such as n-butyllithium is first added to the compound of Formula N-3 to generate an anion, prior to the addition of the borate.
  • a boronic acid derivative such as bis(pinacolato)diboron
  • a palladium catalyst and a base is used.
  • Typical palladium catalysts include but is not limited to palladium chloride (diphenylphosphino)ferrocene).
  • a suitable base includes but is not limited to potassium acetate.
  • a compound of Formula N-3 suitable for use in Scheme N-1 is a compound wherein T 2 is halo or another leaving group such as mesylate, tosylate, or triflate.
  • X 1 , X 2 , X 3 , R 1 , R 31 , and R 32 of the compound of Formula N-3 is as defined for a compound of Formula I-A.
  • a compound of Formula A, B, B′, B′′, C, C′′, D, E, E′′, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1′′, N-3′′, 3-1′′, 3-3′′, 3-4′′, 3-5′′, 3-6′′, N-1′′, or N-3′′ is provided as its salt, including but not limited to hydrochloride, acetate, formate, nitrate, sulfate, and boronate.
  • a palladium compound including but not limited to palladium chloride (diphenylphosphino)ferrocene) and palladium tetrakis (triphenylphosphine), is used in the synthesis of a compound of Formula A, B, B′, B′′, C, C′′, D, E, E′′, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1′′, N-3′′, 3-1′′, 3-3′′, 3-4′′, 3-5′′, 3-6′′, N-1′′, or N-3′′.
  • palladium chloride diphenylphosphino
  • triphenylphosphine palladium tetrakis
  • a palladium compound When a palladium compound is present in the synthesis of a compound of Formula A, B, B′, B′′, C, C′′, D, E, E′′, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1′′, N-3′′, 3-1′′, 3-3′′, 3-4′′, 3-5′′, 3-6′′, N-1′′, or N-3′′, it is present in an amount ranging from about 0.005 molar equivalents to about 0.5 molar equivalents, from about 0.05 molar equivalents to about 0.20 molar equivalents, from about 0.05 molar equivalents to about 0.25 molar equivalents, from about 0.07 molar equivalents to about 0.15 molar equivalents, or about 0.8 molar equivalents to about 0.1 molar equivalents of the compound of Formula A, B, B′, B′′, C, D, E, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1, or N-3.
  • a a palladium compound including but not limited to palladium chloride (diphenylphosphino)ferrocene) and palladium tetrakis (triphenylphosphine) is present in the synthesis of a compound of Formula A, B, B′, B′′, C, C′′, D, E, E′′, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1′′, N-3′′, 3-1′′, 3-3′′, 3-4′′, 3-5′′, 3-6′′, N-1′′, or N-3′′ in about 0.07, about 0.08, about 0.09, about 0.10, about 0.11, about 0.12, about 0.13, about 0.14, or about 0.15 molar equivalents of a starting material of Formula A, B, B′, B′′, C, C′′, D, E, E′′, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1′′, N-3′′, 3-1′′, 3-3′′, 3-4′′
  • the R G2 moiety is removed, using suitable methods, at any point desired, whereupon the compound of Formula C, 3-1, 3-3, 3-4, 3-5, 3-6, A-2, 4-1, 4-2, N-1 or N-3 has a R 31 hydrogen replacing the R G2 moiety on the amino moiety.
  • This transformation is specifically illustrated for the conversion of a compound of Formula C′′ to a compound of C (i.e., as in Step 4 of Scheme E′) and for the conversion of a compound of Formula 3-6′′ to a compound of Formula 3-6 (ie., as in Step 5 of Scheme D′).
  • This illustration is in no way limiting as to the choice of steps wherein a compound comprising a NR 31 R G2 moiety may be converted to a compound comprising a NR 31 R 32 moiety wherein the R 32 moiety is hydrogen.
  • the invention encompasses methods of synthesis of the compounds of A, B, B′, B′′, C, E, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1 or N-3, wherein one or more of M, M 1 , or R 1 has a protecting group present during one or more steps of the synthesis.
  • Protecting groups suitable for use for a M, M 1 , or R 1 moiety are well known in the art, as well as the methods of incorporation and removal, and the reagents suitable for such transformations.
  • Reaction Schemes 0, P and Q illustrate methods of synthesis of borane reagents useful in preparing intermediates of use in synthesis of the compounds of the invention as described in Reaction Schemes 1 and 2 above, to introduce benzothiazolyl substituents.
  • a compound of Formula O-1 is treated with, for example, nitric acid to produce a compound of Formula O-2.
  • the compound of Formula O-2 is treated with a reducing agent such as stannous chloride to produce a compound of Formula O-3.
  • the compound of 0-3 is treated with sodium nitrate in acide and cupric bromide to produce a compound of Formula O-4.
  • the compound of O-4 is treated a base such as butyl lithium and boron tris-isopropoxide to produce a compound of Formula O-5.
  • a compound of Formula P-1 is treated with, for example, potassium thiocyanate and bromine in acetic acid to produce a compound of Formula P-2.
  • the compound of Formula P-2 is treated with an acteylating reagent such as acetyl chloride to produce a compound of Formula P-3.
  • the compound of P-3 is reacted with, for example, bis(pinacolato)diboron (compound P-4) in the presence of a catalyst such as palladium chloride to produce a compound of Formula P-5.
  • the compound of Formula P-2 is reacted with, for example, methyl carbamic acid chloride to produce a compound of Formula Q-1.
  • the compound of Formula Q-1 is reacted with bis(pinacolato)diboron (compound P-4) in the presence of a catalyst such as Pd 2 (dba) 3 , 2-chlorohexylphosphino-2, 4, 6-triisopropylbiphenyl, a base such as potassium acetate, to produce the compound of Formula Q-2.
  • a catalyst such as Pd 2 (dba) 3 , 2-chlorohexylphosphino-2, 4, 6-triisopropylbiphenyl, a base such as potassium acetate
  • Illustrative compounds of the invention include those of subclass 1a, 1b, 2a, 2b, 3a, 3b, 4a, 4b, 5a, 5b, 6a, 6b, 7a, 7b, 8a, 8b, 9a, 9b, 10a, 10b, 11a, 11b, 12a, 12b, 13a, 13b, 14a, 14b, 15a, 15b, 16a, or 16b, where the substituents R 1 , X 1 , and V are as described below.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is H and X 1 is N
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is CH 3 and X 1 is N
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is Et and X 1 is N
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is iPr and X 1 is N
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is iPr, X 1 is N, and V is NH 2 .
  • R 1 is iPr, X 1 is N, and V is NHCOMe.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is cyclopentyl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenyl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is pyridin-2-yl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is N-methylaminocyclohex-4-yl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is tert-butyl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is 1-cyano-but-4-yl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is 1-cyano-prop-3-yl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is 3-azetidinyl and X 1 is CH
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • V is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.
  • R 1 is phenylamino, benzyl, phenyl, NHMe, NH 2 , NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO 2 Me.

Abstract

The present invention provides for a method for treating a disease condition associated with PI3-kinase α and/or mTOR in a subject. In another aspect, the invention provides for a method for treating a disease condition associated with PI3-kinase α and/or mTOR in a subject. In yet another aspect, a method of inhibiting phosphorylation of both Akt (S473) and Akt (T308) in a cell is set forth.

Description

    CROSS-REFERENCE
  • This application claims the benefit of U.S. Provisional Application No. 61/446,016, filed on Feb. 23, 2011, which is hereby incorporated herein by reference in its entirety.
  • BACKGROUND OF THE INVENTION
  • Kinase signaling pathways play a central role in numerous biological processes. Defects in various components of signal transduction pathways have been found to account for a vast number of diseases, including numerous forms of cancer, inflammatory disorders, metabolic disorders, vascular and neuronal diseases (Gaestel et al. Current Medicinal Chemistry (2007) 14:2214-2234). In recent years, kinases that are associated with oncogenic signaling pathways have emerged as important drug targets in the treatment of various diseases including many types of cancers.
  • The mammalian target of rapamycin (mTOR), also known as mechanistic target of rapamycin, is a serine/threonine protein kinase that regulates cell growth, translational control, angiogenesis and/or cell survival. mTOR is encoded by the FK506 binding protein 12-rapamycin associated protein 1 (FRAP1) gene. mTOR is the catalytic subunit of two complexes, mTORC1 and mTORC2. mTORC1 is composed of mTOR, regulatory associated protein of mTOR (Raptor), mammalian LST8/G-protein β-subunit like protein (mLST8/GβL), PRAS40, and DEPTOR. mTOR Complex 2 (mTORC2) is composed of mTOR, rapamycin-insensitive companion of mTOR (Rictor), GβL, and mammalian stress-activated protein kinase interacting protein 1 (mSIN1).
  • Apart from their subunits, mTORC1 and mTORC2 are distinguished by their differential sensitivities to rapamycin and its analogs (also known as rapalogs). Rapamycin binds to and allosterically inhibits mTORC1, but mTORC2 is generally rapamycin-insensitive. As a result of this rapamycin-insensitive mTOR signaling mediated by mTORC2, cancer cells treated with rapamycin analogs usually display only partial inhibition of mTOR signaling, which can lead to enhanced survival and resistance to rapamycin treatment.
  • Another group of kinases involved in cellular functions that are commonly deregulated in diseases is the Phosphatidylinositol 3-kinases (PI 3-kinases or PI3Ks) family of enzymes. These lipid kinases phosphorylate the 3-position hydroxyl group of the inositol ring of phosphatidylinositol (PtdIns), activating signaling cascades associated with such processes as cell growth, proliferation, differentiation, motility, survival and intracellular trafficking. Disruption of these processes involving PI3K leads to many diseases including cancer, allergic contact dermatitis, rheumatoid arthritis, osteoarthritis, inflammatory bowel diseases, chronic obstructive pulmonary disorder, psoriasis, multiple sclerosis, asthma, disorders related to diabetic complications, and inflammatory complications of the cardiovascular system such as acute coronary syndrome.
  • The PI3K family comprises 15 kinases with distinct substrate specificities, expression patterns, and modes of regulation. The class I PI3Ks (p110α, p110β, p110δ, and p110γ) are typically activated by tyrosine kinases or G-protein coupled receptors to generate phosphatidylinositol-3,4,5-trisphosphate (PIP3), which engages downstream effectors such as those in the Akt/PDK1 pathway, mTOR, the Tec family kinases, and the Rho family GTPases.
  • The alpha (α) isoform of type I PI3K has been implicated in a variety of human cancers. Angiogenesis has been shown to selectively require the α isoform of PI3K in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453; 662-6). Mutations in the gene coding for PI3K α or mutations which lead to upregulation of PI3K a are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers. Often, mutations in the gene coding for PI3K α are point mutations clustered within several hotspots in helical and kinase domains, such as E542K, E545K, and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. While other PI3K isoforms such as PI3K δ or PI3K γ are expressed primarily in hematopoietic cells, PI3K α, along with PI3K β, is expressed constitutively.
  • The delta (δ) isoform of class I PI3K has been implicated, in particular, in a number of diseases and biological processes. PI3K δ is expressed primarily in hematopoietic cells including leukocytes such as T-cells, dendritic cells, neutrophils, mast cells, B-cells, and macrophages. PI3K δ is integrally involved in mammalian immune system functions such as T-cell function, B-cell activation, mast cell activation, dendritic cell function, and neutrophil activity. Due to its integral role in immune system function, PI3K δ is also involved in a number of diseases related to undesirable immune response such as allergic reactions, inflammatory diseases, inflammation mediated angiogenesis, rheumatoid arthritis, auto-immune diseases such as lupus, asthma, emphysema and other respiratory diseases. Other class I PI3K involved in immune system function includes PI3K γ, which plays a role in leukocyte signaling and has been implicated in inflammation, rheumatoid arthritis, and autoimmune diseases such as lupus.
  • PI3K β has been implicated primarily in various types of cancer including PTEN-negative cancer (Edgar et al. Cancer Research (2010) 70(3): 1164-1172), and HER2-overexpressing cancer such as breast cancer and ovarian cancer.
  • SUMMARY OF THE INVENTION
  • Due to the diverse essential functions of mTOR and PI3Ks, drugs that bind to and inhibit a broad range of kinase isoforms and complexes with low specificity can lead to deleterious side effects. For example, excessive inhibition of PI3K β may lead to undesirable effects on metabolic pathways and disruption of insulin signaling. Alternatively, excessive inhibition of PI3K δ and/or PI3K γ may disrupt or reduce immune function. The present disclosure provides an alternative approach that effectively targets disease-related pathways, while limiting undesirable side effects.
  • Accordingly, the invention provides a method for treating a disease condition associated with PI3-kinase α and/or mTOR in a subject, comprising administering to said subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase α inhibitor and an mTOR inhibitor, wherein the PI3-kinase α inhibitor exhibits selective inhibition of PI3-kinase α relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) ascertained by an in vitro kinase assay, wherein the one or more type I PI3-kinase is selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In one aspect, the combination comprises a therapeutic effective amount of a PI3-kinase α inhibitor and a therapeutic effective amount of an mTOR inhibitor. In another aspect, the combination comprises a synergistically effective therapeutic amount of PI3-kinase α inhibitor and an mTOR inhibitor, wherein the PI3-kinase α inhibitor and/or the mTOR inhibitor is present in a sub-therapeutic amount.
  • In some embodiments, the disease condition associated with PI3-kinase α and/or mTOR can include but are not limited to a neoplastic condition, autoimmune disease, inflammatory disease, fibrotic disease and kidney disease. For example, the neoplastic condition can be NSCLC, head and neck squamous cell carcinoma, pancreatic, breast and ovarian cancers, renal cell carcinoma, prostate cancer, neuroendocrine cancer, endometrial cancers, and other forms of cancer.
  • The invention further provides a method of inhibiting phosphorylation of both Akt (S473) and Akt (T308) in a cell, comprising contacting a cell with an effective amount of a PI3-kinase α inhibitor and an mTOR inhibitor that selectively inhibits both mTORC1 and mTORC2 activity relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) as ascertained by a cell-based assay or an in vitro kinase assay, wherein the PI3-kinase α inhibitor exhibits selective inhibition of PI3-kinase α relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) ascertained by an in vitro kinase assay, wherein the one or more type I PI3-kinase is selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α relative to all other type I phosphatidylinositol 3-kinases (PI3-kinase) consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • For instance, the PI3-kinase α inhibitor utilized in the subject methods inhibits PI3-kinase α with an IC50 value of about 500 nM or less, 400 nM or less, 300 nM or less, 200 nM or less, 100 nM or less, 10 nM or less, 1 nM or less as ascertained in an in vitro kinase assay. In another instance, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is at least 2, 5, 10, 50, 100, 1000 times less than its IC50 value against one, two, three or all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is less than about 200 nM, and said IC50 value is at least 2, 5 or 10 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α and/or PI3-kinase β with an IC50 value that is at least 5 times less than its IC50 value against PI3-kinase γ or PI3-kinase δ. In yet other embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α and/or PI3-kinase β with an IC50 value that is at least 50 times less than its IC50 value against PI3-kinase γ or PI3-kinase δ. In still other embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is at least 50 times less than its IC50 value against PI3-kinase γ or PI3-kinase δ.
  • In some embodiments of the methods of the invention, the mTOR inhibitor binds to and directly inhibits both mTORC1 and mTORC2. For example, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 500 nM or less, 400 nM or less, 300 nM or less, 200 nM or less, 100 nM or less, 50 nM or less, 10 nM or less, or 1 nM or less, as ascertained in an in vitro kinase assay. In another embodiment, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 10 nM or less as ascertained in an in vitro kinase assay, and the mTOR inhibitor is substantially inactive against one or more types I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. Alternatively, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay, and the IC50 value is at least 2, 5 or 10 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the mTor inhibitor inhibits mTORC1 selectively. For example, the mTor inhibitor inhibits mTORC1 with an IC50 value of about 1000 nM or less, 500 nM or less, 100 nM or less, 50 nM or less, 10 nM or less, as ascertained in an in vitro kinase. In some embodiments, the mTor inhibitor is rapamycin or an analogue of rapamycin. In other embodiments, the mTor inhibitor is sirolimus (rapamycin), deforolimus (AP23573, MK-8669), everolimus (RAD-001), temsirolimus (CCI-779), zotarolimus (ABT-578), or biolimus A9 (umirolimus).
  • In some embodiments, the mTOR inhibitor is a compound of Formula I:
  • Figure US20160235758A1-20160818-C00001
  • or a pharmaceutically acceptable salt thereof, wherein:
  • X1 is N or C-E1, X2 is N or C, X3 is N or C, X4 is C—R9 or N, X5 is N or C-E1, X6 is C or N, and X7 is C or N; and wherein no more than two nitrogen ring atoms are adjacent;
  • R1 is H, -L-C1-10alkyl, -L-C3-8cycloalkyl, —C3-8cycloalkyl, -L-aryl, -L-heteroaryl, -L-C1-10alkylaryl, -L-C1-10alkylhetaryl, -L-C1-10alkylheterocylyl, -L-C2-10alkenyl, -L-C2-10alkynyl, -L-C2-10alkenyl-C3-8cycloalkyl, -L-C2-10alkynyl-C3-8cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocylyl, -L-heteroalkyl-C3-8cycloalkyl, -L-aralkyl, -L-heteroaralkyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • E1 and E2 are independently —(W1)j—R4;
  • M1 is a 5, 6, 7, 8, 9, or -10 membered ring system, wherein the ring system is monocyclic or bicyclic, substituted with R5 and additionally optionally substituted with one or more —(W2)k—R2;
  • each k is 0 or 1;
  • j in E1 or j in E2, is independently 0 or 1;
  • W1 is —O—, —NR7—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • W2 is —O—, —NR7—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)C(O)N(R8)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • R2 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32, aryl (e.g. bicyclic aryl, unsubstituted aryl, or substituted monocyclic aryl), hetaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl —C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl (e.g. C2-10alkyl-monocyclic aryl, C1-10alkyl-substituted monocyclic aryl, or C1-10alkylbicycloaryl), C1-10alkylhetaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylhetaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylhetaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocylyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heteroalkyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl (e.g. monocyclic aryl-C2-10alkyl, substituted monocyclic aryl-C1-10alkyl, or bicycloaryl-C1-10alkyl), aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, hetaryl-C3-8cycloalkyl, hetaryl-heteroalkyl, or hetaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R3 and R4 are independently hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2C(═O) NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32 aryl, hetaryl, C1-4alkyl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl, C1-10alkylhetaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylhetaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylhetaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocylyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, hetaryl-C3-8cycloalkyl, heteroalkyl, hetaryl-heteroalkyl, or hetaryl-heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R5 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32;
  • each of R31, R32, and R33 is independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or hetaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35;
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, hetaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • each of R7′ and R8′ is independently hydrogen, C1-10alkyl, C2-10alkenyl, aryl, heteroaryl, heterocyclyl or C3-10cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R6;
  • R6 is halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2NR31R32, —NR31R32, or —NR34R35; and
  • R9 is H, halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2NR31R32, —NR31R32, or —NR34R35.
  • In other embodiments, the PI3-kinase α inhibitor is a compound of formula:
  • Figure US20160235758A1-20160818-C00002
  • or its pharmaceutically acceptable salts thereof, wherein:
  • W1′ is N, NR3′, or CR3′; W2 is N, NR4′, CR4′, or C═O; W3′ is N, NR5 or CR5; W4′ is N, wherein no more than two N atoms and no more than two C═O groups are adjacent;
  • W5′ is N;
  • W6 is N or CR8′;
  • Wa′ and Wb′ are independently N or CR9′;
  • one of We′ and Wd′ is N, and the other is O, NR10′, or S;
  • R1′ and R2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety; R5′, R6′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R9′ is alkyl or halo; and
  • R10′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In still other embodiments, the PI3-kinase α inhibitor is a compound of formula:
  • Figure US20160235758A1-20160818-C00003
  • or its pharmaceutically acceptable salts thereof, where:
  • X is O or S or N;
  • W1′ is S, N, NR3′ or CR3′, W2′ is N or CR4′, W3′ is S, N or CR5′, W4′ is N or C, and W7′ is N or C, wherein no more than two N atoms and no more than two C═O groups are adjacent;
  • W5′ is N or CR7′;
  • W6′ is N or CR8′;
  • R1′ and R2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety; and
  • R5′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • For any of the methods of the invention, the PI3-kinase α inhibitor and/or the mTOR inhibitor can be administered parenterally, orally, intraperitoneally, intravenously, intraarterially, transdermally, intramuscularly, liposomally, via local delivery by catheter or stent, subcutaneously, intraadiposally, or intrathecally. In some embodiments, the PI3-kinase α inhibitor and/or the mTOR inhibitor are co-administered to the subject in the same formulation. In other embodiments, the PI3-kinase α inhibitor and/or the mTOR inhibitor are co-administered to the subject in different formulations.
  • The invention also provides a pharmaceutical composition comprising a combination of an amount of PI3-kinase α inhibitor and an amount of mTOR inhibitor, wherein said combination provides a synergistic therapeutic effect in a subject in need thereof. For example, the pharmaceutical composition is formulated in an oral dosage. In some embodiments, at least one of the amounts is administered as a sub-therapeutic amount. In other embodiments, the pharmaceutical composition is formulated a tablet or a capsule. For example, the PI3-kinase α inhibitor and the mTOR inhibitor are packaged as separate tablets. In other embodiments, the PI3-kinase α inhibitor and the mTOR inhibitor are formulated as a single oral dosage form.
  • The invention also provides a method comprising: (a) determining the presence in a subject of a mutation in PI3-kinase α that is associated with a disease condition mediated by PI3-kinase α; and (b) administering to said subject a pharmaceutical composition of the invention. For example, the mutation is in the nucleotide sequence coding for PI3-kinase α. Exemplary mutations can include without limitation, deletion, insertion, translation, which can result in point mutations, frame shifts, and/or translation of the nucleic acid sequence coding for PI3-kinase α. In another example, the mutation is in the amino acid sequence of PI3-kinase α.
  • In some embodiments of any of the methods of the invention, the subject or cell comprises a mutation in PI3-kinase α which is associated with a disease condition mediated by PI3-kinase α.
  • INCORPORATION BY REFERENCE
  • All publications, patents, and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent, or patent application was specifically and individually indicated to be incorporated by reference.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The novel features of the invention are set forth with particularity in the appended claims. A better understanding of the features and advantages of the present invention will be obtained by reference to the following detailed description that sets forth illustrative embodiments, in which the principles of the invention are utilized, and the accompanying drawings of which:
  • FIG. 1 is a schematic illustration of multiple and distinct signaling pathways that are activated in human cancer.
  • FIG. 2 is a graph showing the synergistic effect of combined treatment with a PI3-kinase α inhibitor (Compound A) and an mTor inhibitor (Compound B) on tumor weight in a preclinical breast cancer model.
  • FIG. 3 is a western blot depicting the synergistic effect of combined treatment with Compound A and Compound B in terms of downregulating Akt and S6 phosphorylation.
  • FIG. 4 is a graph showing the synergistic effect of combined treatment with Compound A and rapamycin in terms of reducing tumor volume of a preclinical breast cancer model.
  • FIG. 5 shows A) an illustration of the distinct signaling pathways mediated by mTORC1 and mTORC2 and B) a western blot depicting sensitivity of mTORC1-dependent NRDG1 phosphorylation to Compound B, but not rapamycin.
  • FIG. 6 shows A) a graph depicting the selectivity of Compound B over Compound A in a PTEN-mutant negative control tumor model, B) a western blot depicting selective inhibition of Akt, S6, and 4EBP phosphorylation by Compound B and not Compound A in PTEN mutant cells, and C) a chart depicting the specificity of Compound A in inhibiting PI3K a over mTOR and other PI3K isoforms, and the specificity of Compound B in inhibiting mTOR over PI3K isoforms.
  • FIG. 7 is a western blot depicting differential inhibition of Akt phosphorylation at serine 473 over threonine 308 by Compound B (top panel). Also shown is the comparison of Akt phosphorylation inhibition for Pan-PI3K inhibitor versus Compound B.
  • FIG. 8 is a graph showing that Pan-PI3K inhibitor, but not Compound A, blocks B cell function in vivo. Mice were immunized with TNP-Ficoll and treated with 1) vehicle; 2) 70 mg/kg GDC0941; 3) 30 mg/kg Compound A; 4) 60 mg/kg Compound A; or 5) 120 mg/kg Compound A for 7 days. Antibody production was measured as a percentage of control group that were treated with vehicle.
  • FIG. 9 shows, left panel, a graph showing reduction in tumor weight of a breast cancer model using 70 mg/kg Pan-PI3K inhibitor and 60 mg/kg compound A and, right panel, reduced presence of MZB cells in mouse spleen for 70 mg/kg Pan-PI3K inhibitor compared to 60 mg/kg Compound A.
  • FIG. 10 illustrates the frequency of PI3K a mutation in various human cancers.
  • FIG. 11 is a western blot depicting inhibition of the PI3K pathway by Compound A in cell lines with elevated PI3K a activity. The left column shows data from MDA-MB-361 breast cancer cells harboring PIK3CA mutation. The middle column shows data from MDA-MB-453 breast cancer cells harboring PIK3CA mutation. The right column shows data from SKBr3breast cancer cells harboring HER2 mutation.
  • FIG. 12 shows A) a western blot showing inhibition of Akt phosphorylation at serine 473 by Compound A; and B) reduced inhibition of Akt phosphorylation at serine 473 by Compound A in a PTEN-mutant cell line.
  • FIG. 13 is a chart showing that Compound A preferentially inhibits proliferation of tumor cells harboring PI3K a mutations.
  • DETAILED DESCRIPTION OF THE INVENTION
  • Several aspects of the invention are described below with reference to example applications for illustration. It should be understood that numerous specific details, relationships, and methods are set forth to provide a full understanding of the invention. One having ordinary skill in the relevant art, however, will readily recognize that the invention can be practiced without one or more of the specific details or with other methods. The present invention is not limited by the illustrated ordering of acts or events, as some acts may occur in different orders and/or concurrently with other acts or events. Furthermore, not all illustrated acts or events are required to implement a methodology in accordance with the present invention.
  • The terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention. As used herein, the singular forms “a”, “an” and “the” are intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, to the extent that the terms “including”, “includes”, “having”, “has”, “with”, or variants thereof are used in either the detailed description and/or the claims, such terms are intended to be inclusive in a manner similar to the term “comprising”.
  • The term “about” or “approximately” means within an acceptable error range for the particular value as determined by one of ordinary skill in the art, which will depend in part on how the value is measured or determined, i.e., the limitations of the measurement system. For example, “about” can mean within 1 or more than 1 standard deviation, per the practice in the art. Alternatively, “about” can mean a range of up to 20%, preferably up to 10%, more preferably up to 5%, and more preferably still up to 1% of a given value. Alternatively, particularly with respect to biological systems or processes, the term can mean within an order of magnitude, preferably within 5-fold, and more preferably within 2-fold, of a value. Where particular values are described in the application and claims, unless otherwise stated the term “about” meaning within an acceptable error range for the particular value should be assumed.
  • “Treatment”, “treating”, “palliating” and “ameliorating”, as used herein, are used interchangeably. These terms refer to an approach for obtaining beneficial or desired results including but not limited to therapeutic benefit and/or a prophylactic benefit. By therapeutic benefit is meant eradication or amelioration of the underlying disorder being treated. Also, a therapeutic benefit is achieved with the eradication or amelioration of one or more of the physiological symptoms associated with the underlying disorder such that an improvement is observed in the patient, notwithstanding that the patient may still be afflicted with the underlying disorder. For prophylactic benefit, the compositions may be administered to a patient at risk of developing a particular disease, or to a patient reporting one or more of the physiological symptoms of a disease, even though a diagnosis of this disease may not have been made.
  • As used herein, the term “neoplastic condition” refers to the presence of cells possessing abnormal growth characteristics, such as uncontrolled proliferation, immortality, metastatic potential, rapid growth and proliferation rate, perturbed oncogenic signaling, and certain characteristic morphological features. This includes the abnormal growth of: (1) tumor cells (tumors) that proliferate by expressing a mutated tyrosine kinase or overexpression of a receptor tyrosine kinase; (2) benign and malignant cells of other proliferative diseases in which aberrant tyrosine kinase activation occurs; (3) any tumors that proliferate by receptor tyrosine kinases; (4) any tumors that proliferate by aberrant serine/threonine kinase activation; and (5) benign and malignant cells of other proliferative diseases in which aberrant serine/threonine kinase activation occurs.
  • The term “effective amount” or “therapeutically effective amount” refers to that amount of an inhibitor described herein that is sufficient to effect the intended application including but not limited to disease treatment, as defined below. The therapeutically effective amount may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or downregulation of activity of a target protein. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • A “sub-therapeutic amount” of an agent or therapy is an amount less than the effective amount for that agent or therapy, but when combined with an effective or sub-therapeutic amount of another agent or therapy can produce a result desired by the physician, due to, for example, synergy in the resulting efficacious effects, or reduced side effects.
  • A “synergistically effective therapeutic amount” of an agent or therapy is an amount which, when combined with an effective or sub-therapeutic amount of another agent or therapy, produces a greater effect than when either of the two agents are therapies are used alone. In some embodiments, a syngergistically effective therapeutic amount of an agent or therapy produces a greater effect when used in combination than the additive effects of each of the two agents or therapies when used alone.
  • As used herein, “agent” or “biologically active agent” refers to a biological, pharmaceutical, or chemical compound or other moiety. Non-limiting examples include simple or complex organic or inorganic molecule, a peptide, a protein, an oligonucleotide, an antibody, an antibody derivative, antibody fragment, a vitamin derivative, a carbohydrate, a toxin, or a chemotherapeutic compound. Various compounds can be synthesized, for example, small molecules and oligomers (e.g., oligopeptides and oligonucleotides), and synthetic organic compounds based on various core structures. In addition, various natural sources can provide compounds for screening, such as plant or animal extracts, and the like. A skilled artisan can readily recognize that there is no limit as to the structural nature of the agents of the present invention.
  • The term “agonist” as used herein refers to a compound having the ability to initiate or enhance a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the term “agonist” is defined in the context of the biological role of the target polypeptide. While preferred agonists herein specifically interact with (e.g., bind to) the target, compounds that initiate or enhance a biological activity of the target polypeptide by interacting with other members of the signal transduction pathway of which the target polypeptide is a member are also specifically included within this definition.
  • The terms “antagonist” and “inhibitor” are used interchangeably, and they refer to a compound having the ability to inhibit a biological function of a target protein, whether by inhibiting the activity or expression of the target protein. Accordingly, the terms “antagonist” and “inhibitors” are defined in the context of the biological role of the target protein. While preferred antagonists herein specifically interact with (e.g., bind to) the target, compounds that inhibit a biological activity of the target protein by interacting with other members of the signal transduction pathway of which the target protein is a member are also specifically included within this definition. A preferred biological activity inhibited by an antagonist is associated with the development, growth, or spread of a tumor, or an undesired immune response as manifested in autoimmune disease.
  • The phrase “mTOR inhibitor that binds to and directly inhibits both mTORC1 and mTORC2 kinases” when used: herein refers to an mTOR inhibitor that interacts with and reduces the kinase activity of both mTORC1 and mTORC2 complexes.
  • An “anti-cancer agent”, “anti-tumor agent” or “chemotherapeutic agent” refers to any agent useful in the treatment of a neoplastic condition. One class of anti-cancer agents comprises chemotherapeutic agents. “Chemotherapy” means the administration of one or more chemotherapeutic drugs and/or other agents to a cancer patient by various methods, including intravenous, oral, intramuscular, intraperitoneal, intravesical, subcutaneous, transdermal, buccal, or inhalation or in the form of a suppository.
  • The term “cell proliferation” refers to a phenomenon by which the cell number has changed as a result of division. This term also encompasses cell growth by which the cell morphology has changed (e.g., increased in size) consistent with a proliferative signal.
  • The terms “co-administration,” “administered in combination with,” and their grammatical equivalents, encompass administration of two or more agents to an animal so that both agents and/or their metabolites are present in the animal at the same time. Co-administration includes simultaneous administration in separate compositions, administration at different times in separate compositions, or administration in a composition in which both agents are present. Co-administered agents may be in the same formulation. Co-administered agents may also be in different formulations.
  • A “therapeutic effect,” as used herein, encompasses a therapeutic benefit and/or a prophylactic benefit as described above. A prophylactic effect includes delaying or eliminating the appearance of a disease or condition, delaying or eliminating the onset of symptoms of a disease or condition, slowing, halting, or reversing the progression of a disease or condition, or any combination thereof.
  • The term “pharmaceutically acceptable salt” refers to salts derived from a variety of organic and inorganic counter ions well known in the art. Pharmaceutically acceptable acid addition salts can be formed with inorganic acids and organic acids. Inorganic acids from which salts can be derived include, for example, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. Organic acids from which salts can be derived include, for example, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like. Pharmaceutically acceptable base addition salts can be formed with inorganic and organic bases. Inorganic bases from which salts can be derived include, for example, sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum, and the like. Organic bases from which salts can be derived include, for example, primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines, basic ion exchange resins, and the like, specifically such as isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, and ethanolamine. In some embodiments, the pharmaceutically acceptable base addition salt is chosen from ammonium, potassium, sodium, calcium, and magnesium salts.
  • “Pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions of the invention is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • “Signal transduction” is a process during which stimulatory or inhibitory signals are transmitted into and within a cell to elicit an intracellular response. A modulator of a signal transduction pathway refers to a compound which modulates the activity of one or more cellular proteins mapped to the same specific signal transduction pathway. A modulator may augment (agonist) or suppress (antagonist) the activity of a signaling molecule.
  • The term “selective inhibition” or “selectively inhibit” as applied to a biologically active agent refers to the agent's ability to selectively reduce the target signaling activity as compared to off-target signaling activity, via direct or interact interaction with the target.
  • “Subject” refers to an animal, such as a mammal, for example a human. The methods described herein can be useful in both human therapeutics, pre-clinical, and veterinary applications. In some embodiments, the subject is a mammal, and in some embodiments, the subject is human.
  • The term “in vivo” refers to an event that takes place in a subject's body.
  • The term “in vitro” refers to an event that takes places outside of a subject's body. For example, an in vitro assay encompasses any assay run outside of a subject assay. In vitro assays encompass cell-based assays in which cells alive or dead are employed. In vitro assays also encompass a cell-free assay in which no intact cells are employed.
  • When ranges are used herein for physical properties, such as molecular weight, or chemical properties, such as chemical formulae, all combinations and subcombinations of ranges and specific embodiments therein are intended to be included. The term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary from, for example, between 1% and 15% of the stated number or numerical range. The term “comprising” (and related terms such as “comprise” or “comprises” or “having” or “including”) includes those embodiments, for example, an embodiment of any composition of matter, composition, method, or process, or the like, that “consist of” or “consist essentially of” the described features.
  • The following abbreviations and terms have the indicated meanings throughout: PI3K=Phosphoinositide 3-kinase; PI=phosphatidylinositol.
  • Unless otherwise stated, the connections of compound name moieties are at the rightmost recited moiety. That is, the substituent name starts with a terminal moiety, continues with any linking moieties, and ends with the linking moiety. For example, heteroarylthio C1-4 alkyl has a heteroaryl group connected through a thio sulfur to a C1-4 alkyl radical that connects to the chemical species bearing the substituent. This condition does not apply where a formula such as, for example “-L-C1-10 alkyl-C3-8cycloalkyl” is represented. In such case, the terminal group is a C3-8cycloalkyl group attached to a linking C1-10 alkyl moiety which is attached to an element L, which is itself connected to the chemical species bearing the substituent.
  • “Alkyl” refers to a straight or branched hydrocarbon chain radical consisting solely of carbon and hydrogen atoms, containing no unsaturation, having from one to ten carbon atoms (e.g., C1-C10 alkyl). Whenever it appears herein, a numerical range such as “1 to 10” refers to each integer in the given range; e.g., “1 to 10 carbon atoms” means that the alkyl group may consist of 1 carbon atom, 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms, although the present definition also covers the occurrence of the term “alkyl” where no numerical range is designated. In some embodiments, it is a C1-C4 alkyl group. Typical alkyl groups include, but are in no way limited to, methyl, ethyl, propyl, isopropyl, n-butyl, iso-butyl, sec-butyl isobutyl, tertiary butyl, pentyl, isopentyl, neopentyl, hexyl, septyl, octyl, nonyl, decyl, and the like. The alkyl is attached to the rest of the molecule by a single bond, for example, methyl (Me), ethyl (Et), n-propyl, 1-methylethyl (iso-propyl), n-butyl, n-pentyl, 1,1-dimethylethyl (t-butyl), 3-methylhexyl, 2-methylhexyl, and the like. Unless stated otherwise specifically in the specification, an alkyl group is optionally substituted by one or more of substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —ORa, —SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —OC(O)N(Ra)2, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)C(O)N(Ra)2, —N(Ra)C(NRa)N(Ra)2, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2 where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • The term “halo” or “halogen” refers to fluoro, chloro, bromo, or iodo.
  • The term “haloalkyl” refers to an alkyl group substituted with one or more halo groups, for example chloromethyl, 2-bromoethyl, 3-iodopropyl, trifluoromethyl, perfluoropropyl, 8-chlorononyl, and the like.
  • “Acyl” refers to the groups (alkyl)-C(O)—, (aryl)-C(O)—, (heteroaryl)-C(O)—, (heteroalkyl)-C(O)—, and (heterocycloalkyl)-C(O)—, wherein the group is attached to the parent structure through the carbonyl functionality. In some embodiments, it is a C1-C10 acyl radical which refers to the total number of chain or ring atoms of the alkyl, aryl, heteroaryl or heterocycloalkyl portion of the acyloxy group plus the carbonyl carbon of acyl, i.e three other ring or chain atoms plus carbonyl. If the R radical is heteroaryl or heterocycloalkyl, the hetero ring or chain atoms contribute to the total number of chain or ring atoms. Unless stated otherwise specifically in the specification, the “R” of an acyloxy group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —ORa, SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —OC(O)N(Ra)2, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)C(O)N(Ra)2, —N(Ra)C(NRa)N(Ra)2, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • “Cycloalkyl” refers to a monocyclic or polycyclic radical that contains only carbon and hydrogen, and may be saturated, or partially unsaturated. Cycloalkyl groups include groups having from 3 to 10 ring atoms (i.e., C2-C10 cycloalkyl). Whenever it appears herein, a numerical range such as “3 to 10” refers to each integer in the given range; e.g., “3 to 10 carbon atoms” means that the cycloalkyl group may consist of 3 carbon atoms, etc., up to and including 10 carbon atoms. In some embodiments, it is a C3-C8 cycloalkyl radical. In some embodiments, it is a C3-C5 cycloalkyl radical. Illustrative examples of cycloalkyl groups include, but are not limited to the following moieties: cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloseptyl, cyclooctyl, cyclononyl, cyclodecyl, norbornyl, and the like. Unless stated otherwise specifically in the specification, a cycloalkyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —ORa, —SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —OC(O)N(Ra)2, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)C(O)N(Ra)2, —N(Ra)C(NRa)N(Ra)2, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O), OR′ (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • The term “C1-10alkyl C3-8cycloalkyl” is used to describe an alkyl group, branched or straight chain and containing 1 to 10 carbon atoms, attached to a linking cycloalkyl group which contains 3 to 8 carbons, such as for example, 2-methyl cyclopropyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “bicycloalkyl” refers to a structure consisting of two cycloalkyl moieties, unsubstituted or substituted, that have two or more atoms in common. If the cycloalkyl moieties have exactly two atoms in common they are said to be “fused”. Examples include, but are not limited to, bicyclo[3.1.0]hexyl, perhydronaphthyl, and the like. If the cycloalkyl moieties have more than two atoms in common they are said to be “bridged”. Examples include, but are not limited to, bicyclo[3.2.1]heptyl (“norbornyl”), bicyclo[2.2.2]octyl, and the like.
  • As used herein, the term “heteroatom” or “ring heteroatom” is meant to include oxygen (O), nitrogen (N), sulfur (S), phosphorus (P), and silicon (Si).
  • “Heteroalkyl”, “heteroalkenyl” and “heteroalkynyl” include optionally substituted alkyl, alkenyl and alkynyl radicals and which have one or more skeletal chain atoms selected from an atom other than carbon, e.g., oxygen, nitrogen, sulfur, phosphorus or combinations thereof. A numerical range may be given, e.g., C1-C4 heteroalkyl which refers to the chain length in total, which in this example is 4 atoms long. For example, a —CH2OCH2CH3 radical is referred to as a “C4” heteroalkyl, which includes the heteroatom center in the atom chain length description. Connection to the rest of the molecule may be through either a heteroatom or a carbon in the heteroalkyl chain. A heteroalkyl group may be substituted with one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, —ORa, —SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)S(O)tRa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • The term “heteroalkylaryl” refers to a heteroalkyl group as defined above which is attached to an aryl group, and may be attached at a terminal point or through a branched portion of the heteroalkyl, for example, an benzyloxymethyl moiety. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroalkylheteroaryl” refers likewise to a heteroalkyl group which is attached to a heteroaryl moiety, for example, an ethoxymethylpyridyl group. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroalkyl-heterocyclyl” refers to a heteroalkyl group as defined above, which is attached to a heterocyclic group, for example, 4(3-aminopropyl)-N-piperazinyl. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroalkyl-C3-8cycloalkyl” refers to a heteroalkyl group as defined above, which is attached to a cyclic alkyl containing 3 to 8 carbons, for example, 1-aminobutyl-4-cyclohexyl. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterobicycloalkyl” refers to a bicycloalkyl structure, which is unsubstituted or substituted, in which at least one carbon atom is replaced with a heteroatom independently selected from oxygen, nitrogen, and sulfur.
  • The term “heterospiroalkyl” refers to a spiroalkyl structure, which is unsubstituted or substituted, in which at least one carbon atom is replaced with a heteroatom independently selected from oxygen, nitrogen, and sulfur.
  • An “alkene” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon double bond, and an “alkyne” moiety refers to a group consisting of at least two carbon atoms and at least one carbon-carbon triple bond. The alkyl moiety, whether saturated or unsaturated, may be branched, straight chain, or cyclic.
  • “Alkenyl” refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one double bond, and having from two to ten carbon atoms (i.e., C2-C10 alkenyl). Whenever it appears herein, a numerical range such as “2 to 10” refers to each integer in the given range; e.g., “2 to 10 carbon atoms” means that the alkenyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkenyl comprises two to eight carbon atoms. In other embodiments, an alkenyl comprises two to five carbon atoms (e.g., C2-C5 alkenyl). The alkenyl is attached to the rest of the molecule by a single bond, for example, ethenyl (i.e., vinyl), prop-1-enyl (i.e., allyl), but-1-enyl, pent-1-enyl, penta-1,4-dienyl, and the like. Unless stated otherwise specifically in the specification, an alkenyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —ORa, —SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —OC(O)N(Ra)2, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)C(O)N(Ra)2, —N(Ra)C(NRa)N(Ra)2, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • The term “C2-10 alkenyl-heteroalkyl” refers to a group having an alkenyl moiety, containing 2 to 10 carbon atoms and is branched or straight chain, which is attached to a linking heteroalkyl group, such as, for example, allyloxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C2-10 alkynyl-heteroalkyl” refers to a group having an alkynyl moiety, which is unsubstituted or substituted, containing 2 to 10 carbon atoms and is branched or straight chain, which is attached to a linking heteroalkyl group, such as, for example, 4-but-1-ynoxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “haloalkenyl” refers to an alkenyl group substituted with one or more halo groups.
  • Unless otherwise specified, the term “cycloalkenyl” refers to a cyclic aliphatic 3 to 8 membered ring structure, optionally substituted with alkyl, hydroxy and halo, having 1 or 2 ethylenic bonds such as methylcyclopropenyl, trifluoromethylcyclopropenyl, cyclopentenyl, cyclohexenyl, 1,4-cyclohexadienyl, and the like.
  • “Alkynyl” refers to a straight or branched hydrocarbon chain radical group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having from two to ten carbon atoms (i.e., C2-C10 alkynyl). Whenever it appears herein, a numerical range such as “2 to 10” refers to each integer in the given range; e.g., “2 to 10 carbon atoms” means that the alkynyl group may consist of 2 carbon atoms, 3 carbon atoms, etc., up to and including 10 carbon atoms. In certain embodiments, an alkynyl comprises two to eight carbon atoms. In other embodiments, an alkynyl has two to five carbon atoms (e.g., C2-C5 alkynyl). The alkynyl is attached to the rest of the molecule by a single bond, for example, ethynyl, propynyl, butynyl, pentynyl, hexynyl, and the like. Unless stated otherwise specifically in the specification, an alkynyl group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —ORa, SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —OC(O)N(Ra)2, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)C(O)N(Ra)2, N(Ra)C(NRa)N(Ra)2, —N(Ra)S(O)tRa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • The term C2-10 alkynyl-C3-8 cycloalkyl refers to a group containing an alkynyl group, containing 2 to 10 carbons and branched or straight chain, which is attached to a linking cycloalkyl group containing 3 to 8 carbons, such as, for example 3-prop-3-ynyl-cyclopent-1yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “haloalkynyl” refers to an alkynyl group substituted with one or more independent halo groups.
  • “Amino” or “amine” refers to a —N(Ra)2 radical group, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl, unless stated otherwise specifically in the specification. When a —N(Ra)2 group has two Ra other than hydrogen they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring. For example, —N(Ra)2 is meant to include, but not be limited to, 1-pyrrolidinyl and 4-morpholinyl. Unless stated otherwise specifically in the specification, an amino group is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —ORa, —SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —OC(O)N(Ra)2, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)C(O)N(Ra)2, —N(Ra)C(NRa)N(Ra)2, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl and each of these moieties may be optionally substituted as defined herein.
  • “Amide” or “amido” refers to a chemical moiety with formula —C(O)N(R)2 or —NHC(O)R, where R is selected from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon), each of which moiety may itself be optionally substituted. In some embodiments it is a C1-C4 amido or amide radical, which includes the amide carbonyl in the total number of carbons in the radical. The R2′ of —N(R)2 of the amide may optionally be taken together with the nitrogen to which it is attached to form a 4-, 5-, 6-, or 7-membered ring. Unless stated otherwise specifically in the specification, an amido group is optionally substituted independently by one or more of the substituents as described herein for alkyl, cycloalkyl, aryl, heteroaryl, or heterocycloalkyl. An amide may be an amino acid or a peptide molecule attached to a compound of Formula (I), thereby forming a prodrug. Any amine, hydroxy, or carboxyl side chain on the compounds described herein can be amidified. The procedures and specific groups to make such amides are known to those of skill in the art and can readily be found in reference sources such as Greene and Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley & Sons, New York, N.Y., 1999, which is incorporated herein by reference in its entirety.
  • “Aromatic” or “aryl” refers to an aromatic radical with six to ten ring atoms (e.g., C6-C10 aromatic or C6-C10 aryl) which has at least one ring having a conjugated pi electron system which is carbocyclic (e.g., phenyl, fluorenyl, and naphthyl). Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals. Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in “-yl” by removal of one hydrogen atom from the carbon atom with the free valence are named by adding “-idene” to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene. Whenever it appears herein, a numerical range such as “6 to 10” refers to each integer in the given range; e.g., “6 to 10 ring atoms” means that the aryl group may consist of 6 ring atoms, 7 ring atoms, etc., up to and including 10 ring atoms. The term includes monocyclic or fused-ring polycyclic (i.e., rings which share adjacent pairs of ring atoms) groups. Unless stated otherwise specifically in the specification, an aryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, trifluoromethyl, trifluoromethoxy, nitro, trimethylsilanyl, —ORa, —SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —OC(O)N(W)2, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)C(O)N(Ra)2, N(Ra)C(NRa)N(Ra)2, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(W)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • “Heteroaryl” or, alternatively, “heteroaromatic” refers to a 5- to 18-membered aromatic radical (e.g., C5-C13 heteroaryl) that includes one or more ring heteroatoms selected from nitrogen, oxygen and sulfur, and which may be a monocyclic, bicyclic, tricyclic or tetracyclic ring system. Whenever it appears herein, a numerical range such as “5 to 18” refers to each integer in the given range; e.g., “5 to 18 ring atoms” means that the heteroaryl group may consist of 5 ring atoms, 6 ring atoms, etc., up to and including 18 ring atoms. Bivalent radicals derived from univalent heteroaryl radicals whose names end in “-yl” by removal of one hydrogen atom from the atom with the free valence are named by adding “-idene” to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene. An N-containing “heteroaromatic” or “heteroaryl” moiety refers to an aromatic group in which at least one of the skeletal atoms of the ring is a nitrogen atom. The polycyclic heteroaryl group may be fused or non-fused. The heteroatom(s) in the heteroaryl radical is optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heteroaryl is attached to the rest of the molecule through any atom of the ring(s). Examples of heteroaryls include, but are not limited to, azepinyl, acridinyl, benzimidazolyl, benzindolyl, 1,3-benzodioxolyl, benzofuranyl, benzooxazolyl, benzo[d]thiazolyl, benzothiadiazolyl, benzo[b][1,4]dioxepinyl, benzo[b][1,4]oxazinyl, 1,4-benzodioxanyl, benzonaphthofuranyl, benzoxazolyl, benzodioxolyl, benzodioxinyl, benzoxazolyl, benzopyranyl, benzopyranonyl, benzofuranyl, benzofuranonyl, benzofurazanyl, benzothiazolyl, benzothienyl (benzothiophenyl), benzothieno[3,2-d]pyrimidinyl, benzotriazolyl, benzo[4,6]imidazo[1,2-a]pyridinyl, carbazolyl, cinnolinyl, cyclopenta[d]pyrimidinyl, 6,7-dihydro-5H-cyclopenta[4,5]thieno[2,3-d]pyrimidinyl, 5,6-dihydrobenzo[h]quinazolinyl, 5,6-dihydrobenzo[h]cinnolinyl, 6,7-dihydro-5H-benzo[6,7]cyclohepta[1,2-c]pyridazinyl, dibenzofuranyl, dibenzothiophenyl, furanyl, furazanyl, furanonyl, furo[3,2-c]pyridinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyrimidinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyridazinyl, 5,6,7,8,9,10-hexahydrocycloocta[d]pyridinyl, isothiazolyl, imidazolyl, indazolyl, indolyl, indazolyl, isoindolyl, indolinyl, isoindolinyl, isoquinolyl, indolizinyl, isoxazolyl, 5,8-methano-5,6,7,8-tetrahydroquinazolinyl, naphthyridinyl, 1,6-naphthyridinonyl, oxadiazolyl, 2-oxoazepinyl, oxazolyl, oxiranyl, 5,6,6a,7,8,9,10,10a-octahydrobenzo[h]quinazolinyl, 1-phenyl-1H-pyrrolyl, phenazinyl, phenothiazinyl, phenoxazinyl, phthalazinyl, pteridinyl, purinyl, pyranyl, pyrrolyl, pyrazolyl, pyrazolo[3,4-d]pyrimidinyl, pyridinyl, pyrido[3,2-d]pyrimidinyl, pyrido[3,4-d]pyrimidinyl, pyrazinyl, pyrimidinyl, pyridazinyl, pyrrolyl, quinazolinyl, quinoxalinyl, quinolinyl, isoquinolinyl, tetrahydroquinolinyl, 5,6,7,8-tetrahydroquinazolinyl, 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidinyl, 6,7,8,9-tetrahydro-5H-cyclohepta[4,5]thieno[2,3-d]pyrimidinyl, 5,6,7,8-tetrahydropyrido[4,5-c]pyridazinyl, thiazolyl, thiadiazolyl, thiapyranyl, triazolyl, tetrazolyl, triazinyl, thieno[2,3-d]pyrimidinyl, thieno[3,2-d]pyrimidinyl, thieno[2,3-c]pridinyl, and thiophenyl (i.e. thienyl). Unless stated otherwise specifically in the specification, a heteraryl moiety is optionally substituted by one or more substituents which are independently: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, —ORa, —SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O), ORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heterocycloalkylalkyl, heteroaryl or heteroarylalkyl.
  • The terms “aryl-alkyl”, “arylalkyl” and “aralkyl” are used to describe a group wherein the alkyl chain can be branched or straight chain forming a linking portion with the terminal aryl, as defined above, of the aryl-alkyl moiety. Examples of aryl-alkyl groups include, but are not limited to, optionally substituted benzyl, phenethyl, phenpropyl and phenbutyl such as 4-chlorobenzyl, 2,4-dibromobenzyl, 2-methylbenzyl, 2-(3-fluorophenyl)ethyl, 2-(4-methylphenyl)ethyl, 2-(4-(trifluoromethyl)phenyl)ethyl, 2-(2-methoxyphenyl)ethyl, 2-(3-nitrophenyl)ethyl, 2-(2,4-dichlorophenyl)ethyl, 2-(3,5-dimethoxyphenyl)ethyl, 3-phenylpropyl, 3-(3-chlorophenyl)propyl, 3-(2-methylphenyl)propyl, 3-(4-methoxyphenyl)propyl, 3-(4-(trifluoromethyl)phenyl)propyl, 3-(2,4-dichlorophenyl)propyl, 4-phenylbutyl, 4-(4-chlorophenyl)butyl, 4-(2-methylphenyl)butyl, 4-(2,4-dichlorophenyl)butyl, 4-(2-methoxphenyl)butyl, and 10-phenyldecyl. Either portion of the moiety is unsubstituted or substituted.
  • The term “C1-10alkylaryl” as used herein refers to an alkyl group, as defined above, containing 1 to 10 carbon atoms, branched or unbranched, wherein the aryl group replaces one hydrogen on the alkyl group, for example, 3-phenylpropyl. Either portion of the moiety is unsubstituted or substituted.
  • The term C2-10 alkyl monocycloaryl” refers to a group containing a terminal alkyl group, branched or straight chain and containing 2 to 10 atoms attached to a linking aryl group which has only one ring, such as for example, 2-phenyl ethyl. Either portion of the moiety is unsubstituted or substituted.
  • The term “C1-10 alkyl bicycloaryl” refers to a group containing a terminal alkyl group, branched or straight chain and containing 2 to 10 atoms attached to a linking aryl group which is bicyclic, such as for example, 2-(1-naphthyl)-ethyl. Either portion of the moiety is unsubstituted or substituted.
  • The terms “aryl-cycloalkyl” and “arylcycloalkyl” are used to describe a group wherein the terminal aryl group is attached to a cycloalkyl group, for example phenylcyclopentyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “heteroaryl-C3-8cycloalkyl” and “heteroaryl-C3-8cycloalkyl” are used to describe a group wherein the terminal heteroaryl group is attached to a cycloalkyl group, which contains 3 to 8 carbons, for example pyrid-2-yl-cyclopentyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroaryl-heteroalkyl” refers to a group wherein the terminal heteroaryl group is attached to a linking heteroalkyl group, such as for example, pyrid-2-yl methylenoxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “aryl-alkenyl”, “arylalkenyl” and “aralkenyl” are used to describe a group wherein the alkenyl chain can be branched or straight chain forming a linking portion of the aralkenyl moiety with the terminal aryl portion, as defined above, for example styryl (2-phenylvinyl), phenpropenyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “aryl-C2-10alkenyl” means an arylalkenyl as described above wherein the alkenyl moiety contains 2 to 10 carbon atoms such as for example, styryl (2-phenylvinyl), and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C2-10alkenyl-aryl” is used to describe a group wherein the terminal alkenyl group, which contains 2 to 10 carbon atoms and can be branched or straight chain, is attached to the aryl moiety which forms the linking portion of the alkenyl-aryl moiety, such as for example, 3-propenyl-naphth-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “aryl-alkynyl”, “arylalkynyl” and “aralkynyl” are used to describe a group wherein the alkynyl chain can be branched or straight chain forming a linking portion of the aryl-alkynyl moiety with the terminal aryl portion, as defined above, for example 3-phenyl-1-propynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “aryl-C2-10alkynyl” means an arylalkynyl as described above wherein the alkynyl moiety contains two to ten carbons, such as, for example 3-phenyl-1-propynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C2-10alkynyl-aryl” means a group containing an alkynyl moiety attached to an aryl linking group, both as defined above, wherein the alkynyl moiety contains two to ten carbons, such as, for example 3-propynyl-naphth-1-yl. Either portion of the moiety is unsubstituted or substituted.
  • The terms “aryl-oxy”, “aryloxy” and “aroxy” are used to describe a terminal aryl group attached to a linking oxygen atom. Typical aryl-oxy groups include phenoxy, 3,4-dichlorophenoxy, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “aryl-oxyalkyl”, “aryloxyalkyl” and “aroxyalkyl” are used to describe a group wherein an alkyl group is substituted with a terminal aryl-oxy group, for example pentafluorophenoxymethyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C1-10alkoxy-C1-10alkyl” refers to a group wherein an alkoxy group, containing 1 to 10 carbon atoms and an oxygen atom within the branching or straight chain, is attached to a linking alkyl group, branched or straight chain which contains 1 to 10 carbon atoms, such as, for example methoxypropyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C1-10alkoxy-C2-10alkenyl” refers to a group wherein an alkoxy group, containing 1 to 10 carbon atoms and an oxygen atom within the branching or straight chain, is attached to a linking alkenyl group, branched or straight chain which contains 1 to 10 carbon atoms, such as, for example 3-methoxybut-2-en-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C1-10alkoxy-C2-10alkynyl” refers to a group wherein an alkoxy group, containing 1 to 10 carbon atoms and an oxygen atom within the branching or straight chain, is attached to a linking alkynyl group, branched or straight chain which contains 1 to 10 carbon atoms, such as, for example 3-methoxybut-2-in-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocycloalkenyl” refers to a cycloalkenyl structure, which is unsubstituted or substituted in which at least one carbon atom is replaced with a heteroatom selected from oxygen, nitrogen, and sulfur.
  • The terms “heteroaryl-oxy”, “heteroaryl-oxy”, “heteroaryloxy”, “heteroaryloxy”, “hetaroxy” and “heteroaroxy” are used to describe a terminal heteroaryl group, which is unsubstituted or substituted, attached to a linking oxygen atom. Typical heteroaryl-oxy groups include 4,6-dimethoxypyrimidin-2-yloxy and the like.
  • The terms “heteroarylalkyl”, “heteroarylalkyl”, “heteroaryl-alkyl”, “heteroaryl-alkyl”, “hetaralkyl” and “heteroaralkyl” are used to describe a group wherein the alkyl chain can be branched or straight chain forming a linking portion of the heteroaralkyl moiety with the terminal heteroaryl portion, as defined above, for example 3-furylmethyl, thenyl, furfuryl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroaryl-C1-10alkyl” is used to describe a heteroaryl alkyl group as described above where the alkyl group contains 1 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • The term “C1-10alkyl-heteroaryl” is used to describe a alkyl attached to a hetary group as described above where the alkyl group contains 1 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • The terms “heteroarylalkenyl”, “heteroarylalkenyl”, “heteroaryl-alkenyl”, “heteroaryl-alkenyl”, “hetaralkenyl” and “heteroaralkenyl” are used to describe a heteroarylalkenyl group wherein the alkenyl chain can be branched or straight chain forming a linking portion of the heteroaralkenyl moiety with the terminal heteroaryl portion, as defined above, for example 3-(4-pyridyl)-1-propenyl. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroaryl-C2-10alkenyl” group is used to describe a group as described above wherein the alkenyl group contains 2 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • The term “C2-10alkenyl-heteroaryl” is used to describe a group containing an alkenyl group, which is branched or straight chain and contains 2 to 10 carbon atoms, and is attached to a linking heteroaryl group, such as, for example 2-styryl-4-pyridyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “heteroarylalkynyl”, “heteroarylalkynyl”, “heteroaryl-alkynyl”, “heteroaryl-alkynyl”, “hetaralkynyl” and “heteroaralkynyl” are used to describe a group wherein the alkynyl chain can be branched or straight chain forming a linking portion of the heteroaralkynyl moiety with the heteroaryl portion, as defined above, for example 4-(2-thienyl)-1-butynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroaryl-C2-10alkynyl” is used to describe a heteroarylalkynyl group as described above wherein the alkynyl group contains 2 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • The term “C2-10alkynyl-heteroaryl” is used to describe a group containing an alkynyl group which contains 2 to 10 carbon atoms and is branched or straight chain, which is attached to a linking heteroaryl group such as, for example, 4(but-1-ynyl) thien-2-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocyclyl” refers to a four-, five-, six-, or seven-membered ring containing one, two, three or four heteroaroms independently selected from nitrogen, oxygen and sulfur. The four-membered ring has zero double bonds, the five-membered ring has zero to two double bonds, and the six- and seven-membered rings have zero to three double bonds. The term “heterocyclyl” also includes bicyclic groups in which the heterocyclyl ring is fused to another monocyclic heterocyclyl group, or a four- to se-membered aromatic or nonaromatic carbocyclic ring. The heterocyclyl group can be attached to the parent molecular moiety through any carbon atom or nitrogen atom in the group.
  • “Heterocycloalkyl” refers to a stable 3- to 18-membered non-aromatic ring radical that comprises two to twelve carbon atoms and from one to six heteroatoms selected from nitrogen, oxygen and sulfur. Whenever it appears herein, a numerical range such as “3 to 18” refers to each integer in the given range; e.g., “3 to 18 ring atoms” means that the heterocycloalkyl group may consist of 3 ring atoms, 4 ring atoms, etc., up to and including 18 ring atoms. In some embodiments, it is a C5-C10 heterocycloalkyl. In some embodiments, it is a C4-C10 heterocycloalkyl. In some embodiments, it is a C3-C10 heterocycloalkyl. Unless stated otherwise specifically in the specification, the heterocycloalkyl radical is a monocyclic, bicyclic, tricyclic or tetracyclic ring system, which may include fused or bridged ring systems. The heteroatoms in the heterocycloalkyl radical may be optionally oxidized. One or more nitrogen atoms, if present, are optionally quaternized. The heterocycloalkyl radical is partially or fully saturated. The heterocycloalkyl may be attached to the rest of the molecule through any atom of the ring(s). Examples of such heterocycloalkyl radicals include, but are not limited to, dioxolanyl, thienyl[1,3]dithianyl, decahydroisoquinolyl, imidazolinyl, imidazolidinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, oxazolidinyl, piperidinyl, piperazinyl, 4-piperidonyl, pyrrolidinyl, pyrazolidinyl, quinuclidinyl, thiazolidinyl, tetrahydrofuryl, trithianyl, tetrahydropyranyl, thiomorpholinyl, thiamorpholinyl, 1-oxo-thiomorpholinyl, and 1,1-dioxo-thiomorpholinyl. Unless stated otherwise specifically in the specification, a heterocycloalkyl moiety is optionally substituted by one or more substituents which independently are: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, hydroxy, halo, cyano, nitro, oxo, thioxo, trimethylsilanyl, —OR′, SRa, —OC(O)—Ra, —N(Ra)2, —C(O)Ra, —C(O)ORa, —C(O)N(Ra)2, —N(Ra)C(O)ORa, —N(Ra)C(O)Ra, —N(Ra)S(O)tORa (where t is 1 or 2), —S(O)tORa (where t is 1 or 2), —S(O)tN(Ra)2 (where t is 1 or 2), or PO3(Ra)2, where each Ra is independently hydrogen, alkyl, fluoroalkyl, carbocyclyl, carbocyclylalkyl, aryl, aralkyl, heterocycloalkyl, heteroaryl or heteroarylalkyl.
  • “Heterocycloalkyl” also includes bicyclic ring systems wherein one non-aromatic ring, usually with 3 to 7 ring atoms, contains at least 2 carbon atoms in addition to 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen, as well as combinations comprising at least one of the foregoing heteroatoms; and the other ring, usually with 3 to 7 ring atoms, optionally contains 1-3 heteroatoms independently selected from oxygen, sulfur, and nitrogen and is not aromatic.
  • The terms “heterocyclylalkyl”, “heterocyclyl-alkyl”, “hetcyclylalkyl”, and “hetcyclyl-alkyl” are used to describe a group wherein the alkyl chain can be branched or straight chain forming a linking portion of the heterocyclylalkyl moiety with the terminal heterocyclyl portion, as defined above, for example 3-piperidinylmethyl and the like. The term “heterocycloalkylene” refers to the divalent derivative of heterocycloalkyl.
  • The term “C1-10alkyl-heterocycyl” refers to a group as defined above where the alkyl moiety contains 1 to 10 carbon atoms. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocycyl-C1-10alkyl” refers to a group containing a terminal heterocyclic group attached to a linking alkyl group which contains 1 to 10 carbons and is branched or straight chain, such as, for example, 4-morpholinyl ethyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “heterocyclylalkenyl”, “heterocyclyl-alkenyl”, “hetcyclylalkenyl” and “hetcyclyl-alkenyl” are used to describe a group wherein the alkenyl chain can be branched or straight chain forming a linking portion of the heterocyclylalkenyl moiety with the terminal heterocyclyl portion, as defined above, for example 2-morpholinyl-1-propenyl and the like. The term “heterocycloalkenylene” refers to the divalent derivative of heterocyclylalkenyl. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocycyl-C2-10 alkenyl” refers to a group as defined above where the alkenyl group contains 2 to 10 carbon atoms and is branched or straight chain, such as, for example, 4-(N-piperazinyl)-but-2-en-1-yl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “heterocyclylalkynyl”, “heterocyclyl-alkynyl”, “hetcyclylalkynyl” and “hetcyclyl-alkynyl” are used to describe a group wherein the alkynyl chain can be branched or straight chain forming a linking portion of the heterocyclylalkynyl moiety with the terminal heterocyclyl portion, as defined above, for example 2-pyrrolidinyl-1-butynyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocycyl-C2-10 alkynyl” refers to a group as defined above where the alkynyl group contains 2 to 10 carbon atoms and is branched or straight chain, such as, for example, 4-(N-piperazinyl)-but-2-yn-1-yl, and the like.
  • The term “aryl-heterocycyl” refers to a group containing a terminal aryl group attached to a linking heterocyclic group, such as for example, N4-(4-phenyl)-piperazinyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heteroaryl-heterocycyl” refers to a group containing a terminal heteroaryl group attached to a linking heterocyclic group, such as for example, N4-(4-pyridyl)-piperazinyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “carboxylalkyl” refers to a terminal carboxyl (—COOH) group attached to branched or straight chain alkyl groups as defined above.
  • The term “carboxylalkenyl” refers to a terminal carboxyl (—COOH) group attached to branched or straight chain alkenyl groups as defined above.
  • The term “carboxylalkynyl” refers to a terminal carboxyl (—COOH) group attached to branched or straight chain alkynyl groups as defined above.
  • The term “carboxylcycloalkyl” refers to a terminal carboxyl (—COOH) group attached to a cyclic aliphatic ring structure as defined above.
  • The term “carboxylcycloalkenyl” refers to a terminal carboxyl (—COOH) group attached to a cyclic aliphatic ring structure having ethylenic bonds as defined above.
  • The terms “cycloalkylalkyl” and “cycloalkyl-alkyl” refer to a terminal cycloalkyl group as defined above attached to an alkyl group, for example cyclopropylmethyl, cyclohexylethyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “cycloalkylalkenyl” and “cycloalkyl-alkenyl” refer to a terminal cycloalkyl group as defined above attached to an alkenyl group, for example cyclohexylvinyl, cycloheptylallyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “cycloalkylalkynyl” and “cycloalkyl-alkynyl” refer to a terminal cycloalkyl group as defined above attached to an alkynyl group, for example cyclopropylpropargyl, 4-cyclopentyl-2-butynyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “cycloalkenylalkyl” and “cycloalkenyl-alkyl” refer to a terminal cycloalkenyl group as defined above attached to an alkyl group, for example 2-(cyclopenten-1-yl)ethyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The terms “cycloalkenylalkenyl” and “cycloalkenyl-alkenyl” refer to terminal a cycloalkenyl group as defined above attached to an alkenyl group, for example 1-(cyclohexen-3-yl)allyl and the like.
  • The terms “cycloalkenylalkynyl” and “cycloalkenyl-alkynyl” refer to terminal a cycloalkenyl group as defined above attached to an alkynyl group, for example 1-(cyclohexen-3-yl)propargyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “alkoxy” refers to the group —O-alkyl, including from 1 to 8 carbon atoms of a straight, branched, cyclic configuration and combinations thereof attached to the parent structure through an oxygen. Examples include methoxy, ethoxy, propoxy, isopropoxy, cyclopropyloxy, cyclohexyloxy and the like. “Lower alkoxy” refers to alkoxy groups containing one to six carbons. In some embodiments, C1-C4 alkyl, is an alkyl group which encompasses both straight and branched chain alkyls of from 1 to 4 carbon atoms.
  • The term “haloalkoxy” refers to an alkoxy group substituted with one or more halo groups, for example chloromethoxy, trifluoromethoxy, difluoromethoxy, perfluoroisobutoxy, and the like.
  • The term “alkoxyalkoxyalkyl” refers to an alkyl group substituted with an alkoxy moiety which is in turn is substituted with a second alkoxy moiety, for example methoxymethoxymethyl, isopropoxymethoxyethyl, and the like. This moiety is substituted with further substituents or not substituted with other substituents.
  • The term “alkylthio” includes both branched and straight chain alkyl groups attached to a linking sulfur atom, for example methylthio and the like.
  • The term “alkoxyalkyl” refers to an alkyl group substituted with an alkoxy group, for example isopropoxymethyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “alkoxyalkenyl” refers to an alkenyl group substituted with an alkoxy group, for example 3-methoxyallyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “alkoxyalkynyl” refers to an alkynyl group substituted with an alkoxy group, for example 3-methoxypropargyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C2-10alkenylC3-8cycloalkyl” refers to an alkenyl group as defined above substituted with a three to eight membered cycloalkyl group, for example, 4-(cyclopropyl)-2-butenyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “C2-10alkynylC3-8cycloalkyl” refers to an alkynyl group as defined above substituted with a three to eight membered cycloalkyl group, for example, 4-(cyclopropyl)-2-butynyl and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocyclyl-C1-10alkyl” refers to a heterocyclic group as defined above substituted with an alkyl group as defined above having 1 to 10 carbons, for example, 4-(N-methyl)-piperazinyl, and the like. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocyclyl-C2-10alkenyl” refers to a heterocyclic group as defined above, substituted with an alkenyl group as defined above, having 2 to 10 carbons, for example, 4-(N-allyl) piperazinyl, and the like. Moieties wherein the heterocyclic group is substituted on a carbon atom with an alkenyl group are also included. Either portion of the moiety is unsubstituted or substituted.
  • The term “heterocyclyl-C2-10alkynyl” refers to a heterocyclic group as defined above, substituted with an alkynyl group as defined above, having 2 to 10 carbons, for example, 4-(N-propargyl) piperazinyl, and the like. Moieties wherein the heterocyclic group is substituted on a carbon atom with an alkenyl group are also included. Either portion of the moiety is unsubstituted or substituted.
  • The term “oxo” refers to an oxygen that is double bonded to a carbon atom. One in the art understands that an “oxo” requires a second bond from the atom to which the oxo is attached. Accordingly, it is understood that oxo cannot be substituted onto an aryl or heteroaryl ring, unless it forms part of the aromatic system as a tautomer.
  • The term “oligomer” refers to a low-molecular weight polymer, whose number average molecular weight is typically less than about 5000 g/mol, and whose degree of polymerization (average number of monomer units per chain) is greater than one and typically equal to or less than about 50.
  • “Sulfonamidyl” or “sulfonamido” refers to a —S(═O)2—NR′R′ radical, where each R′ is selected independently from the group consisting of hydrogen, alkyl, cycloalkyl, aryl, heteroaryl (bonded through a ring carbon) and heteroalicyclic (bonded through a ring carbon). The R′ groups in NR′R′ of the —S(═O)2—NR′R′ radical may be taken together with the nitrogen to which it is attached to form a 4-, 5-, 6-, or 7-membered ring. A sulfonamido group is optionally substituted by one or more of the substituents described for alkyl, cycloalkyl, aryl, heteroaryl respectively.
  • Compounds described can contain one or more asymmetric centers and may thus give rise to diastereomers and optical isomers. The present invention includes all such possible diastereomers as well as their racemic mixtures, their substantially pure resolved enantiomers, all possible geometric isomers, and pharmaceutically acceptable salts thereof. Compounds may be shown without a definitive stereochemistry at certain positions. The present invention includes all stereoisomers of the disclosed compounds and pharmaceutically acceptable salts thereof. Further, mixtures of stereoisomers as well as isolated specific stereoisomers are also included. During the course of the synthetic procedures used to prepare such compounds, or in using racemization or epimerization procedures known to those skilled in the art, the products of such procedures can be a mixture of stereoisomers.
  • The present invention includes all manner of rotamers and conformationally restricted states of an inhibitor of the invention.
  • Substituents for alkyl, heteroalkyl, cycloalkyl, heterocycloalkyl monovalent and divalent derivative radicals (including those groups often referred to as alkylene, alkenyl, heteroalkylene, heteroalkenyl, alkynyl, cycloalkyl, heterocycloalkyl, cycloalkenyl, and heterocycloalkenyl) can be one or more of a variety of groups selected from, but not limited to: alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, —OR′, ═O, ═NR7—, ═N—OR′, —NR′R″, —SR′, -halogen, —SiR′R″R′″, —OC(O)R′, —C(O)R′, —CO2R′, —C(O)NR′R″, —OC(O)NR′R″, —NR″C(O)R′, —NR7—C(O)NR″R′″, —NR″C(O)OR′, —NR—C(NR′R″)═NR′″, —S(O)R′, —S(O)2R′, —S(O)2NR′R″, —NRSO2R′, —CN and NO2 in a number ranging from zero to (2m′+1), where m′ is the total number of carbon atoms in such radical. R′, R″, R′″ and R′″ each preferably independently refer to hydrogen, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl (e.g., aryl substituted with 1-3 halogens), substituted or unsubstituted alkyl, alkoxy or thioalkoxy groups, or arylalkyl groups. When an inhibitor of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R′, R″, R′″ and R′″ groups when more than one of these groups is present.
  • When R′ and R″ or R″ and R′″ are attached to the same nitrogen atom, they can be combined with the nitrogen atom to form a 4-, 5-, 6-, or 7-membered ring. For example, —NR′R″ is meant to include, but not be limited to, 1-pyrrolidinyl, 4 piperazinyl, and 4-morpholinyl. From the above discussion of substituents, one of skill in the art will understand that the term “alkyl” is meant to include groups including carbon atoms bound to groups other than hydrogen groups, such as haloalkyl (e.g., —CF3 and CH2CF3) and acyl (e.g., —C(O)CH3, —C(O)CF3, —C(O)CH2OCH3, and the like).
  • Similar to the substituents described for alkyl radicals above, exemplary substituents for aryl and heteroaryl groups (as well as their divalent derivatives) are varied and are selected from, for example: halogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, —OR′, —NR′R″, —SR′, -halogen, —SiR′R″R′″, —OC(O)R′, —C(O)R′, —CO2R′, —C(O)NR′R″, —OC(O)NR′R″, —NR″C(O)R′, —NR7—C(O)NR″R′″, —NR″C(O)OR′, —NR—C(NR′R″R′″)═NR′″, —NR—C(NR′R″)═NR′″, —S(O)R′, —S(O)2R′, —S(O)2NR′R″, —NRSO2R′, —CN and NO2, —R′, —N3, —CH(Ph)2, fluoro(C1-C4)alkoxo, and fluoro(C1-C4)alkyl, in a number ranging from zero to the total number of open valences on aromatic ring system; and where R′, R″, R″ and R′″ are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl and substituted or unsubstituted heteroaryl. When an inhibitor of the invention includes more than one R group, for example, each of the R groups is independently selected as are each R′, R″, R′″ and R′″ groups when more than one of these groups is present.
  • As used herein, 0-2 in the context of —S(O)(0-2)— are integers of 0, 1, and 2.
  • Two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally form a ring of the formula -T-C(O)—(CRR′)q—U—, wherein T and U are independently —NR—, —O—, —CRR′— or a single bond, and q is an integer of from 0 to 3. Alternatively, two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula -A-(CH2), —B—, wherein A and B are independently CRR′—, —O—, —NR—, —S—, —S(O)—, —S(O)2—, —S(O)2NR′— or a single bond, and r is an integer of from 1 to 4. One of the single bonds of the new ring so formed may optionally be replaced with a double bond. Alternatively, two of the substituents on adjacent atoms of aryl or heteroaryl ring may optionally be replaced with a substituent of the formula —(CRR′)s—X′—(C″R′″)d—, where s and d are independently integers of from 0 to 3, and X′ is —O—, —NR7—, —S—, —S(O)—, —S(O)2—, or —S(O)2NR′—. The substituents R, R′, R″ and R′″ are preferably independently selected from hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, and substituted or unsubstituted heteroaryl.
  • Unless otherwise stated, structures depicted herein are also meant to include compounds which differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of a hydrogen by a deuterium or tritium, or the replacement of a carbon by 13C- or 14C-enriched carbon are within the scope of this invention.
  • The compounds of the present invention may also contain unnatural proportions of atomic isotopes at one or more of atoms that constitute such compounds. For example, the compounds may be radiolabeled with radioactive isotopes, such as for example tritium (3H), iodine-125 (125I) or carbon-14 (14C). All isotopic variations of the compounds of the present invention, whether radioactive or not, are encompassed within the scope of the present invention.
  • Methods
  • In one aspect, the present invention provides a method for treating a disease condition associated with PI3-kinase α and/or mTOR in a subject. The method typically comprises administering to a subject simultaneously or sequentially a therapeutically effective amount of a combination of a PI3-kinase α inhibitor and an mTOR inhibitor.
  • As used herein, a therapeutically effective amount of a combination of a PI3-kinase α inhibitor and an mTOR inhibitor refers to a combination of a PI3-kinase α inhibitor and an mTOR inhibitor, wherein the combination is sufficient to effect the intended application including but not limited to disease treatment, as defined herein. Encompassed in this subject method is the use of therapeutically effective amount of a PI3-kinase α inhibitor and/or an mTOR inhibitor in combination to effect such treatment. Also contemplated in the subject methods is the use of a sub-therapeutic amount of a PI3-kinase α inhibitor and/or an mTOR inhibitor in the combination for treating an intended disease condition. The individual inhibitors, though present in sub-therapeutic amounts, synergistically yield an efficacious effect and/or reduced a side effect in an intended application.
  • Accordingly, in a separate but related aspect, the present invention provides for a method for treating a disease condition associated with PI3-kinase α and/or mTOR in a subject, comprising administering to the subject simultaneously or sequentially a synergistically effective therapeutic amount of a combination of a PI3-kinase α inhibitor and an mTOR inhibitor.
  • The therapeutically effective amount of the subject combination of compounds may vary depending upon the intended application (in vitro or in vivo), or the subject and disease condition being treated, e.g., the weight and age of the subject, the severity of the disease condition, the manner of administration and the like, which can readily be determined by one of ordinary skill in the art. The term also applies to a dose that will induce a particular response in target cells, e.g., reduction of proliferation or downregulation of activity of a target protein. The specific dose will vary depending on the particular compounds chosen, the dosing regimen to be followed, whether it is administered in combination with other compounds, timing of administration, the tissue to which it is administered, and the physical delivery system in which it is carried.
  • The PI3-kinase α inhibitor utilized in the subject methods typically exhibits selective inhibition of PI3-kinase α relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) including, e.g., PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • Selective inhibition of PI3-kinase α can be ascertained by an in vitro or an in vivo method. Any assay known in the art may be used, including without limitation, immunoassays, immunoprecipitation, fluorescence or cell-based assays. In some embodiments, an in vitro assay is used to determine selective inhibition of PI3-kinase α by an assay which measures the activity of the PI3Kα protein relative to the activity of another PI3-kinase such as PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. For example, a time resolved FRET assay that indirectly measures PIP3 product formed by the activity of a PI3-K may be used to determine an IC50 value for a test compound for PI3-kinase α and/or any of the other PI3-kinases.
  • As used herein, the term “IC50” refers to the half maximal inhibitory concentration of an inhibitor in inhibiting biological or biochemical function. This quantitative measure indicates how much of a particular inhibitor is needed to inhibit a given biological process (or component of a process, i.e. an enzyme, cell, cell receptor or microorganism) by half. In other words, it is the half maximal (50%) inhibitory concentration (IC) of a substance (50% IC, or IC50). EC50 refers to the plasma concentration required for obtaining 50% of a maximum effect in vivo.
  • Determination of IC50 can be made by determining and constructing a dose-response curve and examining the effect of different concentrations of an inhibitor on reversing agonist activity. In vitro assays that are useful in making these determinations are referred to as “in vitro kinase assays.”
  • In some embodiments, an in vitro kinase assay includes the use of labeled ATP as phosphodonor, and following the kinase reaction the substrate peptide is captured on an appropriate filter. Unreacted labeled ATP and metabolites are resolved from the radioactive peptide substrate by various techniques, involving trichloroacetic acid precipitation and extensive washing. Addition of several positively charged residues allows capture on phosphocellulose paper followed by washing. Radioactivity incorporated into the substrate peptide is detected by scintillation counting. This assay is relatively simple, reasonably sensitive, and the peptide substrate can be adjusted both in terms of sequence and concentration to meet the assay requirements.
  • Other exemplary kinase assays are detailed in U.S. Pat. No. 5,759,787 and U.S. application Ser. No. 12/728,926, both of which are incorporated herein by reference.
  • In other embodiments, a cell-based assay is used to ascertain selective inhibition of PI3-kinase α. For example, an inhibitor can be shown to be selective for PI3-kinase α if it selectively downregulates PI3-kinase signal transduction in cells that express PI3-kinase α, preferably in cells that exhibit abnormally high level or activity of PI3-kinase α. A variety of cells having PI3-kinase α mutations and hence exhibiting such PI3-kinase α abnormalities are known in the art. Non-limiting examples of cell lines harboring such mutations include those that carry point mutations, deletions, substitutions, or translation of nucleic acid sequence of the PI3-kinase α gene. Examples of such cell lines include but are not limited to BT20 (H1047R mutation), MCF-7 (E545K mutation), MDA-MB-361 (E545K mutation), MDA-MB-453 (H1047R mutation), T47D (H1047R mutation), Hec-1A (G1049R mutation) and HCT-116 (H1047R mutation). Other cell lines having mutations in the PI3Kα protein may be used, such as cells harboring mutations in the p85, C2, helical or kinase domains.
  • In addition, inhibition of PI3-kinase α activity can be determined by a reduction in signal transduction of the PI3-kinase α pathway. A wide variety of readouts can be utilized to establish a reduction of the output of such signaling pathway. Some non-limiting exemplary readouts include (1) a decrease in phosphorylation of Akt at residues, including but not limited to S473 and T308; (2) a decrease in activation of Akt as evidenced by a reduction of phosphorylation of Akt substrates including but not limited to FoxO1/O3a T24/32, GSK3α/β S21/9, and TSC2 T1462; (3) a decrease in phosphorylation of signaling molecules downstream of PI3-kinase α, including but not limited to ribosomal S6 S240/244, 70S6K T389, and 4EBP1 T37/46; (4) inhibition of proliferation of cells including but not limited to normal or neoplastic cells, mouse embryonic fibroblasts, leukemic blast cells, cancer stem cells, and cells that mediate autoimmune reactions; (5) induction of apoptosis of cells or cell cycle arrest; (6) reduction of cell chemotaxis; and (7) an increase in binding of 4EBP1 to eIF4E. The term “eIF4E” refers to a 24-1(1) eukaryotic translation initiation factor involved in directing ribosomes to the cap structure of mRNAs, having human gene locus 4q21-q25.
  • In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α relative to one, two or three other type I phosphatidylinositol 3-kinases (PI3-kinases) consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In other embodiments, some of the subject inhibitors selectively inhibit PI3-kinase α and PI3-kinase γ as compared to the rest of the type I PI3-kinases. In yet other embodiments, some of the subject inhibitors selectively inhibit PI3-kinase α and PI3-kinase β as compared to the rest of the type I PI3-kinases. In still yet other embodiments, some of the subject inhibitors selectively inhibit PI3-kinase α and PI3-kinase 6 as compared to the rest of the type I PI3-kinases.
  • In some embodiments, the subject methods utilizes a PI3-kinase α inhibitor with an IC50 value of about or less than a predetermined value, as ascertained in an in vitro kinase assay. In some embodiments, the PI3-kinase α inhibitor inhibits PI3-kinase α with an IC50 value of about 1 nM or less, 2 nM or less, 5 nM or less, 7 nM or less, 10 nM or less, 20 nM or less, 30 nM or less, 40 nM or less, 50 nM or less, 60 nM or less, 70 nM or less, 80 nM or less, 90 nM or less, 100 nM or less, 120 nM or less, 140 nM or less, 150 nM or less, 160 nM or less, 170 nM or less, 180 nM or less, 190 nM or less, 200 nM or less, 225 nM or less, 250 nM or less, 275 nM or less, 300 nM or less, 325 nM or less, 350 nM or less, 375 nM or less, 400 nM or less, 425 nM or less, 450 nM or less, 475 nM or less, 500 nM or less, 550 nM or less, 600 nM or less, 650 nM or less, 700 nM or less, 750 nM or less, 800 nM or less, 850 nM or less, 900 nM or less, 950 nM or less, 1 μM or less, 1.2 μM or less, 1.3 μM or less, 1.4 μM or less, 1.5 μM or less, 1.6 μM or less, 1.7 μM or less, 1.8 μM or less, 1.9 μM or less, 2 μM or less, 5 μM or less, 10 μM or less, 15 μM or less, 20 μM or less, 25 μM or less, 30 μM or less, 40 μM or less, 50 μM, 60 μM, 70 μM, 80 μM, 90 μM, 100 μM, 200 μM, 300 μM, 400 μM, or 500 μM, or less.
  • In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, or 1000 times less than its IC50 value against one, two, or three other type I PI3-kinase(s) selected from the group consisting of PI3-kinase PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is less than about 1 nM, 2 nM, 5 nM, 7 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 120 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 225 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM, 950 nM, 1 μM, 1.2 μM, 1.3 μM, 1.4 μM, 1.5 μM, 1.6 μM, 1.7 μM, 1.8 μM, 1.9 μM, 2 μM, 5 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM, 40 μM, 50 μM, 60 μM, 70 μM, 80 μM, 90 μM, 100 μM, 200 μM, 300 μM, 400 μM, or 500 μM, and said IC50 value is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, or 1000 times less than its IC50 value against one, two or three other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In some embodiments, the PI3-kinase α inhibitor inhibits PI3-kinase α with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay.
  • In some instances, the PI3-kinase α inhibitor inhibits PI3-kinase α with an IC50 value of about 200 nM or less as ascertained in an in vitro kinase assay and the IC50 value is at least 5, 10, 15, 20, 25, 50, 100, or 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is less than about 100 nM, and said IC50 value is at 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • In some instances, the PI3-kinase α inhibitor inhibits PI3-kinase α with an IC50 value of about 50 nM or less as ascertained in an in vitro kinase assay and the IC50 value is at least 5, 10, 15, 20, 25, 50, 100, or 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is less than about 20 nM, and said IC50 value is at 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an IC50 value that is less than about 20 nM, and said IC50 value is at 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • In some instances, the PI3-kinase α inhibitor inhibits PI3-kinase α with an IC50 value of about 20 nM or less as ascertained in an in vitro kinase assay and the IC50 value is at least 100 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • Alternatively, the PI3-kinase α inhibitor inhibits PI3-kinase α with an EC50 value of about 10 μM or less, 5 μM or less, 2.5 μM or less, 1 μM or less, 500 nM or less, 100 nM or less, 75 nM or less, 50 nM or less, 25 nM or less, 10 nM or less, 5 nM or less, 1 nM or less, 500 μM or less, or 100 μM or less as ascertained in an in vitro kinase assay.
  • In some embodiments, the PI3-kinase α inhibitor selectively inhibits PI3-kinase α with an EC50 value that is at least 5, 10, 15, 20, 25, 50, 100, or 1000 times less than its EC50 value against one, two or there other type I PI3-kinases selected from the group consisting of PI3-kinase PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the PI3-kinase α inhibitor inhibits PI3-kinase α with an EC50 value of about 10 μM or less, 5 μM or less, 2.5 μM or less, 1 μM or less, 500 nM or less, 100 nM or less, 75 nM or less, 50 nM or less, 25 nM or less, 10 nM or less, 5 nM or less, 1 nM or less, 500 pM or less, or 100 pM or less as ascertained in an in vitro kinase assay, and such EC50 value is at least 5, 10, 15, 20, 25, 50, or 100, 1000 times less than its EC50 value against one, two or three other type I PI3-kinases selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • The mTOR inhibitor utilized in the subject methods is typically highly selective for the target molecule. In one aspect, the mTOR inhibitor binds to and directly inhibits both mTORC1 and mTORC2. Such ability can be ascertained using any method known in the art or described herein. For example, inhibition of mTorC1 and/or mTorC2 activity can be determined by a reduction in signal transduction of the PI3K/Akt/mTor pathway. A wide variety of readouts can be utilized to establish a reduction of the output of such signaling pathway. Some non-limiting exemplary readouts include (1) a decrease in phosphorylation of Akt at residues, including but not limited to S473 and T308; (2) a decrease in activation of Akt as evidenced by a reduction of phosphorylation of Akt substrates including but not limited to FoxO1/O3a T24/32, GSK3α/β S21/9, and TSC2 T1462; (3) a decrease in phosphorylation of signaling molecules downstream of mTor, including but not limited to ribosomal S6 S240/244, 70S6K T389, and 4EBP1 T37/46; (4) inhibition of proliferation of cells including but not limited to normal or neoplastic cells, mouse embryonic fibroblasts, leukemic blast cells, cancer stem cells, and cells that mediate autoimmune reactions; (5) induction of apoptosis of cells or cell cycle arrest; (6) reduction of cell chemotaxis; and (7) an increase in binding of 4EBP1 to eIF4E.
  • mTor exists in two types of complexes, mTorC1 containing the raptor subunit and mTorC2 containing rictor. As known in the art, “rictor” refers to a cell growth regulatory protein having human gene locus 5p13.1. These complexes are regulated differently and have a different spectrum of substrates. For instance, mTorC1 phosphorylates S6 kinase (S6K) and 4EBP1, promoting increased translation and ribosome biogenesis to facilitate cell growth and cell cycle progression. S6K also acts in a feedback pathway to attenuate PI3K/Akt activation. Thus, inhibition of mTorC1 (e.g. by a biologically active agent as discussed herein) results in activation of 4EBP1, resulting in inhibition of (e.g. a decrease in) RNA translation.
  • mTorC2 is generally insensitive to rapamycin and selective inhibitors and is thought to modulate growth factor signaling by phosphorylating the C-terminal hydrophobic motif of some AGC kinases such as Akt. In many cellular contexts, mTorC2 is required for phosphorylation of the S473 site of Akt. Thus, mTorC1 activity is partly controlled by Akt whereas Akt itself is partly controlled by mTorC2.
  • Growth factor stimulation of PI3K causes activation of Akt by phosphorylation at the two key sites, S473 and T308. It has been reported that full activation of Akt requires phosphorylation of both S473 and T308Active. Akt promotes cell survival and proliferation in many ways including suppressing apoptosis, promoting glucose uptake, and modifying cellular metabolism. Of the two phosphorylation sites on Akt, activation loop phosphorylation at T308, mediated by PDK1, is believed to be indispensable for kinase activity, while hydrophobic motif phosphorylation at S473 enhances Akt kinase activity.
  • Selective mTor inhibition may also be determined by expression levels of the mTor genes, its downstream signaling genes (for example by RT-PCR), or expression levels of the proteins (for example by immunocytochemistry, immunohistochemistry, Western blots) as compared to other PI3-kinases or protein kinases.
  • Cell-based assays for establishing selective inhibition of mTorC1 and/or mTorC2 can take a variety of formats. This generally will depend on the biological activity and/or the signal transduction readout that is under investigation. For example, the ability of the agent to inhibit mTorC1 and/or mTorC2 to phosphorylate the downstream substrate(s) can be determined by various types of kinase assays known in the art. Representative assays include but are not limited to immunoblotting and immunoprecipitation with antibodies such as anti-phosphotyrosine, anti-phosphoserine or anti-phosphothreonine antibodies that recognize phosphorylated proteins. Alternatively, antibodies that specifically recognize a particular phosphorylated form of a kinase substrate (e.g. anti-phospho AKT S473 or anti-phospho AKT T308) can be used. In addition, kinase activity can be detected by high throughput chemiluminescent assays such as AlphaScreen™ (available from Perkin Elmer) and eTag™ assay (Chan-Hui, et al. (2003) Clinical Immunology 111: 162-174). In another aspect, single cell assays such as flow cytometry as described in the phosflow experiment can be used to measure phosphorylation of multiple downstream mTOR substrates in mixed cell populations.
  • One advantage of the immunoblotting and phosflow methods is that the phosphorylation of multiple kinase substrates can be measured simultaneously. This provides the advantage that efficacy and selectivity can be measured at the same time. For example, cells may be contacted with an mTOR inhibitor at various concentrations and the phosphorylation levels of substrates of both mTOR and other kinases can be measured. In one aspect, a large number of kinase substrates are assayed in what is termed a “comprehensive kinase survey.” Selective mTOR inhibitors are expected to inhibit phosphorylation of mTOR substrates without inhibiting phosphorylation of the substrates of other kinases. Alternatively, selective mTOR inhibitors may inhibit phosphorylation of substrates of other kinases through anticipated or unanticipated mechanisms such as feedback loops or redundancy.
  • Effect of inhibition of mTorC1 and/or mTorC2, or PI3-kinase α can be established by cell colony formation assay or other forms of cell proliferation assay. A wide range of cell proliferation assays are available in the art, and many of which are available as kits. Non-limiting examples of cell proliferation assays include testing for tritiated thymidine uptake assays, BrdU (5′-bromo-2′-deoxyuridine) uptake (kit marketed by Calibochem), MTS uptake (kit marketed by Promega), MTT uptake (kit marketed by Cayman Chemical), CyQUANT® dye uptake (marketed by Invitrogen).
  • Apoptosis and cell cycle arrest analysis can be performed with any methods exemplified herein as well other methods known in the art. Many different methods have been devised to detect apoptosis. Exemplary assays include but are not limited to the TUNEL (TdT-mediated dUTP Nick-End Labeling) analysis, ISEL (in situ end labeling), and DNA laddering analysis for the detection of fragmentation of DNA in populations of cells or in individual cells, Annexin-V analysis that measures alterations in plasma membranes, detection of apoptosis related proteins such p53 and Fas.
  • A cell-based assay typically proceeds with exposing the target cells (e.g., in a culture medium) to a test compound which is a potential mTorC1 and/or mTorC2 selective inhibitor, or a PI3-kinase inhibitor and then assaying for readout under investigation. Depending on the nature of the candidate mTor inhibitors or PI3-kinase α inhibitors, they can directly be added to the cells or in conjunction with carriers. For instance, when the agent is nucleic acid, it can be added to the cell culture by methods well known in the art, which include without limitation calcium phosphate precipitation, microinjection or electroporation. Alternatively, the nucleic acid can be incorporated into an expression or insertion vector for incorporation into the cells. Vectors that contain both a promoter and a cloning site into which a polynucleotide can be operatively linked are well known in the art. Such vectors are capable of transcribing RNA in vitro or in vitro, and are commercially available from sources such as Stratagene (La Jolla, Calif.) and Promega Biotech (Madison, Wis.). In order to optimize expression and/or in vitro transcription, it may be necessary to remove, add or alter 5′ and/or 3′ untranslated portions of the clones to eliminate extra, potential inappropriate alternative translation initiation codons or other sequences that may interfere with or reduce expression, either at the level of transcription or translation. Alternatively, consensus ribosome binding sites can be inserted immediately 5′ of the start codon to enhance expression. Examples of vectors are viruses, such as baculovirus and retrovirus, bacteriophage, adenovirus, adeno-associated virus, cosmid, plasmid, fungal vectors and other recombination vehicles typically used in the art which have been described for expression in a variety of eukaryotic and prokaryotic hosts, and may be used for gene therapy as well as for simple protein expression. Among these are several non-viral vectors, including DNA/liposome complexes, and targeted viral protein DNA complexes. To enhance delivery to a cell, the nucleic acid or proteins of this invention can be conjugated to antibodies or binding fragments thereof which bind cell surface antigens. Liposomes that also comprise a targeting antibody or fragment thereof can be used in the methods of this invention. Other biologically acceptable carriers can be utilized, including those described in, for example, REMINGTON'S PHARMACEUTICAL SCIENCES, 19th Ed. (2000), in conjunction with the subject compounds.
  • The subject agents can also be utilized to inhibit phosphorylation of both Akt (S473) and Akt (T308) in a cell. Accordingly, the present invention provides a method comprises the step of contacting a cell with an effective amount of such biologically active agent such that Akt phosphorylation at residues S473 and T308 is simultaneously inhibited. In one aspect, the biologically active agent inhibits phosphorylation of S473 of Akt more effectively than phosphorylation of T308 of Akt when tested at a comparable molar concentration, preferably at an identical molar concentration.
  • Inhibition of Akt phosphorylation can be determined using any methods known in the art or described herein. Representative assays include but are not limited to immunoblotting and immunoprecipitation with antibodies such as anti-phosphotyrosine antibodies that recognize the specific phosphorylated proteins. Cell-based ELISA kit quantifies the amount of activated (phosphorylated at S473) Akt relative to total Akt protein is also available (SuperArray Biosciences).
  • In practicing the subject methods, any cells that express PI3-kinase α, mTorC1, mTorC2 and/or Akt can be utilized. Non-limiting examples of specific cell types whose proliferation can be inhibited include fibroblast, cells of skeletal tissue (bone and cartilage), cells of epithelial tissues (e.g. liver, lung, breast, skin, bladder and kidney), cardiac and smooth muscle cells, neural cells (glia and neurones), endocrine cells (adrenal, pituitary, pancreatic islet cells), melanocytes, and many different types of haemopoietic cells (e.g., cells of B-cell or T-cell lineage, and their corresponding stem cells, lymphoblasts). Also of interest are cells exhibiting a neoplastic propensity or phenotype. Of particular interest is the type of cells that differentially expresses (over-expresses or under-expresses) a disease-causing gene. The types of diseases involving abnormal functioning of genes include but are not limited to autoimmune diseases, cancer, obesity, hypertension, diabetes, neuronal and/or muscular degenerative diseases, cardiac diseases, endocrine disorders, and any combinations thereof.
  • In some embodiments, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1 nM, 2 nM, 5 nM, 7 nM, 10 nM, 20 nM, 30 nM, 40 nM, 50 nM, 60 nM, 70 nM, 80 nM, 90 nM, 100 nM, 120 nM, 140 nM, 150 nM, 160 nM, 170 nM, 180 nM, 190 nM, 200 nM, 225 nM, 250 nM, 275 nM, 300 nM, 325 nM, 350 nM, 375 nM, 400 nM, 425 nM, 450 nM, 475 nM, 500 nM, 550 nM, 600 nM, 650 nM, 700 nM, 750 nM, 800 nM, 850 nM, 900 nM, 950 nM, 1 μM, 1.2 μM, 1.3 μM, 1.4 μM, 1.5 μM, 1.6 μM, 1.7 μM, 1.8 μM, 1.9 μM, 2 μM, 5 μM, 10 μM, 15 μM, 20 μM, 25 μM, 30 μM, 40 μM, 50 μM, 60 μM, 70 μM, 80 μM, 90 μM, 100 μM, 200 μM, 300 μM, 400 μM, or 500 μM or less as ascertained in an in vitro kinase assay, and said IC50 value is at least 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 100, or 1000 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. For example, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 200, 100, 75, 50, 25, 10, 5, 1 or 0.5 nM or less as ascertained in an in vitro kinase assay. In one instance, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay. Alternatively, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 10 nM or less as ascertained in an in vitro kinase assay.
  • In some embodiments, the present invention provides the use of an mTOR inhibitor, wherein the mTOR inhibitor directly binds to and inhibits both mTORC1 and mTORC2 with an IC50 value of about or less than a predetermined value, as ascertained in an in vitro kinase assay. In some embodiments, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1 nM or less, 2 nM or less, 5 nM or less, 7 nM or less, 10 nM or less, 20 nM or less, 30 nM or less, 40 nM or less, 50 nM or less, 60 nM or less, 70 nM or less, 80 nM or less, 90 nM or less, 100 nM or less, 120 nM or less, 140 nM or less, 150 nM or less, 160 nM or less, 170 nM or less, 180 nM or less, 190 nM or less, 200 nM or less, 225 nM or less, 250 nM or less, 275 nM or less, 300 nM or less, 325 nM or less, 350 nM or less, 375 nM or less, 400 nM or less, 425 nM or less, 450 nM or less, 475 nM or less, 500 nM or less, 550 nM or less, 600 nM or less, 650 nM or less, 700 nM or less, 750 nM or less, 800 nM or less, 850 nM or less, 900 nM or less, 950 nM or less, 1 μM or less, 1.2 μM or less, 1.3 μM or less, 1.4 μM or less, 1.5 μM or less, 1.6 μM or less, 1.7 μM or less, 1.8 μM or less, 1.9 μM or less, 2 μM or less, 5 μM or less, 10 μM or less, 15 μM or less, 20 μM or less, 25 μM or less, 30 μM or less, 40 μM or less, 50 μM or less, 60 μM or less, 70 μM or less, 80 μM or less, 90 μM or less, 100 μM or less, 200 μM or less, 300 μM or less, 400 μM or less, or 500 μM or less.
  • In some embodiments, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1 nM or less, 2 nM or less, 5 nM or less, 7 nM or less, 10 nM or less, 20 nM or less, 30 nM or less, 40 nM or less, 50 nM or less, 60 nM or less, 70 nM or less, 80 nM or less, 90 nM or less, 100 nM or less, 120 nM or less, 140 nM or less, 150 nM or less, 160 nM or less, 170 nM or less, 180 nM or less, 190 nM or less, 200 nM or less, 225 nM or less, 250 nM or less, 275 nM or less, 300 nM or less, 325 nM or less, 350 nM or less, 375 nM or less, 400 nM or less, 425 nM or less, 450 nM or less, 475 nM or less, 500 nM or less, 550 nM or less, 600 nM or less, 650 nM or less, 700 nM or less, 750 nM or less, 800 nM or less, 850 nM or less, 900 nM or less, 950 nM or less, 1 μM or less, 1.2 μM or less, 1.3 μM or less, 1.4 μM or less, 1.5 μM or less, 1.6 μM or less, 1.7 μM or less, 1.8 μM or less, 1.9 μM or less, 2 μM or less, 5 μM or less, 10 μM or less, 15 μM or less, 20 μM or less, 25 μM or less, 30 μM or less, 40 μM or less, 50 μM or less, 60 μM or less, 70 μM or less, 80 μM or less, 90 μM or less, 100 μM or less, 200 μM or less, 300 μM or less, 400 μM or less, or 500 μM or less, and the mTOR inhibitor is substantially inactive against one or more types I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. In some embodiments, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 10 nM or less as ascertained in an in vitro kinase assay, and the mTOR inhibitor is substantially inactive against one or more types I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • As used herein, the terms “substantially inactive” refers to an inhibitor that inhibits the activity of its target by less than approximately 1%, 5%, 10%, 15% or 20% of its maximal activity in the absence of the inhibitor, as determined by an in vitro enzymatic assay (e.g. in vitro kinase assay).
  • In other embodiments, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 1000, 500, 100, 75, 50, 25, 10, 5, 1, or 0.5 nM or less as ascertained in an in vitro kinase assay, and said IC50 value is at least 2, 5, 10, 15, 20, 50, 100 or 100 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ. For example, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay, and said IC50 value is at least 5 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the mTOR inhibitor inhibits both mTORC1 and mTORC2 with an IC50 value of about 100 nM or less as ascertained in an in vitro kinase assay, and said IC50 value is at least 5 times less than its IC50 value against all other type I PI3-kinases selected from the group consisting of PI3-kinase α, PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • In some embodiments, the mTOR inhibitor utilized in the subject methods inhibits one of mTORC1 and mTORC2 selectively with an IC50 value of about 1000, 500, 100, 75, 50, 25, 10, 5, 1, or 0.5 nM or less as ascertained in an in vitro kinase. For example, the mTOR inhibitor utilized in the subject methods inhibits mTORC1 selectively with an IC50 value of about 1000, 500, 100, 75, 50, 25, 10, 5, 1, or 0.5 nM or less as ascertained in an in vitro kinase. For example, rapamycin and rapamycin derivatives or analogues have been shown to primarily inhibit mTORC1 and not mTORC2. Suitable mTORC1 inhibitors compounds include, for example, sirolimus (rapamycin), deforolimus (AP23573, MK-8669), everolimus (RAD-001), temsirolimus (CCI-779), zotarolimus (ABT-578), and biolimus A9 (umirolimus).
  • PI3-kinase α inhibitors or mTOR inhibitors suitable for use in the subject methods can be selected from a variety types of molecules. For example, an inhibitor can be biological or chemical compound such as a simple or complex organic or inorganic molecule, peptide, peptide mimetic, protein (e.g. antibody), liposome, or a polynucleotide (e.g. small interfering RNA, microRNA, anti-sense, aptamer, ribozyme, or triple helix). Some exemplary classes of chemical compounds suitable for use in the subject methods are detailed in the sections below.
  • The advantages of selective inhibition of a cellular target as a way of treating a disease condition mediated by such target are manifold. Because healthy cells depend on the signaling pathways that are activated in cancers for survival, inhibition of these pathways during cancer treatment can cause harmful side effects. In order for a method of treating cancer to be successful without causing excessive damage to healthy cells, a very high degree of specificity in targeting the aberrant signaling component or components is desirable. Moreover, cancer cells may depend on overactive signaling for their survival (known as the oncogene addiction hypothesis). In this way, cancer cells are frequently observed to adapt to drug inhibition of an aberrant signaling component by selecting for mutations in the same pathway that overcome the effect of the drug. Therefore, cancer therapies may be more successful in overcoming the problem of drug resistance if they target a signaling pathway as a whole, or target more than one component within a signaling pathway.
  • Without being bound by theory, selective inhibition of PI3-kinase α provides a more targeted treatment to a disease condition mediated by PI3-kinase without disrupting one or more pathways that are implicated by one or more other type I phosphatidylinositol 3-kinases, namely PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • Some signaling pathways that contain PI3K and mTOR are illustrated in FIG. 1. One major downstream effector of PI3K and mTOR signaling is the Akt serine/threonine kinase. Akt possesses a protein domain known as a PH domain, or Pleckstrin Homology domain, which binds to phosphoinositides with high affinity. In the case of the PH domain of Akt, it binds either PIP3 (phosphatidylinositol (3,4,5)-trisphosphate, PtdIns(3,4,5)P3) or PIP2 (phosphatidylinositol (3,4)-bisphosphate, PtdIns(3,4)P2). PI3K phosphorylates PIP2 in response to signals from chemical messengers, such as ligand binding to G protein-coupled receptors or receptor tyrosine kinases. Phosphorylation by PI3K converts PIP2 to PIP3, recruiting Akt to the cell membrane where it is phosphorylated at serine 473 (S473) by mTORC2. Phosphorylation of Akt at another site, threonine 308 (T308), is not directly dependent on mTORC2, but requires PI3K activity. Therefore, PI3K activity towards Akt can be isolated from mTOR activity by examining Akt threonine 308 phosphorylation status in cells lacking mTORC2 activity.
  • The subject methods are useful for treating a disease condition associated with PI3-kinase α and/or mTOR. Any disease condition that results directly or indirectly from an abnormal activity or expression level of PI3-kinase α and/or mTOR can be an intended disease condition.
  • A vast diversity of disease conditions associated with PI3-kinase α and/or mTOR have been reported. PI3-kinase α has been implicated, for example, in a variety of human cancers. Angiogenesis has been shown to selectively require the α isoform of PI3K in the control of endothelial cell migration. (Graupera et al, Nature 2008; 453; 662-6). Mutations in the gene coding for PI3K a or mutations which lead to upregulation of PI3K a are believed to occur in many human cancers such as lung, stomach, endometrial, ovarian, bladder, breast, colon, brain and skin cancers. Often, mutations in the gene coding for PI3K a are point mutations clustered within several hotspots in helical and kinase domains, such as E542K, E545K, and H1047R. Many of these mutations have been shown to be oncogenic gain-of-function mutations. Because of the high rate of PI3K a mutations, targeting of this pathway provides valuable therapeutic opportunities. While other PI3K isoforms such as PI3K δ or PI3K γ are expressed primarily in hematopoietic cells, PI3K a, along with PI3K (3, is expressed constitutively.
  • Disease conditions associated with PI3-kinase α and/or mTOR can also be characterized by abnormally high level of activity and/or expression of downstream messengers of PI3-kinase α. For example, proteins or messengers such as PIP2, PIP3, PDK, Akt, PTEN, PRAS40, GSK-313, p21, p27 may be present in abnormal amounts which can be identified by any assays known in the art.
  • Deregulation of the mTOR pathway is emerging as a common theme in diverse human diseases and as a consequence drugs that target mTOR have therapeutic value. The diseases associated with deregulation of mTORC1 include, but are not limited to, tuberous sclerosis complex (TSC) and lymphangioleiomyomatosis (LAM), both of which are caused by mutations in TSC1 or TSC2 tumor suppressors. Patients with TSC develop benign tumors that when present in brain, however, can cause seizures, mental retardation and death. LAM is a serious lung disease Inhibition of mTORC1 may help patients with Peutz-Jeghers cancer-prone syndrome caused by the LKB 1 mutation. mTORC1 may also have role in the genesis of sporadic cancers. Inactivation of several tumor suppressors, in particular PTEN, p53, VHL and NF1, has been linked to mTORC1 activation. Rapamycin and its analogues (eg CCI-779, RAD001 and AP23573) inhibit TORC1 and have shown moderate anti-cancer activity in phase II clinical trials. However, due to the negative signal from S6K1 to the insulin/PI3K/Akt pathway, it is important to note that inhibitors of mTORC1, like rapalogs, can activate PKB/Akt. If this effect persists with chronic rapamycin treatment, it may provide cancer cells with an increased survival signal that may be clinically undesirable. The PI3K/Akt pathway is activated in many cancers. Activated Akt regulates cell survival, cell proliferation and metabolism by phosphorylating proteins such as BAD, FOXO, NF-KB, p21Cip1, p27Kip1, GSK3β and others. Akt might also promote cell growth by phosphorylating TSC2. Akt activation probably promotes cellular transformation and resistance to apoptosis by collectively promoting growth, proliferation and survival, while inhibiting apoptotic pathways. The combination of an inhibitor of mTORC1 and mTORC2 and a PI3-kinase α inhibitor is beneficial for treatment of tumors with elevated Akt phosphorylation, and should down-regulate cell growth, cell survival and cell proliferation.
  • Where desired, the subject to be treated is tested prior to treatment using a diagnostic assay to determine the sensitivity of tumor cells to a PI3Kα kinase inhibitor. Any method known in the art that can determine the sensitivity of the tumor cells of a subject to a PI3Kα kinase inhibitor can be employed. Where the subject is tested prior to treatment using a diagnostic assay to determine the sensitivity of tumor cells to an PI3Kα kinase inhibitor, in one embodiment, when the subject is identified as one whose tumor cells are predicted to have low sensitivity to an PI3Kα kinase inhibitor as a single agent, are likely to display enhanced sensitivity in the presence of an mTOR inhibitor, or vice versa, when the subject is administered, simultaneously or sequentially, a therapeutically effective amount of a combination of an PI3Kα kinase inhibitor and an mTOR inhibitor. In another embodiment, when the subject is identified as one whose tumor cells are predicted to have high sensitivity to an PI3Kα kinase inhibitor as a single agent, but may also display enhanced sensitivity in the presence of an mTOR inhibitor based on the results described herein, the subject is administered, simultaneously or sequentially, a therapeutically effective amount of a combination of an PI3Kα kinase inhibitor and an mTOR inhibitor. In these methods one or more additional anti-cancer agents or treatments can be co-administered simultaneously or sequentially with the PI3Kα kinase inhibitor and mTOR inhibitor, as judged to be appropriate by the administering physician given the prediction of the likely responsiveness of the subject to the combination of PI3Kα kinase inhibitor and mTOR inhibitor, in combination with any additional circumstances pertaining to the individual subject.
  • Accordingly, in some embodiments, the present invention provides for a method comprising: (a) determining the presence in a subject of a mutation in PI3-kinase α that is associated with a disease condition mediated by PI3-kinase α; and (b) administering to said subject a pharmaceutical composition of the invention.
  • In yet another embodiment, the present invention provides for a method of inhibiting phosphorylation of both Akt (S473) and Akt (T308) in a cell, comprising contacting a cell with an effective amount of a PI3-kinase α inhibitor and an mTOR inhibitor, biologically active agent that selectively inhibits both mTORC1 and mTORC2 activity relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) as ascertained by a cell-based assay or an in vitro kinase assay, wherein the PI3-kinase α inhibitor exhibits selective inhibition of PI3-kinase α relative to one or more type I phosphatidylinositol 3-kinases (PI3-kinase) ascertained by an in vitro kinase assay, wherein the one or more type I PI3-kinase is selected from the group consisting of PI3-kinase β, PI3-kinase γ, and PI3-kinase δ.
  • The data presented in the Examples herein below demonstrate that the anti-tumor effects of a combination of an mTOR inhibitor and PI3K a inhibitor are superior to the anti-tumor effects of either inhibitor by itself, and co-administration of an mTOR inhibitor with a PI3K a inhibitor can be effective for treatment of a neoplastic condition associated with PI3-kinase α and/or mTOR. Non-limiting examples of such conditions include but are not limited to Acanthoma, Acinic cell carcinoma, Acoustic neuroma, Acral lentiginous melanoma, Acrospiroma, Acute eosinophilic leukemia, Acute lymphoblastic leukemia, Acute megakaryoblastic leukemia, Acute monocytic leukemia, Acute myeloblastic leukemia with maturation, Acute myeloid dendritic cell leukemia, Acute myeloid leukemia, Acute promyelocytic leukemia, Adamantinoma, Adenocarcinoma, Adenoid cystic carcinoma, Adenoma, Adenomatoid odontogenic tumor, Adrenocortical carcinoma, Adult T-cell leukemia, Aggressive NK-cell leukemia, AIDS-Related Cancers, AIDS-related lymphoma, Alveolar soft part sarcoma, Ameloblastic fibroma, Anal cancer, Anaplastic large cell lymphoma, Anaplastic thyroid cancer, Angioimmunoblastic T-cell lymphoma, Angiomyolipoma, Angiosarcoma, Appendix cancer, Astrocytoma, Atypical teratoid rhabdoid tumor, Basal cell carcinoma, Basal-like carcinoma, B-cell leukemia, B-cell lymphoma, Bellini duct carcinoma, Biliary tract cancer, Bladder cancer, Blastoma, Bone Cancer, Bone tumor, Brain Stem Glioma, Brain Tumor, Breast Cancer, Brenner tumor, Bronchial Tumor, Bronchioloalveolar carcinoma, Brown tumor, Burkitt's lymphoma, Cancer of Unknown Primary Site, Carcinoid Tumor, Carcinoma, Carcinoma in situ, Carcinoma of the penis, Carcinoma of Unknown Primary Site, Carcinosarcoma, Castleman's Disease, Central Nervous System Embryonal Tumor, Cerebellar Astrocytoma, Cerebral Astrocytoma, Cervical Cancer, Cholangiocarcinoma, Chondroma, Chondrosarcoma, Chordoma, Choriocarcinoma, Choroid plexus papilloma, Chronic Lymphocytic Leukemia, Chronic monocytic leukemia, Chronic myelogenous leukemia, Chronic Myeloproliferative Disorder, Chronic neutrophilic leukemia, Clear-cell tumor, Colon Cancer, Colorectal cancer, Craniopharyngioma, Cutaneous T-cell lymphoma, Degos disease, Dermatofibrosarcoma protuberans, Dermoid cyst, Desmoplastic small round cell tumor, Diffuse large B cell lymphoma, Dysembryoplastic neuroepithelial tumor, Embryonal carcinoma, Endodermal sinus tumor, Endometrial cancer, Endometrial Uterine Cancer, Endometrioid tumor, Enteropathy-associated T-cell lymphoma, Ependymoblastoma, Ependymoma, Epithelioid sarcoma, Erythroleukemia, Esophageal cancer, Esthesioneuroblastoma, Ewing Family of Tumor, Ewing Family Sarcoma, Ewing's sarcoma, Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Extramammary Paget's disease, Fallopian tube cancer, Fetus in fetu, Fibroma, Fibrosarcoma, Follicular lymphoma, Follicular thyroid cancer, Gallbladder Cancer, Gallbladder cancer, Ganglioglioma, Ganglioneuroma, Gastric Cancer, Gastric lymphoma, Gastrointestinal cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Stromal Tumor, Gastrointestinal stromal tumor, Germ cell tumor, Germinoma, Gestational choriocarcinoma, Gestational Trophoblastic Tumor, Giant cell tumor of bone, Glioblastoma multiforme, Glioma, Gliomatosis cerebri, Glomus tumor, Glucagonoma, Gonadoblastoma, Granulosa cell tumor, Hairy Cell Leukemia, Hairy cell leukemia, Head and Neck Cancer, Head and neck cancer, Heart cancer, Hemangioblastoma, Hemangiopericytoma, Hemangiosarcoma, Hematological malignancy, Hepatocellular carcinoma, Hepatosplenic T-cell lymphoma, Hereditary breast-ovarian cancer syndrome, Hodgkin Lymphoma, Hodgkin's lymphoma, Hypopharyngeal Cancer, Hypothalamic Glioma, Inflammatory breast cancer, Intraocular Melanoma, Islet cell carcinoma, Islet Cell Tumor, Juvenile myelomonocytic leukemia, Kaposi Sarcoma, Kaposi's sarcoma, Kidney Cancer, Klatskin tumor, Krukenberg tumor, Laryngeal Cancer, Laryngeal cancer, Lentigo maligna melanoma, Leukemia, Leukemia, Lip and Oral Cavity Cancer, Liposarcoma, Lung cancer, Luteoma, Lymphangioma, Lymphangiosarcoma, Lymphoepithelioma, Lymphoid leukemia, Lymphoma, Macroglobulinemia, Malignant Fibrous Histiocytoma, Malignant fibrous histiocytoma, Malignant Fibrous Histiocytoma of Bone, Malignant Glioma, Malignant Mesothelioma, Malignant peripheral nerve sheath tumor, Malignant rhabdoid tumor, Malignant triton tumor, MALT lymphoma, Mantle cell lymphoma, Mast cell leukemia, Mediastinal germ cell tumor, Mediastinal tumor, Medullary thyroid cancer, Medulloblastoma, Medulloblastoma, Medulloepithelioma, Melanoma, Melanoma, Meningioma, Merkel Cell Carcinoma, Mesothelioma, Mesothelioma, Metastatic Squamous Neck Cancer with Occult Primary, Metastatic urothelial carcinoma, Mixed Mullerian tumor, Monocytic leukemia, Mouth Cancer, Mucinous tumor, Multiple Endocrine Neoplasia Syndrome, Multiple Myeloma, Multiple myeloma, Mycosis Fungoides, Mycosis fungoides, Myelodysplastic Disease, Myelodysplastic Syndromes, Myeloid leukemia, Myeloid sarcoma, Myeloproliferative Disease, Myxoma, Nasal Cavity Cancer, Nasopharyngeal Cancer, Nasopharyngeal carcinoma, Neoplasm, Neurinoma, Neuroblastoma, Neuroblastoma, Neurofibroma, Neuroma, Nodular melanoma, Non-Hodgkin Lymphoma, Non-Hodgkin lymphoma, Nonmelanoma Skin Cancer, Non-Small Cell Lung Cancer, Ocular oncology, Oligoastrocytoma, Oligodendroglioma, Oncocytoma, Optic nerve sheath meningioma, Oral Cancer, Oral cancer, Oropharyngeal Cancer, Osteosarcoma, Osteosarcoma, Ovarian Cancer, Ovarian cancer, Ovarian Epithelial Cancer, Ovarian Germ Cell Tumor, Ovarian Low Malignant Potential Tumor, Paget's disease of the breast, Pancoast tumor, Pancreatic Cancer, Pancreatic cancer, Papillary thyroid cancer, Papillomatosis, Paraganglioma, Paranasal Sinus Cancer, Parathyroid Cancer, Penile Cancer, Perivascular epithelioid cell tumor, Pharyngeal Cancer, Pheochromocytoma, Pineal Parenchymal Tumor of Intermediate Differentiation, Pineoblastoma, Pituicytoma, Pituitary adenoma, Pituitary tumor, Plasma Cell Neoplasm, Pleuropulmonary blastoma, Polyembryoma, Precursor T-lymphoblastic lymphoma, Primary central nervous system lymphoma, Primary effusion lymphoma, Primary Hepatocellular Cancer, Primary Liver Cancer, Primary peritoneal cancer, Primitive neuroectodermal tumor, Prostate cancer, Pseudomyxoma peritonei, Rectal Cancer, Renal cell carcinoma, Respiratory Tract Carcinoma Involving the NUT Gene on Chromosome 15, Retinoblastoma, Rhabdomyoma, Rhabdomyosarcoma, Richter's transformation, Sacrococcygeal teratoma, Salivary Gland Cancer, Sarcoma, Schwannomatosis, Sebaceous gland carcinoma, Secondary neoplasm, Seminoma, Serous tumor, Sertoli-Leydig cell tumor, Sex cord-stromal tumor, Sezary Syndrome, Signet ring cell carcinoma, Skin Cancer, Small blue round cell tumor, Small cell carcinoma, Small Cell Lung Cancer, Small cell lymphoma, Small intestine cancer, Soft tissue sarcoma, Somatostatinoma, Soot wart, Spinal Cord Tumor, Spinal tumor, Splenic marginal zone lymphoma, Squamous cell carcinoma, Stomach cancer, Superficial spreading melanoma, Supratentorial Primitive Neuroectodermal Tumor, Surface epithelial-stromal tumor, Synovial sarcoma, T-cell acute lymphoblastic leukemia, T-cell large granular lymphocyte leukemia, T-cell leukemia, T-cell lymphoma, T-cell prolymphocytic leukemia, Teratoma, Terminal lymphatic cancer, Testicular cancer, Thecoma, Throat Cancer, Thymic Carcinoma, Thymoma, Thyroid cancer, Transitional Cell Cancer of Renal Pelvis and Ureter, Transitional cell carcinoma, Urachal cancer, Urethral cancer, Urogenital neoplasm, Uterine sarcoma, Uveal melanoma, Vaginal Cancer, Verner Morrison syndrome, Verrucous carcinoma, Visual Pathway Glioma, Vulvar Cancer, Waldenstrom's macroglobulinemia, Warthin's tumor, Wilms' tumor, or any combination thereof.
  • In other embodiments, the methods of using a PI3Kα inhibitor and an mTOR inhibitor described herein are applied to the treatment of heart conditions including atherosclerosis, heart hypertrophy, cardiac myocyte dysfunction, elevated blood pressure and vasoconstriction. The invention also relates to a method of treating diseases related to vasculogenesis or angiogenesis in a mammal that comprises administering to said mammal a therapeutically effective amount of a PI3Kα inhibitor and an mTOR inhibitor of the present invention, or any pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof.
  • In some embodiments, said method is for treating a disease selected from the group consisting of tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, atherosclerosis, inflammatory bowel disease, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • In some embodiments, the invention provides for the use of a PI3Kα inhibitor and an mTOR inhibitor for treating a disease condition associated with PI3-kinase α and/or mTOR, including, but not limited to, conditions related to an undesirable, over-active, harmful or deleterious immune response in a mammal, collectively termed “autoimmune disease.” Autoimmune disorders include, but are not limited to, Crohn's disease, ulcerative colitis, psoriasis, psoriatic arthritis, juvenile arthritis and ankylosing spondilitis, Other non-limiting examples of autoimmune disorders include autoimmune diabetes, multiple sclerosis, systemic lupus erythematosus (SLE), rheumatoid spondylitis, gouty arthritis, allergy, autoimmune uveitis, nephrotic syndrome, multisystem autoimmune diseases, autoimmune hearing loss, adult respiratory distress syndrome, shock lung, chronic pulmonary inflammatory disease, pulmonary sarcoidosis, pulmonary fibrosis, silicosis, idiopathic interstitial lung disease, chronic obstructive pulmonary disease, asthma, restenosis, spondyloarthropathies, Reiter's syndrome, autoimmune hepatitis, inflammatory skin disorders, vasculitis oflarge vessels, medium vessels or small vessels, endometriosis, prostatitis and Sjogren's syndrome. Undesirable immune response can also be associated with or result in, e.g., asthma, emphysema, bronchitis, psoriasis, allergy, anaphylaxsis, auto-immune diseases, rhuematoid arthritis, graft versus host disease, transplantation rejection, lung injuries, and lupus erythematosus. The pharmaceutical compositions of the present invention can be used to treat other respiratory diseases including but not limited to diseases affecting the lobes of lung, pleural cavity, bronchial tubes, trachea, upper respiratory tract, or the nerves and muscle for breathing. The compositions of the invention can be further used to treat multiorgan failure.
  • The invention also provides methods of using a PI3Kα inhibitor and an mTOR inhibitor for the treatment of liver diseases (including diabetes), pancreatitis or kidney disease (including proliferative glomerulonephritis and diabetes-induced renal disease) or pain in a mammal.
  • The invention also provides a method of using a PI3Kα inhibitor and an mTOR inhibitor for the treatment of sperm motility. The invention further provides a method of using a PI3Kα inhibitor and an mTOR inhibitor for the treatment of neurological or neurodegenerative diseases including, but not limited to, Alzheimer's disease, Huntington's disease, CNS trauma, and stroke.
  • The invention further provides a method of using a PI3Kα inhibitor and an mTOR inhibitor for the prevention of blastocyte implantation in a mammal.
  • The invention also relates to a method of using a PI3Kα inhibitor and an mTOR inhibitor for treating a disease related to vasculogenesis or angiogenesis in a mammal which can manifest as tumor angiogenesis, chronic inflammatory disease such as rheumatoid arthritis, inflammatory bowel disease, atherosclerosis, skin diseases such as psoriasis, eczema, and scleroderma, diabetes, diabetic retinopathy, retinopathy of prematurity, age-related macular degeneration, hemangioma, glioma, melanoma, Kaposi's sarcoma and ovarian, breast, lung, pancreatic, prostate, colon and epidermoid cancer.
  • The invention further provides a method of using a PI3Kα inhibitor and an mTOR inhibitor for the treatment of disorders involving platelet aggregation or platelet adhesion, including but not limited to Bernard-Soulier syndrome, Glanzmann's thrombasthenia, Scott's syndrome, von Willebrand disease, Hermansky-Pudlak Syndrome, and Gray platelet syndrome.
  • In some embodiments, methods of using a PI3Kα inhibitor and an mTOR inhibitor are provided for treating a disease which is skeletal muscle atrophy, skeletal muscle hypertrophy, leukocyte recruitment in cancer tissue, invasion metastasis, melanoma, Kaposi's sarcoma, acute and chronic bacterial and viral infections, sepsis, glomerulo sclerosis, glomerulo, nephritis, or progressive renal fibrosis.
  • Certain embodiments contemplate a human subject such as a subject that has been diagnosed as having or being at risk for developing or acquiring a disease condition associated with PI3-kinase α and/or mTOR. Certain other embodiments contemplate a non-human subject, for example a non-human primate such as a macaque, chimpanzee, gorilla, vervet, orangutan, baboon or other non-human primate, including such non-human subjects that can be known to the art as preclinical models, including preclinical models for inflammatory disorders. Certain other embodiments contemplate a non-human subject that is a mammal, for example, a mouse, rat, rabbit, pig, sheep, horse, bovine, goat, gerbil, hamster, guinea pig or other mammal. There are also contemplated other embodiments in which the subject or biological source can be a non-mammalian vertebrate, for example, another higher vertebrate, or an avian, amphibian or reptilian species, or another subject or biological source. In certain embodiments of the present invention, a transgenic animal is utilized. A transgenic animal is a non-human animal in which one or more of the cells of the animal includes a nucleic acid that is non-endogenous (i.e., heterologous) and is present as an extrachromosomal element in a portion of its cell or stably integrated into its germ line DNA (i.e., in the genomic sequence of most or all of its cells).
  • Exemplary mTor Inhibitor Compounds
  • In one aspect, the invention provides a compound which is an inhibitor of mTor of the Formula I:
  • Figure US20160235758A1-20160818-C00004
  • or a pharmaceutically acceptable salt thereof, wherein:
  • X1 is N or C-E1, X2 is N or C, X3 is N or C, X4 is C—R9 or N, X5 is N or C-E1, X6 is C or N, and X7 is C or N; and wherein no more than two nitrogen ring atoms are adjacent;
  • R1 is H, -L-C3-8cycloalkyl, —C3-8cycloalkyl, -L-aryl, -L-heteroaryl, -L-C1-10alkylaryl, -L-C1-10alkylhetaryl, -L-C1-10alkylheterocylyl, -L-C2-10alkenyl, -L-C2-10alkynyl, -L-C2-10alkenyl-C3-8cycloalkyl, -L-C2-10alkynyl-C3-8cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocylyl, -L-heteroalkyl-C3-8cycloalkyl, -L-aralkyl, -L-heteroaralkyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • E1 and E2 are independently —(W1)j—R4;
  • M1 is a 5, 6, 7, 8, 9, or -10 membered ring system, wherein the ring system is monocyclic or bicyclic, substituted with R5 and additionally optionally substituted with one or more —(W2)k—R2;
  • each k is 0 or 1;
  • j in E1 or j in E2, is independently 0 or 1;
  • W1 is —O—, —NR7—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • W2 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)C(O)N(R8)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • R2 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32, aryl (e.g. bicyclic aryl, unsubstituted aryl, or substituted monocyclic aryl), hetaryl, C3-8cycloalkyl, C3-8cycloalkyl C3-8cyclo alkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl (e.g. C2-10alkyl-monocyclic aryl, C1-10alkyl-substituted monocyclic aryl, or C1-10alkylbicycloaryl), C1-10alkylhetaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylhetaryl, C2-10alkenylhetero alkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylhetaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocylyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heteroalkyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl (e.g. monocyclic aryl-C2-10alkyl, substituted monocyclic aryl-C1-10alkyl, or bicycloaryl-C1-10alkyl), aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, hetaryl-C3-8cycloalkyl, hetaryl-heteroalkyl, or hetaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R3 and R4 are independently hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31—C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31—SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR3)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32 aryl, hetaryl, C1-4alkyl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl, C1-10alkylhetaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylhetaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylhetaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocylyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, hetaryl-C3-8cycloalkyl, heteroalkyl, hetaryl-heteroalkyl, or hetaryl-heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R″, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R″, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R5 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32;
  • each of R31, R32, and R33 is independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or hetaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35;
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, hetaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • each of R7′ and R8′ is independently hydrogen, C1-10alkyl, C2-10alkenyl, aryl, heteroaryl, heterocyclyl or C3-10cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R6;
  • R6 is halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2NR31R32, —NR31R32, or —NR34R35; and
  • R9 is H, halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, hetaryl-C1-10alkyl, hetaryl-C2-10alkenyl, hetaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or hetaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or —NR34R35.
  • M1 is a 5, 6, 7, 8, 9, or -10 membered ring system, wherein the ring system is monocyclic or bicyclic.
  • The monocyclic M1 ring is unsubstituted or substituted with one or more R5 substituents (including 0, 1, 2, 3, 4, or 5 R5 substituents). In some embodiments, the monocyclic M1 ring is aromatic (including phenyl) or heteroaromatic (including but not limited to pyridinyl, pyrrolyl, imidazolyl, thiazolyl, or pyrimidinyl). The monocyclic M1 ring may be a 5 or 6 membered ring (including but not limited to pyridinyl, pyrrolyl, imidazolyl, thiazolyl, or pyrimidinyl). In some embodiments, M2 is a five membered heteroaromatic group with one heteroatom, wherein the heteroatom is N, S, or O. In another embodiment, M2 is a five membered heteroaromatic group with two heteroatoms, wherein the heteroatoms are nitrogen and oxygen or nitrogen and sulfur.
  • The bicyclic M1 ring is unsubstituted or substituted with one or more R5 substituents (including 0, 1, 2, 3, 4, 5, 6 or 7 R5 substituents). Bicyclic M1 ring is a 7, 8, 9, or 10 membered aromatic or heteroaromatic. Examples of an aromatic bicyclic M1 ring include naphthyl. In other embodiments the bicyclic M1 ring is heteroaromatic and includes but is not limited to benzothiazolyl, quinolinyl, quinazolinyl, benzoxazolyl, and benzoimidazolyl.
  • The invention also provides compounds wherein M1 is a moiety having a structure of Formula M1-A or Formula M1-B:
  • Figure US20160235758A1-20160818-C00005
  • wherein W1, W2, and W7 are independently N or C—R5; W4 and W10 are independently N—R5, O, or S; W6 and W8 are independently N or C—R5; W5 and W9 are independently N or C—R2; and W3 is C or N, provided no more than two N and/or N—R5 are adjacent and no two 0 or S are adjacent.
  • In some embodiments of the invention, the M1 moiety of Formula M1-A is a moiety of Formula M1-A1, Formula M1-A2, Formula M1-A3, or Formula M1-A4:
  • Figure US20160235758A1-20160818-C00006
  • wherein W4 is N—R5, O, or S; W6 is N or C—R5 and W5 is N or C—R2.
  • Some nonlimiting examples of the M1 moiety of Formula M1-A include:
  • Figure US20160235758A1-20160818-C00007
  • wherein R5 is —(W1)k—R53 or R55; each k is independently 0 or 1, n is 0, 1, 2, or 3, and —(W1)k—R53 and R55 are as defined above.
  • In other embodiments of the invention, the M1 moiety of Formula M1-B is a moiety of Formula M1-B1, Formula M1-B2, Formula M1-B3, or Formula M1-B4:
  • Figure US20160235758A1-20160818-C00008
  • wherein W10 is N—R5, O, or S, Wg is N or C—R5, and W5 is N or C—R2.
  • Some nonlimiting examples of the M1 moiety of Formula M1-B include:
  • Figure US20160235758A1-20160818-C00009
  • wherein R′5 is —(W1)k—R53 or R55; k is 0 or 1, n is 0, 1, 2, or 3, and —(W1)k—R53 and R55 are as defined above.
  • The invention also provides compounds wherein M1 is a moiety having a structure of Formula M1-C or Formula M1-D:
  • Figure US20160235758A1-20160818-C00010
  • wherein W12, W13, W14, and W15 are independently N or C—R5; W11 and W18 are independently N—R5, O, or S; W16 and W17 are independently N or C—R5; provided no more than two N are adjacent.
  • In other embodiments of the invention, the M1 moiety of Formula M1-C or Formula M1-D is a moiety of Formula M1-C1 or Formula M1-D1:
  • Figure US20160235758A1-20160818-C00011
  • wherein W11 and W18 are N—R5, O, or S; and W16 and W17 are N or C—R5.
  • Some nonlimiting examples of the M1 moiety of Formula M1-C and Formula M1-D include:
  • Figure US20160235758A1-20160818-C00012
  • wherein R′5 is —(W1)k—R53 or R55; k is 0 or 1, and —(W1)k—R53 and R55 are as defined above.
  • The invention also provides compounds wherein M1 is a moiety having a structure of Formula M1-E:
  • Figure US20160235758A1-20160818-C00013
  • wherein X11, X12, X13, X14, X15, X16, and X17 are independently N, or C—R5; provided that no more than two N are adjacent.
  • In some embodiments of the invention, the M1 moiety having a structure of Formula M1-E, is a moiety having a structure of Formula M1-E1, M1-E2, M1-E3, M1-E4, M1-E5, M1-E6, M1-E7, or M1-E8:
  • Figure US20160235758A1-20160818-C00014
  • In some embodiments of the invention, the M1 moiety having a structure of Formula M1-E, is a moiety having a structure:
  • Figure US20160235758A1-20160818-C00015
  • Some nonlimiting examples of the M1 moiety of Formula M1-E include:
  • Figure US20160235758A1-20160818-C00016
  • wherein R′5 is —(W1)k—R53 or R55; k is 0 or 1, n is 0, 1, 2, or 3, and —(W1)k—R53 or R55 are as defined above. In some embodiments, k is 0, and R5 is R53.
  • In some embodiments, R53 is hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or unsubstituted or substituted C3-C8cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl). In other embodiments, R53 is monocyclic or bicyclic aryl, wherein the R53 aryl is unsubstituted or substituted. Some examples of aryl include but are not limited to phenyl, naphthyl or fluorenyl. In some other embodiments, R53 is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R53 includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R53 includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, and purinyl. Additionally, R53 may be alkylcycloalkyl (including but not limited to cyclopropylethyl, cyclopentylethyl, and cyclobutylpropyl), -alkylaryl (including but not limited to benzyl, phenylethyl, and phenylnaphthyl), alkylhetaryl (including but not limited to pyridinylmethyl, pyrrolylethyl, and imidazolylpropyl), or alkylheterocyclyl (non-limiting examples are morpholinylmethyl, 1-piperazinylmethyl, and azetidinylpropyl). For each of alkylcycloalkyl, alkylaryl, alkylhetaryl, or alkylheterocyclyl, the moiety is connected to M1 through the alkyl portion of the moiety In other embodiments, R53 is unsubstituted or substituted C2-C10alkenyl (including but not limited to alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C10alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl).
  • Further embodiments provide R53 wherein R53 is alkenylaryl, alkenylheteroaryl, alkenylheteroalkyl, or alkenylheterocycicyl, wherein each of alkenyl, aryl, heteroaryl, heteroalkyl, and heterocyclyl is as described herein and wherein the alkenylaryl, alkenylhetaryl, alkenylheteroalkyl, or alkenylheterocycicyl moiety is attached to M1 through the alkenyl. Some nonlimiting examples in include styryl, 3-pyridinylallyl, 2-methoxyethoxyvinyl, and 3-morpholinlylallyl In other embodiments, R53 is -alkynylaryl, -alkynylhetaryl, -alkynylheteroalkyl, -alkynylheterocylyl, -alkynylcycloalkyl, or -alkynylC3-8cycloalkenyl, wherein each of alkynyl, aryl, heteroaryl, heteroalkyl, and heterocyclyl is as described herein and wherein the alkynylaryl, alkynylhetaryl, alkynylheteroalkyl, or alkynylheterocycicyl moiety is attached to M1 through the alkynyl. Alternatively, R53 is -alkoxyalkyl, -alkoxyalkenyl, or -alkoxyalkynyl, wherein each of alkoxy, alkyl, alkenyl, and alkynyl is as described herein and wherein the -alkoxyalkyl, -alkoxyalkenyl, or -alkoxyalkynyl moiety is attached to M1 through the alkoxy. In yet other embodiments, R53 is -heterocyclylalkyl, -heterocyclylalkenyl, or -heterocyclylalkynyl, wherein the heterocyclyl, alkyl, alkenyl, or alkynyl is as described herein and wherein the -heterocyclylalkyl, -heterocyclylalkenyl, or -heterocyclylalkynyl is attached to M1 through the heterocyclyl portion of the moiety. Further, R53 may be aryl-alkenyl, aryl-alkynyl, or aryl-heterocyclyl, wherein the aryl, alkenyl, alkynyl, or heterocyclyl is as described herein and wherein the aryl-alkenyl, aryl-alkynyl, or aryl-heterocyclyl moiety is attached to M1 through the aryl portion of the moiety. In some other embodiments, R53 is heteroaryl-alkyl, heteroaryl-alkenyl, heteroaryl -alkynyl, heteroaryl-cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of heteroaryl, alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, and heterocyclyl is as described herein and wherein the heteroaryl-alkyl, heteroaryl-alkenyl, heteroaryl-alkynyl, heteroaryl-cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl moiety is attached to M1 through the heteroaryl portion of the moiety.
  • For each of the aryl or heteroaryl moieties forming part or all of R53, the aryl or heteroaryl is unsubstituted or is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —NR31R32, —C(═O)NNR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR3)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32 substituents. Additionally, each of the alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moieties forming part of all of R53 is unsubstituted or substituted with one or more halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NNR34R35, or —C(═O)NR31R32 substituents.
  • In other embodiments, R5 is —W1—R53. In some embodiments, R5 is —OR53, including but not limited to Oalkyl (including but not limited to methoxy or ethoxy), -Oaryl (including but not limited to phenoxy), —O-heteroaryl (including but not limited to pyridinoxy) and —O-heterocycloxy(including but not limited to 4-N-piperidinoxy). In some embodiments R5 is —NR6R53 including but not limited to anilinyl, diethylamino, and 4-N-piperidinylamino. In yet other embodiments R5 is —S(O)0-2R53, including but not limited to phenylsulfonyl and pyridinylsulfonyl. The invention also provides compounds wherein R5 is —C(O) (including but not limited to acetyl, benzoyl, and pyridinoyl) or —C(O)O R53 (including but not limited to carboxyethyl, and carboxybenzyl). In other embodiments, R5 is —C(O)N(R6)R53 (including but not limited to C(O)NH(cyclopropyl) and C(O)N(Me)(phenyl)) or —CH(R6)N(R7)R53 (including but not limited to —CH2—NH-pyrrolidinyl, CH2—NHcyclopropyl, and CH2-anilinyl). Alternatively, R5 is —N(R6)C(O)R53 (including but not limited to —NHC(O)phenyl, —NHC(O)cyclopentyl, and to —NHC(O)piperidinyl) or —N(R6)S(O)2 R53 (including but not limited to —NHS(O)2phenyl, —NHS(O)2piperazinyl, and —NHS(O)2methyl. Additionally, R5 is —N(R6)S(O) R53, —CH(R6)N(C(O)OR7) R53, —CH(R7)N(C(O)R7) R53, —CH(R6)N(SO2R7) R53, —CH(R6)N(R7) R53, —CH(R6)C(O)N(R7) R53, —CH(R6)N(R7)C(O) R53, —CH(R6)N(R7)S(O) R53, or —CH(R6)N(R7)S(O)2 R53.
  • Alternatively, R5 is R55. R55 is halo, —OH, —NO2, —CF3, —OCF3, or —CN. In some other embodiments, R55 is —R31, —OR31 (including but not limited to methoxy, ethoxy, and butoxy) —C(O)R31 (non-limiting examples include acetyl, propionyl, and pentanoyl), or —CO2R31 (including but not limited to carboxymethyl, carboxyethyl and carboxypropyl). In further embodiments, R55 is —NR31R32, —C(═O)NR31R32, —SO2NR31R32, or —S(O)0-2R31. In other embodiments, R55 is —NR34R35 or —SO2 NR34R35, wherein R34R35 are taken together with the nitrogen to which R34R35 are attached to form a cyclic moiety. The cyclic moiety so formed may be unsubstituted or substituted, wherein the substituents are selected from the group consisting of alkyl, —C(O)alkyl, —S(O)2alkyl, and —S(O)2aryl. Examples include but are not limited to morpholinyl, piperazinyl, or —SO2-(4-N-methyl-piperazin-1-yl. Additionally, R55 is —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —C(═O)NNR34R35, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32; In yet another embodiment, R55 is —O-aryl, including but not limited to phenoxy, and naphthyloxy.
  • The invention further provides a compound which is an mTor inhibitor, wherein the compound has the Formula I-A:
  • Figure US20160235758A1-20160818-C00017
  • or a pharmaceutically acceptable salt thereof, wherein:
  • X1 is N or C-E1, X2 is N, X3 is C, and X4 is C—R9 or N; or X1 is N or C-E1, X2 is C, X3 is N, and X4 is C—R9 or N;
  • R1 is —H, -L-C1-10alkyl, -L-C3-8cycloalkyl, -L-C1-10alkyl-C3-8cycloalkyl, -L-aryl, -L-heteroaryl, -L-C1-10alkylaryl, -L-C1-10alkylheteroaryl, -L-C1-10alkylheterocyclyl, -L-C2-10alkenyl, -L-C2-10alkynyl, -L-C2-10alkenyl-C3-8cycloalkyl, -L-C2-10alkynyl-C3-8cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C3-8cycloalkyl, -L-aralkyl, -L-heteroaralkyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • M1 is a moiety having the structure of Formula M1-F1 or M1-F2:
  • Figure US20160235758A1-20160818-C00018
  • k is 0 or 1;
  • E1 and E2 are independently —(W1)j—R4;
  • j, in each instance (i.e., in E1 or j in E2), is independently 0 or 1
  • W1 is —O—, —NR7—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • W2 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)C(O)N(R8)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • R2 is hydrogen, halogen, —OH, —R31, —CF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32 aryl (e.g. bicyclic aryl, unsubstituted aryl, or substituted monocyclic aryl), heteroaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl -L-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl (e.g. C2-10alkyl-monocyclic aryl, C1-10alkyl-substituted monocyclic aryl, or C1-10alkylbicycloaryl), C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heteroalkyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl (e.g. monocyclic aryl-C2-10alkyl, substituted monocyclic aryl-C1-10alkyl, or bicycloaryl-C1-10alkyl), aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31 S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R3 and R4 are independently hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32, aryl, heteroaryl, C1-4 alkyl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R5 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32;
  • R31, R32, and R33, in each instance, are independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35;
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, heteroaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • R7′ and R8′ are each independently hydrogen, C1-10alkyl, C2-10alkenyl, aryl, heteroaryl, heterocyclyl or
  • C3-10cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R6;
  • R6 is halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or —NR34R35; and
  • R9 is H, halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or —NR34R35.
  • In some embodiments, X4 is C—R9.
  • The invention also provides an inhibitor as defined above, wherein the compound is of Formula I:
  • Figure US20160235758A1-20160818-C00019
  • or a pharmaceutically acceptable salt thereof, and wherein the substituents are as defined above.
  • In various embodiments the compound of Formula I-B or its pharmaceutically acceptable salt thereof, is a compound having the structure of Formula I-B1 or Formula I-B2:
  • Figure US20160235758A1-20160818-C00020
  • or a pharmaceutically acceptable salt thereof.
  • In various embodiments of Formula I-B1, X1 is N and X2 is N. In other embodiments, X1 is C-E1 and X2 is N. In yet other embodiments, X1 is NH and X2 is C. In further embodiments, X1 is CH-E1 and X2 is C.
  • In various embodiments of Formula I-B2, X1 is N and X2 is C. In further embodiments, X1 is C-E1 and X2 is C.
  • In various embodiments, X1 is C—(W1)j—R4, where j is 0.
  • In another embodiment, X1 is CH. In yet another embodiment, X1 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of X1, it is C—(W1), —R4. In various embodiments of X1, j is 1, and W1 is —O—. In various embodiments of X1, j is 1, and W1 is —NR7—. In various embodiments of X1, j is 1, and W1 is —NH—. In various embodiments of X1, j is 1, and W1 is —S(O)0-2-. In various embodiments of X1, j is 1, and W1 is —C(O)—. In various embodiments of X1, j is 1, and W1 is —C(O)N(R7)—. In various embodiments of X1, j is 1, and W1 is —N(R7)C(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)2—. In various embodiments of X1, j is 1, and W1 is —C(O)O—. In various embodiments of X1, j is 1, and W1 is CH(R7)N(C(O)OR8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)C(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In another embodiment, X1 is CH2. In yet another embodiment, X1 is CH-halogen, where halogen is Cl, F, Br, or I.
  • In another embodiment, X1 is N.
  • In various embodiments, X2 is N. In other embodiments, X2 is C.
  • In various embodiments, E2 is —(W1)j—R4, where j is 0.
  • In another embodiment, E2 is CH. In yet another embodiment, E2 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of E2, it is —(W1)j—R4. In various embodiments of E2, j is 1, and W1 is —O—. In various embodiments of E2, j is 1, and W1 is —NR7—. In various embodiments of E2, j is 1, and W1 is —NH—. In various embodiments of E2, j is 1, and W1 is —S(O)0-2-. In various embodiments of E2, j is 1, and W1 is —C(O)—. In various embodiments of E2, j is 1, and W1 is —C(O)N(R7)—. In various embodiments of E2, j is 1, and W1 is —N(R7)C(O)—. In various embodiments of E2, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of E2, j is 1, and W1 is —N(R7)S(O)2—. In various embodiments of E2, j is 1, and W1 is —C(O)O—. In various embodiments of E2, j is 1, and W1 is CH(R7)N(C(O)OR8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)C(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R)N(R8)S(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In various embodiments when M1 is a moiety of Formula M1-F1, M1 is benzoxazolyl substituted with —(W2)k—R2. In some embodiments, M1 is a benzoxazolyl substituted at the 2-position with (W2)j—R2, In some embodiments, M1 is either a 5-benzoxazolyl or a 6-benzoxazolyl moiety, optionally substituted at the 2-position with —(W2)j—R2. Exemplary Formula M1-F1 M1 moieties include but are not limited to the following:
  • Figure US20160235758A1-20160818-C00021
  • In various embodiments when M1 is a moiety of Formula M1-F2, Formula M1-F2 is an aza-substituted benzoxazolyl moiety having a structure of one of the following formulae:
  • Figure US20160235758A1-20160818-C00022
  • Exemplary Formula M1-F2 M1 moieties include but are not limited to the following:
  • Figure US20160235758A1-20160818-C00023
  • In various embodiments of M1, k is 0. In other embodiments of M1, k is 1, and W2 is selected from one of the following: —O—, —NR7—, —S(O)0-2, —C(O)—, —C(O)N(R7), —N(R7)C(O), or N(R7)C(O)N(R8). In yet another embodiment of M1, k is 1, and W2 is —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8), —CH(R7)N(C(O)R8), or —CH(R7)N(SO2R8). In a further embodiment of M1, k is 1, and W2 is —CH(R7)N(R8), —CH(R7)C(O)N(R8), —CH(R7)N(R8)C(O), or —CH(R7)N(R8)S(O)—. In yet another embodiment of M1, k is 1, and W2 is CH(R7)N(R8)S(O)2.
  • The invention provides an inhibitor of mTor which is a compound of Formula I-C or Formula I-D:
  • Figure US20160235758A1-20160818-C00024
  • or a pharmaceutically acceptable salt thereof, wherein X1 is N or C-E1, X2 is N, and X3 is C; or X1 is N or C-E1, X2 is C, and X3 is N;
  • R1 is —H, -L-C1-10alkyl, -L-C3-8cycloalkyl, -L-C1-10alkyl-C3-8cycloalkyl, -L-aryl, -L-heteroaryl, -L-C1-10alkylaryl, -L-C1-10alkylheteroaryl, -L-C1-10alkylheterocyclyl, -L-C2-10alkenyl, -L-C2-10alkynyl, -L-C2-10alkenyl-C3-8cycloalkyl, -L-C2-10alkynyl-C3-8cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C3-8cycloalkyl, -L-aralkyl, -L-heteroaralkyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • E1 and E2 are independently —(W1)j—R4;
  • j in E1 or j in E2, is independently 0 or 1;
  • W1 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • W2 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)C(O)N(R8)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • k is 0 or 1;
  • R2 is hydrogen, halogen, —OH, —R31, —CF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32 aryl (e.g. bicyclic aryl, unsubstituted aryl, or substituted monocyclic aryl), heteroaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl (e.g. C2-10alkyl-monocyclic aryl, C1-10alkyl-substituted monocyclic aryl, or C1-10alkylbicycloaryl), C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl (e.g. monocyclic aryl-C2-10alkyl, substituted monocyclic aryl-C1-10alkyl, or bicycloaryl-C1-10alkyl), aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31—SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32 s, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R3 and R4 are independently hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31—C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31 S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32, aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl, C1-10alkylhereroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31—C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31—CO2R31, —NR34R35, or —C(═O)NR31R32;
  • R5 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32;
  • R31, R32, and R33, in each instance, are independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35;
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, heteroaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom; and
  • R7′ and R8′ are each independently hydrogen, C1-10alkyl, C2-10alkenyl, aryl, heteroaryl, heterocyclyl or C3-10cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R6; and R6 is halo, —OR31, —SH, NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O) NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, or C2-10alkynyl; or R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, each of which is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O) NR34R″, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or NR34R35.
  • In various embodiments of the compound of Formula I-C, the compound has a structure of Formula I-C1 or Formula I-C2:
  • Figure US20160235758A1-20160818-C00025
  • or a pharmaceutically acceptable salt thereof.
  • In some embodiments of Formula I-C1, X1 is N and X2 is N. In other embodiments, X1 is C-E1 and X2 is N. In yet other embodiments, X1 is NH and X2 is C. In further embodiments, X1 is CH-E1 and X2 is C.
  • In several embodiments of Formula I-C2, X1 is N and X2 is C. In yet other embodiments, X1 is NH and X2 is C. In further embodiments, X1 is CH-E1 and X2 is C.
  • In various embodiments of the compound of Formula I-D, the compound has a structure of Formula I-D1 or Formula I-D2:
  • Figure US20160235758A1-20160818-C00026
  • or a pharmaceutically acceptable salt thereof.
  • In some embodiments of Formula I-D1, X1 is N and X2 is N. In other embodiments, X1 is C-E1 and X2 is N. In yet other embodiments, X1 is NH and X2 is C. In further embodiments, X1 is CH-E1 and X2 is C.
  • In several embodiments of Formula I-D2, X1 is N and X2 is C. In further embodiments, X1 is C-E1 and X2 is C.
  • In various embodiments, X1 is C—(W1)j—R4, where j is 0.
  • In another embodiment, X1 is CH. In yet another embodiment, X1 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of X1, it is C—(W1), —R4. In various embodiments of X1, j is 1, and W1 is —O—. In various embodiments of X1, j is 1, and W1 is —NR7—. In various embodiments of X1, j is 1, and W1 is —NH—. In various embodiments of X1, j is 1, and W1 is —S(O)0-2. In various embodiments of X1, j is 1, and W1 is —C(O)—. In various embodiments of X1, j is 1, and W1 is —C(O)N(R7). In various embodiments of X1, j is 1, and W1 is —N(R7)C(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)2. In various embodiments of X1, j is 1, and W1 is —C(O)O—. In various embodiments of X1, j is 1, and W1 is CH(R7)N(C(O)OR8). In various embodiments of X1, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)C(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In various embodiments, X1 is CH—(W1)j—R4, where j is 0.
  • In another embodiment, X1 is CH2. In yet another embodiment, X1 is CH-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of X1, it is CH—(W1)j—R4. In various embodiments of X1, j is 1, and W1 is —O—. In various embodiments of X1, j is 1, and W1 is NR7—. In various embodiments of X1, j is 1, and W1 is NH—. In various embodiments of X1, j is 1, and W1 is —S(O)0-2. In various embodiments of X1, j is 1, and W1 is —C(O)—. In various embodiments of X1, j is 1, and W1 is —C(O)N(R7). In various embodiments of X1, j is 1, and W1 is N(R7)C(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)2. In various embodiments of X1, j is 1, and W1 is —C(O)O—. In various embodiments of X1, j is 1, and W1 is CH(R7)N(C(O)OR8). In various embodiments of X1, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of X1, j is 1, and W1 is CH(R7)N(R8)C(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of X1, j is 1, and W1 is CH(R7)N(R8)S(O)2—.
  • In another embodiment, X1 is N.
  • In various embodiments, X2 is N. In other embodiments, X2 is C.
  • In various embodiments, E2 is (W1)j—R4, where j is 0.
  • In another embodiment, E2 is CH. In yet another embodiment, E2 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of E2, it is (W1)j—R4. In various embodiments of E2, j is 1, and W1 is —O—. In various embodiments of E2, j is 1, and W1 is NR7—. In various embodiments of E2, j is 1, and W1 is NH—. In various embodiments of E2, j is 1, and W1 is —S(O)0-2. In various embodiments of E2, j is 1, and W1 is —C(O)—. In various embodiments of E2, j is 1, and W1 is —C(O)N(R7). In various embodiments of E2, j is 1, and W1 is N(R7)C(O)—. In various embodiments of E2, j is 1, and W1 is N(R7)S(O)—. In various embodiments of E2, j is 1, and W1 is —N(R7)S(O)2. In various embodiments of E2, j is 1, and W1 is —C(O)O—. In various embodiments of E2, j is 1, and W1 is CH(R7)N(C(O)OR8). In various embodiments of E2, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of E2, j is 1, and W1 is CH(R7)N(R8)C(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In various embodiments, k is 0. In other embodiments, k is 1 and W2 is —O—. In another embodiment, k is 1 and W2 is NR7. In yet another embodiment of, k is 1, and W2 is —S(O)0-2. In another embodiment of, k is 1 and W2 is —C(O)—. In a further embodiment, k is 1 and W2 is —C(O)N(R7). In another embodiment, k is 1, and W2 is N(R7)C(O)—. In another embodiment, k is 1 and W2 is N(R7)C(O)N(R8). In yet another embodiment, k is 1 and W2 is —N(R7)S(O)—. In still yet another embodiment, k is 1 and W2 is —N(R7)S(O)2. In a further embodiment, k is 1 and W2 is —C(O)O—. In another embodiment, k is 1 and W2 is CH(R7)N(C(O)OR8). In another embodiment, k is 1 and W2 is —CH(R7)N(C(O)R8)—. In another embodiment, k is 1 and W2 is —CH(R7)N(SO2R8)—. In a further embodiment, k is 1 and W2 is —CH(R7)N(R8)—. In another embodiment, k is 1 and W2 is CH(R7)C(O)N(R8). In yet another embodiment, k is 1 and W2 is CH(R7)N(R8)C(O)—. In another embodiment, k is 1 and W2 is —CH(R7)N(R8)S(O)—. In yet another embodiment, k is 1 and W2 is CH(R7)N(R8)S(O)2—.
  • The invention also provides a compound which is an mTor inhibitor of Formula I-E:
  • Figure US20160235758A1-20160818-C00027
  • or a pharmaceutically acceptable salt thereof, wherein: X1 is N or C-E1, X2 is N, and X3 is C; or X1 is N or C-E1, X2 is C, and X3 is N;
  • R1 is —H, L-C1-10alkyl, -L-C3-8cycloalkyl, -L-C1-10alkyl-C3-8cycloalkyl, -L-aryl, -L-heteroaryl, -L-C1-10alkylaryl, -L-C1-10alkylheteroaryl, -L-C1-10alkylheterocyclyl, -L-C2-10alkenyl, -L-C2-10alkynyl, -L-C2-10alkenyl-C3-8cycloalkyl, -L-C2-10alkynyl-C3-8cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C3-8cycloalkyl, -L-aralkyl, -L-heteroaralkyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • M1 is a moiety having the structure of Formula M1-F1 or Formula M1-F2:
  • Figure US20160235758A1-20160818-C00028
  • k is 0 or 1;
  • E1 and E2 are independently (W1); —R4;
  • j in E1 or j in E2, is independently 0 or 1;
  • W1 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O), —N(R7)S(O)—, —N(R7)S(O)2, —C(O)O, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8), —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • W2 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)C(O)N(R8), —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • R2 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32 aryl (e.g. bicyclic aryl, unsubstituted aryl, or substituted monocyclic aryl), heteroaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl (e.g. C2-10alkyl-monocyclic aryl, C1-10alkyl-substituted monocyclic aryl, or C1-10alkylbicycloaryl), C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynylC2-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cyclo alkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl (e.g. monocyclic aryl-C2-10alkyl, substituted monocyclic aryl-C1-10alkyl, or bicycloaryl-C1-10alkyl), aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31—SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —OC2R31, —NR34R35, or —C(═O)NR31R32; R3 and R4 are independently hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31 S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32, aryl, heteroaryl, C1-4 alkyl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl, C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31—C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR—SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more halo, —OH, —R31—CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R5 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32;
  • R31, R32, and R33, in each instance, are independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl), (C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35;
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, heteroaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • R7′ and R8′ are each independently hydrogen, C1-10alkyl, C2-10alkenyl, aryl, heteroaryl, heterocyclyl or C3-10cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R6;
  • R6 is halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or —NR34R35; and
  • R9 is H, halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or —NR34R35.
  • In various embodiments of the compound of Formula I-E, the compound has a structure of Formula I-E1 or Formula I-E2:
  • Figure US20160235758A1-20160818-C00029
  • or a pharmaceutically acceptable salt thereof.
  • In some embodiments of Formula I-E1, X1 is N and X2 is N. In other embodiments, X1 is C-E1 and X2 is N. In yet other embodiments, X1 is NH and X2 is C. In further embodiments, X1 is CH-E1 and X2 is C.
  • In several embodiments of Formula I-E2, X1 is N and X2 is C. In further embodiments, X1 is C-E1 and X2 is C.
  • In various embodiments, X1 is C—(W1)j—R4, where j is 0.
  • In another embodiment, X1 is CH. In yet another embodiment, X1 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of X1, it is C—(W1), —R4. In various embodiments of X1, j is 1, and W1 is —O—. In various embodiments of X1, j is 1, and W1 is —NR7—. In various embodiments of X1, j is 1, and W1 is —NH—. In various embodiments of X1, j is 1, and W1 is —S(O)0-2-. In various embodiments of X1, j is 1, and W1 is —C(O)—. In various embodiments of X1, j is 1, and W1 is —C(O)N(R7)—. In various embodiments of X1, j is 1, and W1 is —N(R7)C(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)2—. In various embodiments of X1, j is 1, and W1 is —C(O)O—. In various embodiments of X1, j is 1, and W1 is CH(R7)N(C(O)OR8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)C(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In another embodiment, X1 is N.
  • In various embodiments, X2 is N. In other embodiments, X2 is C.
  • In various embodiments, E2 is —(W1)j—R4, where j is 0.
  • In another embodiment, E2 is CH. In yet another embodiment, E2 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of E2, it is —(W1), —R4. In various embodiments of E2, j is 1, and W1 is —O—. In various embodiments of E2, j is 1, and W1 is —NR7—. In various embodiments of E2, j is 1, and W1 is —NH—. In various embodiments of E2, j is 1, and W1 is —S(O)0-2-. In various embodiments of E2, j is 1, and W1 is —C(O)—. In various embodiments of E2, j is 1, and W1 is —C(O)N(R7)—. In various embodiments of E2, j is 1, and W1 is —N(R7)C(O)—. In various embodiments of E2, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of E2, j is 1, and W1 is —N(R7)S(O)2—. In various embodiments of E2, j is 1, and W1 is —C(O)O—. In various embodiments of E2, j is 1, and W1 is CH(R7)N(C(O)OR8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)C(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In various embodiments when M1 is a moiety of Formula I-E1, M1 is benzoxazolyl substituted with (W2)k—R2. In some embodiments, M1 is a benzoxazolyl moiety, substituted at the 2-position with (W2)k—R2. In some embodiments, M1 is either a 5-benzoxazolyl or a 6-benzoxazolyl moiety, optionally substituted with (W2)k—R2. Exemplary Formula I-E1 M1 moieties include but are not limited to the following:
  • Figure US20160235758A1-20160818-C00030
  • In various embodiments when M1 is a moiety of Formula I-E2, Formula I-E2 is an aza-substituted benzoxazolyl moiety having a structure of one of the following formulae:
  • Figure US20160235758A1-20160818-C00031
  • Exemplary Formula I-E2 M1 moieties include but are not limited to the following:
  • Figure US20160235758A1-20160818-C00032
  • In various embodiments of M1, k is 0. In other embodiments of M1, k is 1 and W2 is —O—. In another embodiment of M1, k is 1 and W2 is —NR7—. In yet another embodiment of M1, k is 1 and W2 is S(O)0-2-. In another embodiment of M1, k is 1 and W2 is —C(O)—. In a further embodiment of M1, k is 1 and W2 is —C(O)N(R7)—. In another embodiment of M1, k is 1 and W2 is N(R7)C(O)—. In another embodiment, k is 1 and W2 is N(R7)C(O)N(R8). In yet another embodiment of M1, k is 1 and W2 is —N(R7)S(O)—. In still yet another embodiment of M1, k is 1 and W2 is —N(R7)S(O)2. In a further embodiment of M1, k is 1 and W2 is —C(O)O—. In another embodiment of M1, k is 1 and W2 is CH(R7)N(C(O)OR8). In another embodiment of M1, k is 1 and W2 is —CH(R7)N(C(O)R8)—. In another embodiment of M1, k is 1 and W2 is —CH(R7)N(SO2R8)—. In a further embodiment of M1, k is 1 and W2 is —CH(R7)N(R8)—. In another embodiment of M1, k is 1 and W2 is —CH(R7)C(O)N(R8)—. In yet another embodiment of M1, k is 1 and W2 is —CH(R7)N(R8)C(O)—. In another embodiment of M1, k is 1 and W2 is —CH(R7)N(R8)S(O)—. In yet another embodiment of M1, k is 1 and W2 is —CH(R7)N(R8)S(O)2—.
  • Additional embodiments of compounds of Formula I, including I-A, I—B, I—C, I-D, I-E and others are described below.
  • In various embodiments of compounds of Formula I, L is absent. In another embodiment, L is —(C═O)—. In another embodiment, L is C(═O)O—. In a further embodiment, L is —C(═O) NR31—. In yet another embodiment, L is —S—. In one embodiment, L is —S(O)—. In another embodiment, L is —S(O)2—. In yet another embodiment, L is —S(O)2NR31—. In another embodiment, L is —NR31—.
  • In various embodiments of compounds of Formula I, R1 is L-C1-10alkyl, which is unsubstituted. In another embodiment, R1 is L-C1-10alkyl, which is substituted by one or more independent R3. In yet another embodiment, R1 is L-unsubstituted C1-10alkyl, where L is absent. In another embodiment, R1 is L-C1-10alkyl, which is substituted by one or more independent R3, and L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C3-8cycloalkyl, which is unsubstituted. In another embodiment, R1 is L-C3-8cycloalkyl, which is substituted by one or more independent R3. In yet another embodiment, R1 is -L-C3-8cycloalkyl, which is unsubstituted, and L is absent. In a further embodiment, R1 is -L-C3-8cycloalkyl which is substituted by one or more independent R3, and L is absent.
  • In various embodiments of compounds of Formula I, R1 is H.
  • In various embodiments of compounds of Formula I, R1 is -L-aryl, which is unsubstituted. In another embodiment, R1 is -L-aryl, which is substituted by one or more independent R3. In another embodiment, R1 is -L-aryl which is unsubstituted, and L is absent. In yet another embodiment, R1 is -L-aryl, which is substituted by one or more independent R3, and L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-heteroaryl, which is unsubstituted. In another embodiment, R1 is -L-heteroaryl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heteroaryl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-heteroaryl, which is substituted by one or more independent R3, and L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C1-10alkyl-C3-8cycloalkyl, which is unsubstituted. In another embodiment, R1 is -L-C1-10alkyl-C3-8cycloalkyl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C1-10alkyl-C3-8cycloalkyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C1-10alkyl-C3-8cycloalkyl, which is substituted by one or more independent R3, and L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C1-10alkylaryl, which is unsubstituted. In another embodiment, R1 is -L-C1-10alkylaryl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C1-10alkylaryl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C1-10alkylaryl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C1-10alkylheteroaryl, which is unsubstituted. In another embodiment, R1 is -L-C1-10alkylheteroaryl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C1-10alkylheteroaryl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C1-10alkylheteroaryl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C1-10alkylheterocyclyl, which is unsubstituted. In another embodiment, R1 is -L-C1-10alkylheterocyclyl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C1-10alkylheterocyclyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C1-10alkylheterocyclyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C2-10alkenyl, which is unsubstituted. In another embodiment, R1 is -L-C2-10alkenyl which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C2-10alkenyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C2-10alkenyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C2-10alkynyl, which is unsubstituted. In another embodiment, R1 is -L-C2-10alkynyl which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C2-10alkynyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C2-10alkynyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C2-10alkenyl-C3-8cycloalkyl, which is unsubstituted. In another embodiment, R1 is -L-C2-10alkenyl-C3-8cycloalkyl which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C2-10alkenyl-C3-8cycloalkyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C2-10alkenyl-C3-8cycloalkyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C2-10alkynyl-C3-8cycloalkyl, which is unsubstituted. In another embodiment, R1 is -L-C2-10alkynyl-C3-8cycloalkyl which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C2-10alkynyl-C3-8cycloalkyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C2-10alkynyl-C3-8cycloalkyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-C2-10alkynyl-C3-8cycloalkyl, which is unsubstituted. In another embodiment, R1 is -L-C2-10alkynyl-C3-8cycloalkyl which is substituted by one or more independent R3. In a further embodiment, R1 is -L-C2-10alkynyl-C3-8cycloalkyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-C2-10alkynyl-C3-8cycloalkyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-heteroalkyl, which is unsubstituted. In another embodiment, R1 is -L-heteroalkyl which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heteroalkyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-heteroalkyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-heteroalkylaryl, which is unsubstituted. In another embodiment, R1 is -L-heteroalkylaryl which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heteroalkylaryl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-heteroalkylaryl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-heteroalkylheteroaryl, which is unsubstituted. In another embodiment, R1 is -L-heteroalkylheteroaryl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heteroalkylheteroaryl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-heteroalkylheteroaryl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula, R1 is -L-heteroalkyl-heterocyclyl, which is unsubstituted. In another embodiment, R1 is -L-heteroalkyl-heterocyclyl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heteroalkyl-heterocyclyl which is unsubstituted, and L is absent. In yet another embodiment, R1 is -L-heteroalkyl-heterocyclyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-heteroalkyl-C3-8cycloalkyl, which is unsubstituted. In another embodiment, R1 is -L-heteroalkyl-C3-8cycloalkyl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heteroalkyl-C3-8cycloalkyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-heteroalkyl-C3-8cycloalkyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-aralkyl, which is unsubstituted. In another embodiment, R1 is -L-aralkyl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-aralkyl which is unsubstituted. In yet another embodiment, R1 is -L-aralkyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-heteroaralkyl, which is unsubstituted. In another embodiment, R1 is -L-heteroaralkyl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heteroaralkyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-heteroaralkyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is -L-heterocyclyl, which is unsubstituted. In another embodiment, R1 is -L-heterocyclyl, which is substituted by one or more independent R3. In a further embodiment, R1 is -L-heterocyclyl which is unsubstituted and L is absent. In yet another embodiment, R1 is -L-heterocyclyl, which is substituted by one or more independent R3, where L is absent.
  • In various embodiments of compounds of Formula I, R1 is a substituent as shown below:
  • Figure US20160235758A1-20160818-C00033
    Figure US20160235758A1-20160818-C00034
    Figure US20160235758A1-20160818-C00035
  • In various embodiments of compounds of Formula I, R2 is hydrogen. In another embodiment, R2 is halogen. In another embodiment, R2 is OH. In another embodiment, R2 is R31. In another embodiment, R2 is CF3. In another embodiment, R2 is OCF3. In another embodiment, R2 is —OR31. In another embodiment, R2 is NR31R32. In another embodiment, R2 is NR34R35. In another embodiment, R2 is —C(O)R31. In another embodiment, R2 is CO2R31. In another embodiment, R2 is —C(═O)NR31R″. In another embodiment, R2 is —C(═O)NR34R35. In another embodiment, R2 is —NO2. In another embodiment, R2 is —CN. In another embodiment, R2 is —S(O)0-2R3. In another embodiment, R2 is —SO2NR31R32. In another embodiment, R2 is —SO2NR34R35. In another embodiment, R2 is —NR31C(═O)R32. In another embodiment, R2 is —NR31C(═O)OR32. In another embodiment, R2 is —NR31C(═O)NR32R33. In another embodiment, R2 is —NR31S(O)0-2R32. In another embodiment, R2 is —C(═S)OR31. In another embodiment, R2 is —C(═O)SR31. In another embodiment, R2 is —NR31C(═NR32)NR33R32. In another embodiment, R2 is —NR31C(═NR32)OR33. In another embodiment, R2 is —NR31C(═NR32)SR33. In another embodiment, R2 is —OC(═O)OR33. In another embodiment, R2 is —OC(═O)NR31R32. In another embodiment, R2 is —OC(═O)SR31. In another embodiment, R2 is —SC(═O)OR31. In another embodiment, R2 is —P(O)OR31OR32. In another embodiment, R2 is —SC(═O)NR31R32. In another embodiment, R2 is monocyclic aryl. In another embodiment, R2 is bicyclic aryl. In another embodiment, R2 is substituted monocyclic aryl. In another embodiment, R2 is heteroaryl. In another embodiment, R2 is C1-4alkyl. In another embodiment, R2 is C1-10alkyl. In another embodiment, R2 is C3-8cycloalkyl. In another embodiment, R2 is C3-8cycloalkyl-C1-10alkyl. In another embodiment, R2 is C1-10alkyl-C3-8cycloalkyl. In another embodiment, R2 is C1-10alkyl-monocyclic aryl. In another embodiment, R2 is C2-10alkyl-monocyclic aryl. In another embodiment, R2 is monocyclic aryl-C2-10alkyl. In another embodiment, R2 is C1-10alkyl-bicyclicaryl. In another embodiment, R2 is bicyclicaryl-C1-10alkyl. In another embodiment, R2 is —C1-10alkylheteroaryl. In another embodiment, R2 is —C1-10alkylheterocyclyl. In another embodiment, R2 is —C2-10alkenyl. In another embodiment, R2 is —C2-10alkynyl. In another embodiment, R2 is C2-10alkenylaryl. In another embodiment, R2 is C2-10alkenylheteroaryl. In another embodiment, R2 is C2-10alkenylheteroalkyl. In another embodiment, R2 is C2-10alkenylheterocycicyl. In another embodiment, R2 is —C2-10alkynylaryl. In another embodiment, R2 is —C2-10alkynylheteroaryl. In another embodiment, R2 is —C2-10alkynylheteroalkyl. In another embodiment, R2 is —C2-10alkynylheterocyclyl. In another embodiment, R2 is —C2-10alkynylC3-8cycloalkyl. In another embodiment, R2 is —C2-10alkynylC3-8cycloalkenyl. In another embodiment, R2 is —C1-10alkoxy-C1-10alkyl. In another embodiment, R2 is —C1-10alkoxy-C2-10alkenyl. In another embodiment, R2 is —C1-10alkoxy-C2-10alkynyl. In another embodiment, R2 is -heterocyclyl C1-10alkyl. In another embodiment, R2 is heterocyclylC2-10alkenyl. In another embodiment, R2 is heterocyclylC2-10alkynyl. In another embodiment, R2 is aryl-C2-10alkyl. In another embodiment, R2 is aryl-C1-10alkyl. In another embodiment, R2 is aryl-C2-10alkenyl. In another embodiment, R2 is aryl-C2-10alkynyl. In another embodiment, R2 is aryl-heterocyclyl. In another embodiment, R2 is heteroaryl-C1-10alkyl. In another embodiment, R2 is heteroaryl-C2-10alkenyl. In another embodiment, R2 is heteroaryl-C2-10alkynyl. In another embodiment, R2 is heteroaryl-C3-8cycloalkyl. In another embodiment, R2 is heteroaryl-heteroalkyl. In another embodiment, R2 is heteroaryl-heterocyclyl.
  • In various embodiments of compounds of Formula I, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is unsubstituted. In various embodiments, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent halo. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OH. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —R31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —CF3. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OCF. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OR31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR34R35. In another embodiment, when R4 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(O)R31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —CO2R31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(═O)NR31R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(═O)NR34R35. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NO2. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —CN. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —S(O)0-2R31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SO2NR31R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SO2NR34R35. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent NR31C(═O)R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═O)OR32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═O)NR32R33. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31S(O)0-2R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(═S)OR31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(═O)SR31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═NR32)NR33R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent, —NR31C(═NR32)OR33. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═NR32)SR33. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)OR33. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)NR31R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)SR31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SC(═O)OR31. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —P(O)OR31OR32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SC(═O)NR31R32. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent alkyl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent heteroalkyl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent alkenyl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent alkynyl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent cycloalkyl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent heterocycloalkyl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent aryl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent arylalkyl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent heteroaryl. In another embodiment, when R2 is bicyclic aryl, monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, monocyclic aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent heteroarylalkyl.
  • In various embodiments of compounds of Formula I, R3 is hydrogen. In another embodiment, R3 is halogen. In another embodiment, R3 is OH. In another embodiment, R3 is R31. In another embodiment, R3 is CF3. In another embodiment, R3 is OCF3. In another embodiment, R3 is —OR31. In another embodiment, R3 is NR31R32. In another embodiment, R3 is NR34R35. In another embodiment, R3 is —C(O)R31. In another embodiment, R3 is CO2R31. In another embodiment, R3 is —C(═O)NR31R32. In another embodiment, R3 is —C(═O)NR34R35. In another embodiment, R3 is —NO2. In another embodiment, R3 is —CN. In another embodiment, R3 is —S(O)0-2R3. In another embodiment, R3 is SO2NR31R32. In another embodiment, R3 is —SO2NR34R35. In another embodiment, R3 is —NR31C(═O)R32. In another embodiment, R3 is —NR31C(═O)OR32. In another embodiment, R3 is —NR31C(═O)NR32R33. In another embodiment, R3 is NR31S(O)0-2R32. In another embodiment, R3 is —C(═S)OR31. In another embodiment, R3 is —C(═O)SR31. In another embodiment, R3 is —NR31C(═NR32)NR33R32. In another embodiment, R3 is —NR31C(═NR32)OR33. In another embodiment, R3 is —NR31C(═NR32)SR33. In another embodiment, R3 is —OC(═O)OR33. In another embodiment, R3 is —OC(═O)NR31R32. In another embodiment, R3 is —OC(═O)SR31. In another embodiment, R3 is —SC(═O)OR31. In another embodiment, R3 is —P(O)OR31OR32. In another embodiment, R3 is —SC(═O)NR31R32. In another embodiment, R3 is aryl. In another embodiment, R2 is heteroaryl. In another embodiment, R3 is C1-4alkyl. In another embodiment, R3 is C1-10alkyl. In another embodiment, R3 is C3-8cycloalkyl. In another embodiment, R3 is C3-8cycloalkyl-C1-10alkyl. In another embodiment, R3 is —C1-10alkyl-C3-8cycloalkyl. In another embodiment, R3 is C2-10alkyl-monocyclic aryl. In another embodiment, R3 is monocyclic aryl-C2-10alkyl. In another embodiment, R3 is C1-10alkyl-bicyclicaryl. In another embodiment, R3 is bicyclicaryl-C1-10alkyl. In another embodiment, R3 is C1-10alkylheteroaryl. In another embodiment, R3 is C1-10alkylheterocyclyl. In another embodiment, R3 is C2-10alkenyl. In another embodiment, R3 is C2-10alkynyl. In another embodiment, R3 is C2-10alkenylaryl. In another embodiment, R3 is C2-10alkenylheteroaryl. In another embodiment, R3 is C2-10alkenylheteroalkyl. In another embodiment, R3 is C2-10alkenylheterocycicyl. In another embodiment, R3 is —C2-10alkynylaryl. In another embodiment, R3 is —C2-10alkynylheteroaryl. In another embodiment, R3 is —C2-10alkynylheteroalkyl. In another embodiment, R3 is C2-10alkynylheterocyclyl. In another embodiment, R3 is —C2-10alkynylC3-8cycloalkyl. In another embodiment, R3 is C2-10alkynylC3-8cycloalkenyl. In another embodiment, R3 is —C1-10alkoxy C1-10alkyl. In another embodiment, R3 is C1-10alkoxy-C2-10alkenyl. In another embodiment, R3 is —C1-10alkoxy-C2-10alkynyl. In another embodiment, R3 is heterocyclyl-C1-10alkyl. In another embodiment, R3 is -heterocyclylC2-10alkenyl. In another embodiment, R3 is heterocyclyl-C2-10alkynyl. In another embodiment, R3 is aryl-C1-10alkyl. In another embodiment, R3 is aryl-C2-10alkenyl. In another embodiment, R3 is aryl-C2-10alkynyl. In another embodiment, R3 is aryl-heterocyclyl. In another embodiment, R3 is heteroaryl-C1-10alkyl. In another embodiment, R3 is heteroaryl-C2-10alkenyl. In another embodiment, R3 is heteroaryl-C2-10alkynyl. In another embodiment, R3 is heteroaryl-C3-8cycloalkyl. In another embodiment, R3 is heteroaryl-heteroalkyl. In another embodiment, R3 is heteroaryl-heterocyclyl.
  • In various embodiments of compounds of Formula I, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is unsubstituted. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent halo. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —OH. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —R31. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —CF3. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —OCF. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent OR31. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent NR31R32. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent NR34R35. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent C(O)R31. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl or heteroalkyl, it is substituted with one or more independent CO2R31. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent C(═O)NR31R32. In another embodiment, when R3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent C(═O)NR34R35. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —NO2. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent CN. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent S(O)0-2R31. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent SO2NR31R32. In another embodiment, when R3 is aryl, heteroaryl, C3-8cycloalkyl, C3-8cycloalkyl-heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent SO2NR34R35. In another embodiment, when R3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl. C2-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent NR31C(═O)R32. In another embodiment, when R3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═O)OR32. In another embodiment, when R3 is aryl, heteroaryl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═O)NR32R33. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31S(O)0-2R32. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent C(═S)OR31. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent C(═O)SR31. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —NR31C(═NR32)NR33R32. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent, —NR31C(═NR32)OR33. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent NR31C(═NR32)SR33. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)OR33. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent OC(═O)NR31R32. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)SR31. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SC(═O)OR31. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —P(O)OR31OR32. In another embodiment, when R3 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SC(═O)NR31R32.
  • In various embodiments of compounds of Formula I, R4 is hydrogen. In another embodiment, R4 is halogen. In another embodiment, R4 is OH. In another embodiment, R4 is R31. In another embodiment, R4 is CF3. In another embodiment, R4 is OCF3. In another embodiment, R4 is —OR31. In another embodiment, R4 is NR31R32. In another embodiment, R4 is NR34R35. In another embodiment, R4 is —C(O)R31. In another embodiment, R4 is —CO2R31. In another embodiment, R4 is —C(═O)NR31R32. In another embodiment, R4 is —C(═O)NR34R35. In another embodiment, R4 is —NO2. In another embodiment, R4 is —CN. In another embodiment, R4 is —S(O)0-2R3. In another embodiment, R4 is —SO2NR31R32. In another embodiment, R4 is —SO2NR34R35. In another embodiment, R4 is —NR31C(═O)R32. In another embodiment, R4 is —NR31C(═O)OR32. In another embodiment, R4 is —NR31C(═O)NR32R33. In another embodiment, R4 is —NR31S(O)0-2R32. In another embodiment, R4 is —C(═S)OR31. In another embodiment, R4 is —C(═O)SR31. In another embodiment, R4 is —NR31C(═NR32)NR33R32. In another embodiment, R4 is —NR31C(═NR32)OR33. In another embodiment, R4 is —NR31C(═NR32)SR33. In another embodiment, R4 is —OC(═O)OR33. In another embodiment, R4 is —OC(═O)NR31R32. In another embodiment, R4 is —OC(═O)SR31. In another embodiment, R4 is —SC(═O)OR31. In another embodiment, R4 is —P(O)OR31OR32. In another embodiment, R4 is —SC(═O)NR31R32. In another embodiment, R4 is aryl. In another embodiment, R4 is heteroaryl. In another embodiment, R4 is C1-4alkyl. In another embodiment, R4 is C1-10alkyl. In another embodiment, R4 is C3-8cycloalkyl. In another embodiment, R4 is C1-10alkyl-C3-8cycloalkyl. In another embodiment, R4 is C1-10alkylaryl. In another embodiment, R4 is C1-10alkylheteroaryl. In another embodiment, R4 is C1-10alkylheterocyclyl. In another embodiment, R4 is C2-10alkenyl. In another embodiment, R4 is C2-10alkynyl. In another embodiment, R4 is C2-10alkynyl-C3-8cycloalkyl. R4 is C2-10alkenyl-C3-8cycloalkyl. In another embodiment, R4 is C2-10alkenylaryl. In another embodiment, R4 is C2-10alkenyl-heteroaryl. In another embodiment, R4 is C2-10alkenylheteroalkyl. In another embodiment, R4 is C2-10alkenylheterocycicyl. In another embodiment, R4 is —C2-10alkynylaryl. In another embodiment, R4 is C2-10alkynylheteroaryl. In another embodiment, R4 is C2-10alkynylheteroalkyl. In another embodiment, R4 is C2-10alkynylheterocyclyl. In another embodiment, R4 is C2-10alkynylC3-8cycloalkyl. In another embodiment, R4 is heterocyclyl C1-10alkyl. In another embodiment, R4 is heterocyclylC2-10alkenyl. In another embodiment, R4 is heterocyclyl-C2-10alkynyl. In another embodiment, R4 is aryl-C1-10alkyl. In another embodiment, R4 is aryl-C2-10alkenyl. In another embodiment, R4 is aryl-C2-10alkynyl. In another embodiment, R4 is aryl-heterocyclyl. In another embodiment, R4 is heteroaryl-C1-10alkyl. In another embodiment, R4 is heteroaryl-C2-10alkenyl. In another embodiment, R4 is heteroaryl-C2-10alkynyl. In another embodiment, R4 is C3-8cycloalkyl-C1-10alkyl. In another embodiment, R4 is C3-8cycloalkyl-C2-10alkenyl. In another embodiment, R4 is C3-8cycloalkyl-C2-10alkynyl.
  • In various embodiments of compounds of Formula I, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is unsubstituted. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent halo. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —OH. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —R31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent CF3. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —OCF. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent OR31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent NR31R32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent NR34R35. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —C(O)R31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent CO2R31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —C(═O)NR31R32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(═O)NR34R35. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —NO2. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —CN. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —S(O)0-2R31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —SO2NR31R32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent SO2NR34R35. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent NR31C(═O)R32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent NR31C(═O)OR32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═O)NR32R33. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31S(O)0-2R32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(═S)OR31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —C(═O)SR31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —NR31C(═NR32)NR33R32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl C2-10alkyl heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent, —NR31C(═NR32)OR33. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —NR31C(═NR32)SR33. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)OR33. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)NR31R32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —OC(═O)SR31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SC(═O)OR31. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, heterocyclyl, heterocyclyl C1-10alkyl, or heteroalkyl, it is substituted with one or more independent —P(O)OR31OR32. In another embodiment, when R4 is aryl, heteroaryl, C1-10alkyl, cycloalkyl, heterocyclyl, heteroalkyl, C2-10alkenyl, C2-10alkynyl, aryl-C2-10alkyl, heterocyclyl C1-10alkyl, or C3-8cycloalkyl-C1-10alkyl, it is substituted with one or more independent —SC(═O)NR31R32.
  • In various embodiments of compounds of Formula I, R5 is hydrogen. In another embodiment, R5 is halogen. In another embodiment, R5 is —OH. In another embodiment, R5 is —R31. In another embodiment, R5 is —CF3. In another embodiment, R5 is —OCF3. In another embodiment, R5 is —OR31. In another embodiment, R5 is —NR31R32. In another embodiment, R5 is —NR34R35. In another embodiment, R5 is —C(O)R31. In another embodiment, R5 is —CO2R31. In another embodiment, R5 is —C(═O)NR31R32. In another embodiment, R5 is —C(═O)NR34R35. In another embodiment, R5 is —NO2. In another embodiment, R5 is —CN. In another embodiment, R5 is —S(O)0-2R31. In another embodiment, R5 is —SO2NR31R32. In another embodiment, R5 is —SO2NR34R35. In another embodiment, R5 is —NR31C(═O)R32. In another embodiment, R5 is —NR31C(═O)OR32. In another embodiment, R5 is —NR31C(═O)NR32R33. In another embodiment, R5 is —NR31S(O)0-2R32. In another embodiment, R5 is —C(═S)OR31. In another embodiment, R5 is —C(═O)SR31. In another embodiment, R5 is —NR31C(═NR32)NR33R32. In another embodiment, R5 is —NR31C(═NR32)OR33. In another embodiment, R5 is —NR31C(═NR32)SR33. In another embodiment, R5 is —OC(═O)OR33. In another embodiment, R5 is —OC(═O)NR31R32. In another embodiment, R5 is —OC(═O)SR31. In another embodiment, R5 is —SC(═O)OR31. In another embodiment, R5 is —P(O)OR31OR32. In another embodiment, R5 is or —SC(═O)NR31R32.
  • In various embodiments of compounds of Formula I, R7 is hydrogen. In another embodiment, R7 is unsubstituted C1-10alkyl. In another embodiment, R7 is unsubstituted C2-10alkenyl. In another embodiment, R7 is unsubstituted aryl. In another embodiment, R7 is unsubstituted heteroaryl. In another embodiment, R7 is unsubstituted heterocyclyl. In another embodiment, R7 is unsubstituted C3-10cycloalkyl. In another embodiment, R7 is C1-10alkyl substituted by one or more independent R6. In another embodiment, R7 is C2-10alkenyl substituted by one or more independent R6. In another embodiment, R7 is aryl substituted by one or more independent R6. In another embodiment, R7 is heteroaryl substituted by one or more independent R6. In another embodiment, R7 is heterocycly substituted by one or more independent R6. In another embodiment, R7 is C3-10cycloalkyl substituted by one or more independent R6.
  • In various embodiments of compounds of Formula I, R8 is hydrogen. In another embodiment, R8 is unsubstituted C1-10alkyl. In another embodiment, R8 is unsubstituted C2-10alkenyl. In another embodiment, R8 is unsubstituted aryl. In another embodiment, R8 is unsubstituted heteroaryl. In another embodiment, R8 is unsubstituted heterocyclyl. In another embodiment, R8 is unsubstituted C3-10cycloalkyl. In another embodiment, R8 is C1-10alkyl substituted by one or more independent R6. In another embodiment, R8 is C2-10alkenyl substituted by one or more independent R6. In another embodiment, R8 is aryl substituted by one or more independent R6. In another embodiment, R8 is heteroaryl substituted by one or more independent R6. In another embodiment, R8 is heterocyclyl substituted by one or more independent R6. In another embodiment, leis C3-10cycloalkyl substituted by one or more independent R6.
  • In various embodiments of compounds of Formula I, R6 is halo, In another embodiment, R6 is —OR31. In another embodiment, R6 is —SH. In another embodiment, R6 is NH2. In another embodiment, R6 is —NR34R35. In another embodiment, R6 is NR31R32. In another embodiment, R6 is —CO2R31. In another embodiment, R6 is CO2aryl. In another embodiment, R6 is —C(═O)NR31R32. In another embodiment, R6 is C(═O) NR34R35. In another embodiment, R6 is NO2. In another embodiment, R6 is —CN. In another embodiment, R6 is —S(O)0-2 C1-10alkyl. In another embodiment, R6 is —S(O)0-2aryl. In another embodiment, R6 is —SO2NR34R35. In another embodiment, R6 is —SO2NR31R32. In another embodiment, R6 is C1-10alkyl. In another embodiment, R6 is C2-10alkenyl. In another embodiment, R6 is C2-10alkynyl. In another embodiment, R6 is unsubstituted aryl-C1-10alkyl. In another embodiment, R6 is unsubstituted aryl-C2-10alkenyl. In another embodiment, R6 is unsubstituted aryl-C2-10alkynyl. In another embodiment, R6 is unsubstituted heteroaryl-C1-10alkyl. In another embodiment, R6 is unsubstituted heteroaryl-C2-10alkenyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent halo. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent cyano. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent nitro. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —OC1-10alkyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C1-10alkyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C2-10alkenyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C2-10alkynyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent (halo)C1-10alkyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent (halo)C2-10alkenyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent (halo)C2-10alkynyl. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —COOH. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C(═O)NR31R32. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C(═O) NR34R35. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent SO2NR34R35. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —SO2 NR31R32. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —NR31R32. In another embodiment, R6 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —NR34R35.
  • In various embodiments of compounds of Formula I, R9 is H. In another embodiment, R9 is halo. In another embodiment, R9 is —OR31. In another embodiment, R9 is —SH. In another embodiment, R9 is NH2. In another embodiment, R9 is —NR34R35. In another embodiment, R9 is —NR31R32. In another embodiment, R9 is —CO2R31. In another embodiment, R9 is CO2aryl. In another embodiment, R9 is —C(═O)NR31R32. In another embodiment, R9 is C(═O) NR34R35. In another embodiment, R9 is NO2. In another embodiment, R9 is CN. In another embodiment, R9 is —S(O)0-2 C1-10alkyl. In another embodiment, R9 is —S(O)0-2aryl. In another embodiment, R9 is SO2NR34R35. In another embodiment, R9 is —SO2NR31R32. In another embodiment, R9 is C1-10alkyl. In another embodiment, R9 is C2-10alkenyl. In another embodiment, R9 is C2-10alkynyl. In another embodiment, R9 is unsubstituted aryl-C1-10alkyl. In another embodiment, R9 is unsubstituted aryl-C2-10alkenyl. In another embodiment, R9 is unsubstituted aryl-C2-10alkynyl. In another embodiment, R9 is unsubstituted heteroaryl-C1-10alkyl. In another embodiment, R9 is unsubstituted heteroaryl-C2-10alkenyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent halo. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent cyano. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent nitro. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —OC1-10alkyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C1-10alkyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C2-10alkenyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C2-10alkynyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent -(halo)C1-10alkyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent -(halo)C2-10alkenyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent -(halo)C2-10alkynyl. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —COOH. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C(═O)NR31R32. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —C(═O) NR34R35. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —SO2NR34R35. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —SO2 NR31R32. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —NR31R32. In another embodiment, R9 is aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, or heteroaryl-C2-10alkenyl substituted by one or more independent —NR34R35.
  • In various embodiments of compounds of Formula I, R31 is H. In some embodiments, R31 is unsubstituted C1-10alkyl. In some embodiments, R31 is substituted C1-10alkyl. In some embodiments, R31 is C1-10alkyl substituted with one or more aryl. In some embodiments, R31 is C1-10alkyl substituted with one or more heteroalkyl. In some embodiments, R31 is C1-10alkyl substituted with one or more heterocyclyl. In some embodiments, R31 is C1-10alkyl substituted with one or more heteroaryl. In some embodiments, when R31 is C1-10alkyl substituted with one or more aryl, each of said aryl substituents is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2-aryl, —C(═O)N(C1-10alkyl)(—C(═O)NH(—C(═O)NR34R35, —C(═O)NH2, —OCF3, —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R31 is C1-10alkyl substituted with one or more heteroalkyl, each of said heteroalkyl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2-aryl, —C(═O)N(C1-10alkyl)(—C(═O)NH(—C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35 substituents. In some embodiments, when R31 is C1-10alkyl substituted with one or more heterocyclyl, each of said heterocyclyl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2-10C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R31 is C1-10alkyl substituted with one or more heteroaryl, each of said heteroaryl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R31 is substituted C1-10alkyl, it is substituted by a combination of aryl, heteroalkyl, heterocyclyl, or heteroaryl groups.
  • In various embodiments of compounds of Formula I, R32 is H. In some embodiments, R32 is unsubstituted C1-10alkyl. In some embodiments, R32 is substituted C1-10alkyl. In some embodiments, R32 is C1-10alkyl substituted with one or more aryl. In some embodiments, R32 is C1-10alkyl substituted with one or more heteroalkyl. In some embodiments, R32 is C1-10alkyl substituted with one or more heterocyclyl. In some embodiments, R32 is C1-10alkyl substituted with one or more heteroaryl. In some embodiments, when R32 is C1-10alkyl substituted with one or more aryl, each of said aryl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R32 is C1-10alkyl substituted with one or more heteroalkyl, each of said heteroalkyl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(—NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R32 is C1-10alkyl substituted with one or more heterocyclyl, each of said heterocyclyl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R32 is C1-10alkyl substituted with one or more heteroaryl, each of said heteroaryl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2-10C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R32 is substituted C1-10alkyl, it is substituted by a combination of aryl, heteroalkyl, heterocyclyl, or heteroaryl groups.
  • In various embodiments of compounds of Formula I, R33 is unsubstituted C1-10alkyl. In some embodiments, R33 is substituted C1-10alkyl. In some embodiments, R33 is C1-10alkyl substituted with one or more aryl. In some embodiments, R33 is C1-10alkyl substituted with one or more heteroalkyl. In some embodiments, R33 is C1-10alkyl substituted with one or more heterocyclyl. In some embodiments, R33 is C1-10alkyl substituted with one or more heteroaryl. In some embodiments, when R33 is C1-10alkyl substituted with one or more aryl, each of said aryl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2N(C1-10alkyl)(C1-10alkyl), —SO2NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R33 is C1-10alkyl substituted with one or more heteroalkyl, each of said heteroalkyl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R33 is C1-10alkyl substituted with one or more heterocyclyl, each of said heterocyclyl group is unsubstituted or substituted with one or more halo, —OH, —C10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R33 is C1-10alkyl substituted with one or more heteroaryl, each of said heteroaryl group is unsubstituted or substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2-10C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35. In some embodiments, when R33 is substituted C1-10alkyl, it is substituted by a combination of aryl, heteroalkyl, heterocyclyl, or heteroaryl groups.
  • In various embodiments of compounds of Formula I, R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, heteroaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen.
  • In some embodiments, the R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form:
  • Figure US20160235758A1-20160818-C00036
  • In another embodiment, X1 is C≡NH2.
  • In various embodiments, X1 is C≡NH—R4, where —NH—R4 is:
  • Figure US20160235758A1-20160818-C00037
    Figure US20160235758A1-20160818-C00038
  • In one embodiment, the invention provides an inhibitor of Formula I-C1 where R5 is H. In another embodiment, the invention provides an inhibitor of Formula I-C2 where R5 is H.
  • In some embodiments, the invention provides an inhibitor of Formula I-C1a:
  • Figure US20160235758A1-20160818-C00039
  • or a pharmaceutically acceptable salt thereof wherein:
  • E2 is H;
  • X1 and X2 are N;
  • R1 is L-C1-10alkyl, -L-C3-8cycloalkyl, -L-C1-10alkylheterocyclyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • R3 is hydrogen, —OH, —OR31, —NR31R32, —C(O)R31, —C(═O)NR31R32, —C(═O)NR34R35, aryl, heteroaryl, C1-4alkyl, C1-10alkyl, C3-8cycloalkyl, or heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, or heterocyclyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • —(W2)k— is —NH—, —N(H)C(O)— or —N(H)S(O)2—; R2 is hydrogen, halogen, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, bicyclic aryl, substituted monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C2-10alkyl-monocyclic aryl, monocyclic aryl-C2-10alkyl, C1-10alkylbicycloaryl, bicycloaryl-C1-10alkyl, substituted C1-10alkylaryl, substituted aryl-C1-10alkyl, C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxyC2-10alkenyl, C1-10alkoxyC2-10alkynyl, heterocyclyl, heterocyclyl C1-10alkyl, heterocyclylC2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, R31—C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R31, R32, and R33, in each instance, are independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted; and
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, heteroaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen.
  • In another aspect, an inhibitor of Formula I-C1 is a compound of Formula I-C1a:
  • Figure US20160235758A1-20160818-C00040
  • or a pharmaceutically acceptable salt thereof, wherein: E2 is —H; X1 is CH and X2 is N;
  • R1 is -L-C1-10alkyl, -L-C3-8cycloalkyl, -L-C1-10alkylheterocyclyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • R3 is hydrogen, —OH, —OR31, —NR31R32, —C(O)R31, —C(═O)NR31R32, —C(═O)NR34R35, aryl, heteroaryl, C1-4alkyl, C1-10alkyl, C3-8cycloalkyl, or heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, or heterocyclyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • —(W2)k— is —NH—, —N(H)C(O)— or —N(H)S(O)2—;
  • R2 is hydrogen, halogen, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, bicyclic aryl, substituted monocyclic aryl, heteroaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C2-10alkyl-monocyclic aryl, monocyclic aryl-C2-10alkyl, C1-10alkylbicycloaryl, bicycloaryl-C1-10alkyl, substituted C1-10alkylaryl, substituted aryl-C1-10alkyl, C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, heterocyclyl, heterocyclyl C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R31, R32, and R33, in each instance, are independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted; and
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, heteroaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen.
  • The invention further provides a compound which is an mTor inhibitor, wherein the compound has the Formula I-A:
  • Figure US20160235758A1-20160818-C00041
  • or a pharmaceutically acceptable salt thereof, wherein:
  • X1 is N or C-E1, X2 is N, X3 is C, and X4 is C—R9 or N; or X1 is N or C-E1, X2 is C, X3 is N, and X4 is C—R9 or N;
  • R1 is —H, -L-C1-10alkyl, -L-C3-8cycloalkyl, -L-C1-10alkyl-C3-8cycloalkyl, -L-aryl, -L-heteroaryl, -L-C1-10alkylaryl, -L-C1-10alkylheteroaryl, -L-C1-10alkylheterocyclyl, -L-C2-10alkenyl, -L-C2-10alkynyl, -L-C2-10alkenyl-C3-8cycloalkyl, -L-C2-10alkynyl-C3-8cycloalkyl, -L-heteroalkyl, -L-heteroalkylaryl, -L-heteroalkylheteroaryl, -L-heteroalkyl-heterocyclyl, -L-heteroalkyl-C3-8cycloalkyl, -L-aralkyl, -L-heteroaralkyl, or -L-heterocyclyl, each of which is unsubstituted or is substituted by one or more independent R3;
  • L is absent, —(C═O)—, —C(═O)O—, —C(═O) N(R31)—, —S—, —S(O)—, —S(O)2—, —S(O)2N(R31)—, or —N(R31)—;
  • M1 is benzothiazolyl substituted with —(W2)k—R2;
  • k is 0 or 1;
  • E1 and E2 are independently —(W1)j—R4;
  • j, in each instance (i.e., in E1 or j in E2), is independently 0 or 1
  • W1 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • W2 is —O—, —S(O)0-2-, —C(O)—, —C(O)N(R7)—, —N(R7)C(O)—, —N(R7)C(O)N(R8)—, —N(R7)S(O)—, —N(R7)S(O)2—, —C(O)O—, —CH(R7)N(C(O)OR8)—, —CH(R7)N(C(O)R8)—, —CH(R7)N(SO2R8)—, —CH(R7)N(R8)—, —CH(R7)C(O)N(R8)—, —CH(R7)N(R8)C(O)—, —CH(R7)N(R8)S(O)—, or —CH(R7)N(R8)S(O)2—;
  • R2 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32, aryl (e.g. bicyclic aryl, unsubstituted aryl, or substituted monocyclic aryl), heteroaryl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl (e.g. C2-10alkyl-monocyclic aryl, C1-10alkyl-substituted monocyclic aryl, or C1-10alkylbicycloaryl), C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl, C1-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycicyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heteroalkyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl (e.g. monocyclic aryl-C2-10alkyl, substituted monocyclic aryl-C1-10alkyl, or bicycloaryl-C1-10alkyl), aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said bicyclic aryl or heteroaryl moiety is unsubstituted, or wherein each of bicyclic aryl, heteroaryl moiety or monocyclic aryl moiety is substituted with one or more independent alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31 S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —NR34R35, or —C(═O)NR31R32;
  • R3 and R4 are independently hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31 S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R32, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, —SC(═O)NR31R32, aryl, heteroaryl, C1-4 alkyl, C1-10alkyl, C3-8cycloalkyl, C1-10alkyl-C3-8cycloalkyl, C3-8cycloalkyl-C1-10alkyl, C3-8cycloalkyl-C2-10alkenyl, C3-8cycloalkyl-C2-10alkynyl, C1-10alkyl-C2-10alkenyl, C1-10alkyl-C2-10alkynyl, C1-10alkylaryl, C1-10alkylheteroaryl, C1-10alkylheterocyclyl, C2-10alkenyl, C2-10alkynyl, C2-10alkenyl-C1-10alkyl, C2-10alkynyl-C1-10alkyl, C2-10alkenylaryl, C2-10alkenylheteroaryl, C2-10alkenylheteroalkyl, C2-10alkenylheterocycleyl, C2-10alkenyl-C3-8cycloalkyl, C2-10alkynyl-C3-8cycloalkyl, C2-10alkynylaryl, C2-10alkynylheteroaryl, C2-10alkynylheteroalkyl, C2-10alkynylheterocyclyl, C2-10alkynyl-C3-8cycloalkenyl, C1-10alkoxy C1-10alkyl, C1-10alkoxy-C2-10alkenyl, C1-10alkoxy-C2-10alkynyl, heterocyclyl, heterocyclyl-C1-10alkyl, heterocyclyl-C2-10alkenyl, heterocyclyl-C2-10alkynyl, aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, aryl-heterocyclyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, heteroaryl-C3-8cycloalkyl, heteroalkyl, heteroaryl-heteroalkyl, or heteroaryl-heterocyclyl, wherein each of said aryl or heteroaryl moiety is unsubstituted or is substituted with one or more independent halo, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR—NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R32, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32, and wherein each of said alkyl, cycloalkyl, heterocyclyl, or heteroalkyl moiety is unsubstituted or is substituted with one or more halo, —OH, —R31, —CF3, —OCF3, —OR31, —O-aryl, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR34R35, or —C(═O)NR31R32;
  • R5 is hydrogen, halogen, —OH, —R31, —CF3, —OCF3, —OR31, —NR31R32, —NR34R35, —C(O)R31, —CO2R31, —C(═O)NR31R32, —C(═O)NR34R35, —NO2, —CN, —S(O)0-2R31, —SO2NR31R32, —SO2NR34R35, —NR31C(═O)R32, —NR31C(═O)OR32, —NR31C(═O)NR32R33, —NR31S(O)0-2R32, —C(═S)OR31, —C(═O)SR31, —NR31C(═NR32)NR33R″, —NR31C(═NR32)OR33, —NR31C(═NR32)SR33, —OC(═O)OR33, —OC(═O)NR31R″, —OC(═O)SR31, —SC(═O)OR31, —P(O)OR31OR32, or —SC(═O)NR31R32;
  • R31, R32, and R33, in each instance, are independently H or C1-10alkyl, wherein the C1-10alkyl is unsubstituted or is substituted with one or more aryl, heteroalkyl, heterocyclyl, or heteroaryl group, wherein each of said aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more halo, —OH, —C1-10alkyl, —CF3, —O-aryl, —OCF3, —OC1-10alkyl, —NH2, —N(C1-10alkyl)(C1-10alkyl), —NH(C1-10alkyl), —NH(aryl), —NR34R35, —C(O)(C1-10alkyl), —C(O)(C1-10alkyl-aryl), —C(O)(aryl), —CO2—C1-10alkyl, —CO2—C1-10alkylaryl, —CO2-aryl, —C(═O)N(C1-10alkyl)(C1-10alkyl), —C(═O)NH(C1-10alkyl), —C(═O)NR34R35, —C(═O)NH2, —OCF3, —O(C1-10alkyl), —O-aryl, —N(aryl)(C1-10alkyl), —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2 C1-10alkylaryl, —S(O)0-2 aryl, —SO2N(aryl), —SO2 N(C1-10alkyl)(C1-10alkyl), —SO2 NH(C1-10alkyl) or —SO2NR34R35;
  • R34 and R35 in —NR34R35, —C(═O)NR34R35, or —SO2NR34R35, are taken together with the nitrogen atom to which they are attached to form a 3-10 membered saturated or unsaturated ring; wherein said ring is independently unsubstituted or is substituted by one or more —NR31R32, hydroxyl, halogen, oxo, aryl, heteroaryl, C1-6alkyl, or O-aryl, and wherein said 3-10 membered saturated or unsaturated ring independently contains 0, 1, or 2 more heteroatoms in addition to the nitrogen atom;
  • R7′ and R8′ are each independently hydrogen, C1-10alkyl, C2-10alkenyl, aryl, heteroaryl, heterocyclyl or C3-10cycloalkyl, each of which except for hydrogen is unsubstituted or is substituted by one or more independent R6;
  • R6 is halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or —NR34R35; and
  • R9 is H, halo, —OR31, —SH, —NH2, —NR34R35, —NR31R32, —CO2R31, —CO2aryl, —C(═O)NR31R32, C(═O)NR34R35, —NO2, —CN, —S(O)0-2 C1-10alkyl, —S(O)0-2aryl, —SO2NR34R35, —SO2NR31R32, C1-10alkyl, C2-10alkenyl, C2-10alkynyl; aryl-C1-10alkyl, aryl-C2-10alkenyl, aryl-C2-10alkynyl, heteroaryl-C1-10alkyl, heteroaryl-C2-10alkenyl, heteroaryl-C2-10alkynyl, wherein each of said alkyl, alkenyl, alkynyl, aryl, heteroalkyl, heterocyclyl, or heteroaryl group is unsubstituted or is substituted with one or more independent halo, cyano, nitro, —OC1-10alkyl, C1-10alkyl, C2-10alkenyl, C2-10alkynyl, haloC1-10alkyl, haloC2-10alkenyl, haloC2-10alkynyl, —COOH, —C(═O)NR31R32, —C(═O)NR34R35, —SO2NR34R35, —SO2 NR31R32, —NR31R32, or —NR34R35.
  • In some embodiments, X4 is C—R9.
  • The invention also provides an inhibitor as defined above, wherein the compound is of Formula I-B:
  • Figure US20160235758A1-20160818-C00042
  • or a pharmaceutically acceptable salt thereof, and wherein the substituents are as defined above.
  • In various embodiments the compound of Formula I-B or its pharmaceutically acceptable salt thereof, is an inhibitor having the structure of Formula I-B1 or Formula I-B2:
  • Figure US20160235758A1-20160818-C00043
  • or a pharmaceutically acceptable salt thereof.
  • In various embodiments of Formula I-B1, X1 is N and X2 is N. In other embodiments, X1 is C-E1 and X2 is N. In yet other embodiments, X1 is NH and X2 is C. In further embodiments, X1 is CH-E1 and X2 is C.
  • In various embodiments of Formula I-B2, X1 is N and X2 is C. In further embodiments, X1 is C-E1 and X2 is C.
  • In various embodiments, X1 is C—(W1)j—R4, where j is 0.
  • In another embodiment, X1 is CH. In yet another embodiment, X1 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of X1, it is C—(W1)j—R4. In various embodiments of X1, j is 1, and W1 is —O—. In various embodiments of X1, j is 1, and W1 is NR′—. In various embodiments of X1, j is 1, and W1 is —NH—. In various embodiments of X1, j is 1, and W1 is —S(O)0-2. In various embodiments of X1, j is 1, and W1 is —C(O)—. In various embodiments of X1, j is 1, and W1 is —C(O)N(R7). In various embodiments of X1, j is 1, and W1 is —N(R7)C(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of X1, j is 1, and W1 is —N(R7)S(O)2—. In various embodiments of X1, j is 1, and W1 is —C(O)O—. In various embodiments of X1, j is 1, and W1 is CH(R7)N(C(O)OR8). In various embodiments of X1, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)C(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of X1, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In another embodiment, X1 is CH2. In yet another embodiment, X1 is CH-halogen, where halogen is Cl, F, Br, or I.
  • In another embodiment, X1 is N.
  • In various embodiments, X2 is N. In other embodiments, X2 is C.
  • In various embodiments, E2 is —(W1)j—R4, where j is 0.
  • In another embodiment, E2 is CH. In yet another embodiment, E2 is C-halogen, where halogen is Cl, F, Br, or I.
  • In various embodiments of E2, it is —(W1), —R4. In various embodiments of E2, j is 1, and W1 is —O—. In various embodiments of E2, j is 1, and W1 is —NR′—. In various embodiments of E2, j is 1, and W1 is —NH—. In various embodiments of E2, j is 1, and W1 is —S(O)0-2-. In various embodiments of E2, j is 1, and W1 is —C(O)—. In various embodiments of E2, j is 1, and W1 is —C(O)N(R7)—. In various embodiments of E2, j is 1, and W1 is —N(R7)C(O)—. In various embodiments of E2, j is 1, and W1 is —N(R7)S(O)—. In various embodiments of E2, j is 1, and W1 is —N(R7)S(O)2—. In various embodiments of E2, j is 1, and W1 is —C(O)O—. In various embodiments of E2, j is 1, and W1 is CH(R7)N(C(O)OR8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(C(O)R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(SO2R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)C(O)N(R8)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)C(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)S(O)—. In various embodiments of E2, j is 1, and W1 is —CH(R7)N(R8)S(O)2—.
  • In various embodiments of Formula I-A, I—B, I—B1 and I-B2, M1 is:
  • Figure US20160235758A1-20160818-C00044
  • In some embodiments of the invention, M1 is benzothiazolyl substituted with —(W2)k—R2. W2 can be —O—, —S(O)0-2-(including but not limited to —S—, —S(O)—, and —S(O)2—), —C(O)—, or —C(O)O—. In other embodiments, W1 is —NR6— or —CH(R6)N(R7)—, wherein R6 and R7 are each independently hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), unsubstituted or substituted C2-C10alkenyl (including but not limited to alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl). Additionally when W2 is —NR6— or —CH(R6)N(R7)—, R6 and R7 are each independently unsubstituted or substituted aryl (including phenyl and naphthtyl). In yet other embodiments, when W2 is —NR6 or —CH(R6)N(R7)—, R6 and R7 are each independently heteroaryl, wherein the heteroaryl is unsubstituted or substituted. R6 and R7 heteroaryl is monocyclic heteroaryl, and includes but is not limited to imidazolyl, pyrrolyl, oxazolyl, thiazolyl, and pyridinyl. In some other embodiments, when W2 is —NR6— or —CH(R6)N(R7)—, R6 and R7 are each independently unsubstituted or substituted heterocyclyl (which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl) or unsubstituted or substituted C3-8cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl). Non limiting exemplary W2 include NH—, —N(cyclopropyl), and N(4-N-piperidinyl).
  • For example, exemplary mTor inhibitors of the invention have the Formulas:
  • Figure US20160235758A1-20160818-C00045
  • Reaction Schemes—mTor Inhibitor Compounds
  • The mTor inhibitor compounds disclosed herein may be prepared by the routes described below. Materials used herein are either commercially available or prepared by synthetic methods generally known in the art. These schemes are not limited to the compounds listed or by any particular substituents employed for illustrative purposes. Numbering does not necessarily correspond to that of claims or other tables.
  • Figure US20160235758A1-20160818-C00046
  • In one embodiment, compounds are synthesized by condensing a functionalized heterocycle A-1 with formamide, to provide a pyrazolopyrimidine A-2. The pyrazolopyrimidine is treated with N-iodosuccinimide, which introduces an iodo substituent in the pyrazole ring as in A-3. The R1 substituent is introduced by reacting the pyrazolopyrimidine A3 with a compound of Formula R1-Lg in the presence of a base such as potassium carbonate to produce a compound of Formula A-4. Other bases that are suitable for use in this step include but are not limited to sodium hydride and potassium t-butoxide. The compound of Formula R1-Lg has a moiety R1 as defined for R1 of a compound of Formula I-A, and wherein Lg is an appropriate leaving group such as halide (including bromo, iodo, and chloro), tosylate, or other suitable leaving group,
  • The substituents corresponding to M1 are thereafter introduced by reacting aryl or heteroaryl boronic acids with the compound of Formula A-4 to obtain compound A-5.
  • Figure US20160235758A1-20160818-C00047
  • Alternatively, Mitsunobu chemistry can be used to obtain alkylated pyrazolopyrimidine A-4, as shown in Scheme A-1. Iodopyrazolopyrimidine A-3 is reacted with a suitable alcohol, in the presence of triphenylphosphine and diisopropylazodicarboxylate (DIAD) to produce pyrazolopyrimidine A-4.
  • Figure US20160235758A1-20160818-C00048
  • The compounds of the invention may be synthesized via a reaction scheme represented generally in Scheme B. The synthesis proceeds via coupling a compound of Formula A with a compound of Formula B to yield a compound of Formula C. The coupling step is typically catalyzed by using, e.g., a palladium catalyst, including but not limited to palladium tetrakis (triphenylphosphine). The coupling is generally performed in the presence of a suitable base, a nonlimiting example being sodium carbonate. One example of a suitable solvent for the reaction is aqueous dioxane.
  • A compound of Formula A for use in Scheme B has a structure of Formula A, wherein T1 is triflate or halo (including bromo, chloro, and iodo), and wherein R1, X1, X2, X3, R31 and R32 are defined as for a compound of Formula I-A. For boronic acids and acid derivatives as depicted in Formula B, M is either M1 or M2. M1 is defined as for a compound of Formula I-A. For example, M1 can be a 5-benzoxazolyl or a 6-benzoxazolyl moiety, including but not limited to those M1 moieties disclosed herein. M2 is a moiety which is synthetically transformed to form M1, after the M2 moiety has been coupled to the bicyclic core of the compound of Formula A.
  • For a compound of Formula B, G is hydrogen or RG1, wherein RG1 is alkyl, alkenyl, or aryl. Alternatively, B(OG)2 is taken together to form a 5- or 6-membered cyclic moiety. In some embodiments, the compound of Formula B is a compound having a structure of Formula E:
  • Figure US20160235758A1-20160818-C00049
  • wherein G is H or RG1; RG1 is alkyl, alkenyl, or aryl. Alternatively,
  • Figure US20160235758A1-20160818-C00050
  • forms a 5- or 6-membered cyclic moiety; and R2 is a RG2 moiety, wherein the RG2 moiety is H, acyl, or an amino protecting group including but not limited to tert-butyl carbamate (Boc), carbobenzyloxy (Cbz), benzyl (Bz), fluorenylmethyloxycarbonyl (FMOC), p-methoxybenzyl (PMB), and the like.
  • Figure US20160235758A1-20160818-C00051
  • In some embodiments, a compound of Formula B is a compound of Formula B′, wherein G is RG1. or a compound of Formula B″, wherein G is hydrogen. Scheme C depicts an exemplary scheme for synthesizing a compound of Formula B′ or, optionally, Formula B″ for use in Reaction Scheme C. This reaction proceeds via reacting a compound of Formula D with a trialkyl borate or a boronic acid derivative to produce a compound of Formula B′. The reaction is typically run a solvent such as dioxane or tetrahydrofuran. The trialkyl borate includes but is not limited to triisopropyl borate and the boronic acid derivative includes but is not limited to bis(pinacolato)diboron.
  • When the reaction is performed with trialkyl borate, a base such as n-butyllithium is first added to the compound of Formula D to generate an anion, prior to the addition of the borate. When the reaction is performed with a boronic acid derivative such as bis(pinacolato)diboron, a palladium catalyst and a base is used. Typical palladium catalysts include but is not limited to palladium chloride (diphenylphosphino)ferrocene). A suitable base includes but is not limited to potassium acetate.
  • A compound of Formula D for use in Scheme C is a compound wherein T2 is halo or another leaving group, and M is as defined above in Scheme B. The compound of Formula B′ may further be converted to a compound of Formula B″ by treatment with an acid such as hydrochloric acid.
  • In one embodiment of a compound of Formula B, B′, B″, or E, the G groups are hydrogen. In another of a compound of Formula B, B′, B″, or E, the G groups are RG1.
  • In some embodiments, no further synthetic transformation of M1 moiety is performed after the coupling reaction when, e.g. M1 is 2-N-acetyl-benzoxazol-5-yl.
  • Some exemplary compounds of Formula B that can be synthesized via Scheme C include but are not limited to compounds of the following formulae:
  • Figure US20160235758A1-20160818-C00052
    Figure US20160235758A1-20160818-C00053
    Figure US20160235758A1-20160818-C00054
  • In other embodiments of the invention, a compound of Formula E is synthesized from a compound of Formula F, as shown in Scheme C-1:
  • Figure US20160235758A1-20160818-C00055
  • Scheme C-1 depicts an exemplary scheme for synthesizing a compound of Formula E. This reaction proceeds via reacting a compound of Formula F with a trialkyl borate or a boronic acid derivative to produce a compound of Formula E. The conditions of the reaction are as described above in Scheme C.
  • A compound of Formula F for use in Scheme C-1 is a compound wherein T2 is halo (including Br, Cl, and I) or another leaving group (including but not limited to triflate, tosylate, and mesylate), and the Gp moiety is H, acyl, or an amino protecting group including but not limited to tert-butyl carbamate (Boc), carbobenzyloxy (Cbz), benzyl (Bz), fluorenylmethyloxycarbonyl (FMOC), p-methoxybenzyl (PMB), and the like.
  • The compound of Formula E, wherein G is alkyl, may further be converted to a compound of Formula E, wherein G is hydrogen, by treatment with an acid such as hydrochloric acid
  • Where desired, deprotection of a substituent (e.g., removal of Boc protection from an amino substituent) on the benzoxazolyl moiety (i.e. M1 of Formula C) is performed after coupling the compound of Formula B to the compound of Formula A.
  • Some exemplary compounds with such protecting groups, include but are not limited to compounds of the following formulae:
  • Figure US20160235758A1-20160818-C00056
  • An exemplary transformation of M2 to M1 can be carried out via Scheme D as shown below.
  • Figure US20160235758A1-20160818-C00057
  • In Step 1, a compound of Formula 3-1 is reacted with boronic acid 3-2, in the presence of palladium tetrakis (triphenylphosphine) and a suitable base, such as sodium carbonate in an aqueous/organic solvent mixture to produce a compound of Formula 3-3. In Step 2, the compound of Formula 3-3 is reacted with about 2 equivalents of nitric acid in acetic acid as solvent to produce a compound of Formula 3-4. Two alternative transformations may be used to effect the next transformation of Step 3. In the first method, the compound of Formula 3-4 is treated with sodium dithionite and sodium hydroxide in water to produce a compound of Formula 3-5. Alternatively, the compound of Formula 3-4 is reduced using palladium on carbon in a suitable solvent under a hydrogen atmosphere to yield a compound of Formula 3-5.
  • In Step 4, compound 3-5 is reacted with about 1.2 equivalents of cyanogen bromide in a solvent such as methanol/tetrahydrofuran mixture to produce a compound of Formula 3-6. The compound of Formula 3-6 may be further transformed by other substitution or derivatization.
  • A compound of Formula 3-1 useful in the method of Scheme D is a compound having a structure of Formula 3-1, wherein T1 is triflate or halo (including bromo, chloro, and iodo), and wherein R1, X1, X2, X3, R31 and R32 are defined as for a compound of Formula I-A.
  • Exemplary compounds having a pyrazolopyrimidine core can be synthesized via Scheme E.
  • Figure US20160235758A1-20160818-C00058
  • In Step 1 of Scheme E, compound A-2 in dimethylformamide (DMF), is reacted with an N-halosuccinimide (NT1S) at about 80° C., to provide compound 4-1, where T1 is iodo or bromo. In Step 2, compound 4-1 in DMF is reacted with a compound R1Tx, in the presence of potassium carbonate, to provide compound 4-2. In Step 4, compound 4-2 is coupled with a compound of Formula B using palladium catalysis such as palladium tetrakis (triphenylphosphine), and in the presence of sodium carbonate, to yield a pyrazolopyrimidine compound as shown.
  • A compound of Formula R1Tx suitable for use in Reaction Scheme E is the compound wherein R1 is cycloalkyl or alkyl and Tx is halo (including bromo, iodo, or chloro) or a leaving group, including but not limited to mesylate or tosylate.
  • Reaction Schemes F-M illustrate methods of synthesis of borane reagents useful in preparing intermediates of use in synthesis of the compounds of the invention as described in Reaction Schemes A, B, and E above, to introduce M1 substituents.
  • Figure US20160235758A1-20160818-C00059
  • Figure US20160235758A1-20160818-C00060
  • Figure US20160235758A1-20160818-C00061
  • Figure US20160235758A1-20160818-C00062
  • Figure US20160235758A1-20160818-C00063
  • Figure US20160235758A1-20160818-C00064
  • Figure US20160235758A1-20160818-C00065
  • Figure US20160235758A1-20160818-C00066
  • Figure US20160235758A1-20160818-C00067
  • In an alternative method of synthesis, a compound of Formula N-1 and a compound of N-2 are coupled to produce a compound of Formula C. The coupling step is typically catalyzed by using, e.g., a palladium catalyst, including but not limited to palladium tetrakis (triphenylphosphine). The coupling is generally performed in the presence of a suitable base, a nonlimiting example being sodium carbonate. One example of a suitable solvent for the reaction is aqueous dioxane.
  • A compound of Formula N-1 for use in Scheme N has a structure of Formula N-1, wherein G is hydrogen or RG1, wherein RG1 is alkyl, alkenyl, or aryl. Alternatively, B(OG)2 of the compound of Formula N-1 is taken together to form a 5- or 6-membered cyclic moiety. R1, X1, X2, X3, R31 and R32 of the compound of Formula N-1 are defined as for a compound of Formula I-A.
  • A compound of Formula N-2 for use in Scheme N has a structure of Formula N-2 wherein T1 is triflate or halo (including bromo, chloro, and iodo). M of the compound of Formula N-2 is either M1 or M2. M1 is defined as for a compound of Formula I For example, M1 can be a 5-benzoxazolyl or a 6-benzoxazolyl moiety, including but not limited to those M1 moieties disclosed herein. M2 is a moiety which is synthetically transformed to form M1, after the M2 moiety has been coupled to the bicyclic core of the compound of Formula N-1.
  • Figure US20160235758A1-20160818-C00068
  • A compound of Formula N-1 may be synthesized as shown in Scheme N-1. A compound of Formula N-1 is reacted with a trialkyl borate or a boronic acid derivative to produce a compound of Formula N-1. The reaction is typically run a solvent such as dioxane or tetrahydrofuran. The trialkyl borate includes but is not limited to triisopropyl borate and the boronic acid derivative includes but is not limited to bis(pinacolato)diboron.
  • When the reaction is performed with trialkyl borate, a base such as n-butyllithium is first added to the compound of Formula N-3 to generate an anion, prior to the addition of the borate. When the reaction is performed with a boronic acid derivative such as bis(pinacolato)diboron, a palladium catalyst and a base is used. Typical palladium catalysts include but is not limited to palladium chloride (diphenylphosphino)ferrocene). A suitable base includes but is not limited to potassium acetate.
  • A compound of Formula N-3 suitable for use in Scheme N-1 is a compound wherein T2 is halo or another leaving group such as mesylate, tosylate, or triflate. X1, X2, X3, R1, R31, and R32 of the compound of Formula N-3 is as defined for a compound of Formula I-A.
  • In some embodiments of the invention, a compound of Formula A, B, B′, B″, C, C″, D, E, E″, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1″, N-3″, 3-1″, 3-3″, 3-4″, 3-5″, 3-6″, N-1″, or N-3″ is provided as its salt, including but not limited to hydrochloride, acetate, formate, nitrate, sulfate, and boronate.
  • In some embodiments of the invention, a palladium compound, including but not limited to palladium chloride (diphenylphosphino)ferrocene) and palladium tetrakis (triphenylphosphine), is used in the synthesis of a compound of Formula A, B, B′, B″, C, C″, D, E, E″, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1″, N-3″, 3-1″, 3-3″, 3-4″, 3-5″, 3-6″, N-1″, or N-3″. When a palladium compound is present in the synthesis of a compound of Formula A, B, B′, B″, C, C″, D, E, E″, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1″, N-3″, 3-1″, 3-3″, 3-4″, 3-5″, 3-6″, N-1″, or N-3″, it is present in an amount ranging from about 0.005 molar equivalents to about 0.5 molar equivalents, from about 0.05 molar equivalents to about 0.20 molar equivalents, from about 0.05 molar equivalents to about 0.25 molar equivalents, from about 0.07 molar equivalents to about 0.15 molar equivalents, or about 0.8 molar equivalents to about 0.1 molar equivalents of the compound of Formula A, B, B′, B″, C, D, E, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1, or N-3. In some embodiments, a a palladium compound, including but not limited to palladium chloride (diphenylphosphino)ferrocene) and palladium tetrakis (triphenylphosphine) is present in the synthesis of a compound of Formula A, B, B′, B″, C, C″, D, E, E″, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1″, N-3″, 3-1″, 3-3″, 3-4″, 3-5″, 3-6″, N-1″, or N-3″ in about 0.07, about 0.08, about 0.09, about 0.10, about 0.11, about 0.12, about 0.13, about 0.14, or about 0.15 molar equivalents of a starting material of Formula A, B, B′, B″, C, C″, D, E, E″, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1″, N-3″, 3-1″, 3-3″, 3-4″, 3-5″, 3-6″, N-1″, or N-3″ that is used to synthesize a compound of Formula A, B, B′, B″, C, C″, D, E, E″, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1″, N-3″, 3-1″, 3-3″, 3-4″, 3-5″, 3-6″, N-1″, or N-3″.
  • In some embodiments of the above reaction schemes B, D, E, N or N-1, another embodiment of the compounds of Formula A, C, 3-1, 3-3, 3-4, 3-5, 3-6, A-2, 4-1, 4-2, N-1 and N-3 is as shown in Schemes B′. D′. E′, N′ or N-1′ below. In these alternative syntheses, producing a compound of Formula C, 3-1, 3-3, 3-4, 3-5, 3-6, A-2, 4-1, 4-2, N-1 or N-3, use compounds that comprise an amino moiety having a RG2 moiety present during one or more of the synthetic steps, wherein RG2 is an amino protecting group including but not limited to tert-butyl carbamate (Boc), carbobenzyloxy (Cbz), benzyl (Bz), fluorenylmethyloxycarbonyl (FMOC), p-methoxybenzyl (PMB), and the like. These compounds include a compound of Formula A″, C″, 3-1″, 3-3″, 3-4″, 3-5″, 3-6″, A-2″, 4-1″, 4-2″, N-1″ or N-3″.
  • The RG2 moiety is removed, using suitable methods, at any point desired, whereupon the compound of Formula C, 3-1, 3-3, 3-4, 3-5, 3-6, A-2, 4-1, 4-2, N-1 or N-3 has a R31 hydrogen replacing the RG2 moiety on the amino moiety. This transformation is specifically illustrated for the conversion of a compound of Formula C″ to a compound of C (i.e., as in Step 4 of Scheme E′) and for the conversion of a compound of Formula 3-6″ to a compound of Formula 3-6 (ie., as in Step 5 of Scheme D′). This illustration is in no way limiting as to the choice of steps wherein a compound comprising a NR31RG2 moiety may be converted to a compound comprising a NR31R32 moiety wherein the R32 moiety is hydrogen.
  • Figure US20160235758A1-20160818-C00069
  • Figure US20160235758A1-20160818-C00070
    Figure US20160235758A1-20160818-C00071
  • Figure US20160235758A1-20160818-C00072
  • Figure US20160235758A1-20160818-C00073
  • Additionally, the invention encompasses methods of synthesis of the compounds of A, B, B′, B″, C, E, 3-1, 3-2, 3-3, 3-4, 3-5, 3-6, N-1 or N-3, wherein one or more of M, M1, or R1 has a protecting group present during one or more steps of the synthesis. Protecting groups suitable for use for a M, M1, or R1 moiety are well known in the art, as well as the methods of incorporation and removal, and the reagents suitable for such transformations.
  • Compounds of the invention where X4 is C—R9 may be prepared by methods analogous to the ones described in the Schemes illustrated above.
  • Reaction Schemes 0, P and Q illustrate methods of synthesis of borane reagents useful in preparing intermediates of use in synthesis of the compounds of the invention as described in Reaction Schemes 1 and 2 above, to introduce benzothiazolyl substituents.
  • Figure US20160235758A1-20160818-C00074
  • A compound of Formula O-1 is treated with, for example, nitric acid to produce a compound of Formula O-2. The compound of Formula O-2 is treated with a reducing agent such as stannous chloride to produce a compound of Formula O-3. The compound of 0-3 is treated with sodium nitrate in acide and cupric bromide to produce a compound of Formula O-4. The compound of O-4 is treated a base such as butyl lithium and boron tris-isopropoxide to produce a compound of Formula O-5.
  • Figure US20160235758A1-20160818-C00075
  • A compound of Formula P-1 is treated with, for example, potassium thiocyanate and bromine in acetic acid to produce a compound of Formula P-2. The compound of Formula P-2 is treated with an acteylating reagent such as acetyl chloride to produce a compound of Formula P-3. The compound of P-3 is reacted with, for example, bis(pinacolato)diboron (compound P-4) in the presence of a catalyst such as palladium chloride to produce a compound of Formula P-5.
  • Figure US20160235758A1-20160818-C00076
  • The compound of Formula P-2 is reacted with, for example, methyl carbamic acid chloride to produce a compound of Formula Q-1. The compound of Formula Q-1 is reacted with bis(pinacolato)diboron (compound P-4) in the presence of a catalyst such as Pd2(dba)3, 2-chlorohexylphosphino-2, 4, 6-triisopropylbiphenyl, a base such as potassium acetate, to produce the compound of Formula Q-2.
  • Some illustrative compounds of the invention which are mTor inhibitors are described below. The compounds of the invention are not limited in any way to the compounds illustrated herein.
  • Figure US20160235758A1-20160818-C00077
    Figure US20160235758A1-20160818-C00078
    Figure US20160235758A1-20160818-C00079
    Figure US20160235758A1-20160818-C00080
    Figure US20160235758A1-20160818-C00081
    Figure US20160235758A1-20160818-C00082
    Figure US20160235758A1-20160818-C00083
    Figure US20160235758A1-20160818-C00084
  • Illustrative compounds of the invention include those of subclass 1a, 1b, 2a, 2b, 3a, 3b, 4a, 4b, 5a, 5b, 6a, 6b, 7a, 7b, 8a, 8b, 9a, 9b, 10a, 10b, 11a, 11b, 12a, 12b, 13a, 13b, 14a, 14b, 15a, 15b, 16a, or 16b, where the substituents R1, X1, and V are as described below.
  • In some embodiments, when R1 is H and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is H and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is CH3 and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is CH3 and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is Et and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is Et and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is iPr and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is iPr and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In one embodiment, R1 is iPr, X1 is N, and V is NH2. In another embodiment, R1 is iPr, X1 is N, and V is NHCOMe. In other embodiments, when R1 is cyclobutyl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is cyclobutyl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is cyclopentyl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is cyclopentyl and X1 is N V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is phenyl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is phenyl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is pyridin-2-yl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is pyridin-2-yl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is N-methylaminocyclohex-4-yl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is N-methylaminocyclohex-4-yl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is N-methylpiperidin-4-yl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is N-methylpiperidin-4-yl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is N-methylaminocyclobut-3-yl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is N-methylaminocyclobut-3-yl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is tert-butyl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is tert-butyl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is 1-cyano-but-4-yl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is 1-cyano-but-4-yl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is 1-cyano-prop-3-yl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is 1-cyano-prop-3-yl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is 3-azetidinyl and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is 3-azetidinyl and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me.
  • In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00085
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00086
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00087
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00088
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00089
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00090
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00091
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00092
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00093
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00094
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00095
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00096
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00097
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00098
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00099
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00100
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00101
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00102
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00103
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00104
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00105
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00106
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00107
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00108
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00109
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00110
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00111
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00112
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00113
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00114
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00115
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00116
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00117
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00118
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00119
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00120
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00121
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00122
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00123
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe. NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00124
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00125
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00126
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00127
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00128
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00129
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00130
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00131
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00132
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00133
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00134
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00135
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00136
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00137
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00138
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00139
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00140
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00141
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00142
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00143
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00144
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00145
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00146
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00147
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00148
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00149
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00150
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00151
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00152
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00153
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00154
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00155
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00156
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00157
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00158
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00159
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00160
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00161
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00162
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00163
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00164
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00165
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00166
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me.
  • In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00167
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00168
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00169
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00170
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00171
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00172
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00173
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00174
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00175
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00176
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00177
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00178
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00179
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00180
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me.
  • In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00181
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00182
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00183
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00184
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00185
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00186
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00187
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00188
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00189
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00190
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00191
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00192
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00193
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00194
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00195
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00196
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00197
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00198
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00199
  • and X1 is CH, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00200
  • and X1 is N, V is phenylamino, benzyl, phenyl, NHMe, NH2, NHEt, NHCOH, NHCOMe, NHCOEt, NHCOiPr, NHCOOMe, CONHMe, or NHSO2Me.
  • In the noted embodiments, pyridin-2-yl
  • Figure US20160235758A1-20160818-C00201
  • is N-methylaminocyclohex-4-yl is
  • Figure US20160235758A1-20160818-C00202
  • N-methylpiperidin-4-yl is
  • Figure US20160235758A1-20160818-C00203
  • and N-methylaminocyclobut-3-yl is
  • Figure US20160235758A1-20160818-C00204
  • Illustrative compounds of the invention include those of subclass 1a, 1b, 2a, 2b, 3a, 3b, 4a, 4b, 5a, 5b, 6a, 6b, 7a, 7b, 8a, 8b, 9a, 9b, 10a, 10b, 11a, 11b, 12a, 12b, 13a, 13b, 14a, 14b, 15a, 15b, 16a, or 16b, where the substituents R1, X1, and V are as described below. In some embodiments, when R1 is H and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is H and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is CH3 and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is CH3 and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is Et and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is Et and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is iPr and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is iPr and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is cyclobutyl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is cyclobutyl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is cyclopentyl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is cyclopentyl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is phenyl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is phenyl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is pyridin-2-yl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is pyridin-2-yl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is N-methylaminocyclohex-4-yl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is N-methylaminocyclohex-4-yl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is N-methylpiperidin-4-yl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is N-methylpiperidin-4-yl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In some embodiments, when R1 is N-methylaminocyclobut-3-yl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is N-methylaminocyclobut-3-yl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is tert-butyl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is tert-butyl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is 1-cyano-but-4-yl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is 1-cyano-but-4-yl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is 1-cyano-prop-3-yl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is 1-cyano-prop-3-yl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is 3-azetidinyl and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is 3-azetidinyl and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00205
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00206
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00207
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00208
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00209
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00210
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00211
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00212
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00213
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00214
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00215
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00216
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00217
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00218
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00219
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00220
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00221
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00222
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00223
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00224
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00225
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00226
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00227
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00228
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00229
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00230
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00231
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00232
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00233
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00234
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00235
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00236
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00237
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00238
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00239
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00240
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00241
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00242
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00243
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00244
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00245
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00246
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00247
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00248
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00249
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00250
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00251
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00252
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00253
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00254
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00255
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00256
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00257
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00258
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00259
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00260
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00261
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00262
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00263
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00264
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00265
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00266
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00267
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00268
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00269
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00270
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00271
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00272
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00273
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00274
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00275
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00276
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00277
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00278
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00279
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00280
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00281
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00282
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00283
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00284
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00285
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00286
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino.
  • In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00287
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00288
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00289
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00290
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00291
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00292
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00293
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00294
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00295
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00296
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00297
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00298
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00299
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00300
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino.
  • In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00301
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00302
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00303
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00304
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00305
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00306
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00307
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00308
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00309
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00310
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00311
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00312
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00313
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00314
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00315
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00316
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00317
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00318
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00319
  • and X1 is CH, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino. In other embodiments, when R1 is
  • Figure US20160235758A1-20160818-C00320
  • and X1 is N, V is cyclopropanecarboxamido, cyclopropylamino, morpholinoethylamino, hydroxyethylamino, or N-morpholino.
  • In the noted embodiments, cyclopropanecarboxamido is
  • Figure US20160235758A1-20160818-C00321
  • cyclopropylamino is
  • Figure US20160235758A1-20160818-C00322
  • 2-morpholinoethylamino is
  • Figure US20160235758A1-20160818-C00323
  • hydroxyethylamino is
  • Figure US20160235758A1-20160818-C00324
  • and N-morpholino is
  • Figure US20160235758A1-20160818-C00325
  • TABLE 1
    Biological activity of several illustrative mTor inhibitor compounds of the invention.
    mTOR PI3K α PI3K β PI3K γ PI3K δ PC3
    Structure IC50 (nM) IC50 (nM) IC50 (nM) IC50 (nM) IC50 (nM) EC50 (nM)
     1
    Figure US20160235758A1-20160818-C00326
    ++++ +++ ++ ++++ +++ ++++
     2
    Figure US20160235758A1-20160818-C00327
    ++++ ++ + +++ +++ +++
     3
    Figure US20160235758A1-20160818-C00328
    ++ + ++ ++ ++
     4
    Figure US20160235758A1-20160818-C00329
    +++ ++ ++ +++ +++ ++
     5
    Figure US20160235758A1-20160818-C00330
    ++++ +++ ++ ++++ +++ ++++
     6
    Figure US20160235758A1-20160818-C00331
    ++++ ++ + ++ +++ +++
     7
    Figure US20160235758A1-20160818-C00332
    ++++ +++ ++ ++ +++ ++
     8
    Figure US20160235758A1-20160818-C00333
    ++++ +++ + +++ +++ ++++
     9
    Figure US20160235758A1-20160818-C00334
    ++++ ++ + +++ +++ ++++
    10
    Figure US20160235758A1-20160818-C00335
    ++ +
    11
    Figure US20160235758A1-20160818-C00336
    +++ +
    12
    Figure US20160235758A1-20160818-C00337
    +++ +
    13
    Figure US20160235758A1-20160818-C00338
    ++ ++ +++ +++
    14
    Figure US20160235758A1-20160818-C00339
    ++ ++ +++ ++
    15
    Figure US20160235758A1-20160818-C00340
    + + + +
    16
    Figure US20160235758A1-20160818-C00341
    + + ++ +
    17
    Figure US20160235758A1-20160818-C00342
    + + + +
    18
    Figure US20160235758A1-20160818-C00343
    + + + +
    19
    Figure US20160235758A1-20160818-C00344
    ++ + + +
    20
    Figure US20160235758A1-20160818-C00345
    ++ ++ + ++
    21
    Figure US20160235758A1-20160818-C00346
    +++ + + + +
    22
    Figure US20160235758A1-20160818-C00347
    ++++ ++++ ++ +++ +++ ++
    23
    Figure US20160235758A1-20160818-C00348
    ++++ ++ + ++ ++
    24
    Figure US20160235758A1-20160818-C00349
    + + + +
    25
    Figure US20160235758A1-20160818-C00350
    +++ ++ ++++ +++
    26
    Figure US20160235758A1-20160818-C00351
    ++++ +++ ++++ +++
    27
    Figure US20160235758A1-20160818-C00352
    ++ + + +++
  • Table 1 shows the biological activity in mTOR and PI3K kinase assays of several compounds of the invention. The scale utilized in Table 1 is as follows: ++++ less than 100 nM; +++ less than 1.0 μM; ++ less than 10 μM; and + greater than 10 μM.
  • In other embodiments, the present invention provides the following compounds:
  • Figure US20160235758A1-20160818-C00353
    Figure US20160235758A1-20160818-C00354
    Figure US20160235758A1-20160818-C00355
  • Any of the compounds shown above may show a biological activity in an mTOR or PI3K inhibition assay of between about 0.5 nM and 25 μM (IC50).
  • Additional compounds which are mTor inhibitors of the invention are shown in Table 2.
  • TABLE 2
    In vitro IC50 values for Illustrative mTor Inhibitor Compounds of the Invention.
    PC3
    mTORC PI3K α PI3K β PI3K γ PI3K δ prolifera-
    # Structure IC50 (nM) IC50 (nM) IC50 (nM) IC50 (nM) IC50 (nM) tion (nM)
     1
    Figure US20160235758A1-20160818-C00356
    ++++ + + ++ ++ +++
     2
    Figure US20160235758A1-20160818-C00357
    +
     3
    Figure US20160235758A1-20160818-C00358
    ++ +
     4
    Figure US20160235758A1-20160818-C00359
    + +
     5
    Figure US20160235758A1-20160818-C00360
    + + +
     6
    Figure US20160235758A1-20160818-C00361
    + + +
     7
    Figure US20160235758A1-20160818-C00362
    +++ + +
     8
    Figure US20160235758A1-20160818-C00363
    + + +
     9
    Figure US20160235758A1-20160818-C00364
    ++++ + +
    10
    Figure US20160235758A1-20160818-C00365
    +++++ + + + + +
    11
    Figure US20160235758A1-20160818-C00366
    +++++++ + + ++ ++ ++++
    12
    Figure US20160235758A1-20160818-C00367
    ++++++ + + ++ + ++++
    13
    Figure US20160235758A1-20160818-C00368
    + + +
    14
    Figure US20160235758A1-20160818-C00369
    + +
    15
    Figure US20160235758A1-20160818-C00370
    +++++++ + + ++++ ++++ ++++
    16
    Figure US20160235758A1-20160818-C00371
    +++++++ + + ++ +++ ++
    17
    Figure US20160235758A1-20160818-C00372
    + + +
    18
    Figure US20160235758A1-20160818-C00373
    + * *
    19
    Figure US20160235758A1-20160818-C00374
    + +
    20
    Figure US20160235758A1-20160818-C00375
    + +
    21
    Figure US20160235758A1-20160818-C00376
    ++++ ++ + ++ ++ +
    22
    Figure US20160235758A1-20160818-C00377
    +++++++ + + + ++
    23
    Figure US20160235758A1-20160818-C00378
    + +
    24
    Figure US20160235758A1-20160818-C00379
    + + +
    25
    Figure US20160235758A1-20160818-C00380
    ++ + +
    26
    Figure US20160235758A1-20160818-C00381
    ++++++ + + ++ +++ ++
    27
    Figure US20160235758A1-20160818-C00382
    +++++ ++
    28
    Figure US20160235758A1-20160818-C00383
    ++ + + + +
    29
    Figure US20160235758A1-20160818-C00384
    +
    30
    Figure US20160235758A1-20160818-C00385
    +++++ + + + +
    31
    Figure US20160235758A1-20160818-C00386
    +++++ + + ++ +
    32
    Figure US20160235758A1-20160818-C00387
    ++ + + + +
    33
    Figure US20160235758A1-20160818-C00388
    ++ + + + +
    34
    Figure US20160235758A1-20160818-C00389
    + + + +
    35
    Figure US20160235758A1-20160818-C00390
    + + + +
    36
    Figure US20160235758A1-20160818-C00391
    ++++++ + +++ ++ +++
    37
    Figure US20160235758A1-20160818-C00392
    + ++ ++ ++
    38
    Figure US20160235758A1-20160818-C00393
    ++ + + + +
    39
    Figure US20160235758A1-20160818-C00394
    ++++++ + + + +
    40
    Figure US20160235758A1-20160818-C00395
    +++ + + + +
    41
    Figure US20160235758A1-20160818-C00396
    ++++++ + + ++++ + +
    42
    Figure US20160235758A1-20160818-C00397
    +++++++ + + +++ +
    43
    Figure US20160235758A1-20160818-C00398
    + + + +
    44
    Figure US20160235758A1-20160818-C00399
    +++ + + +
    45
    Figure US20160235758A1-20160818-C00400
    +
    46
    Figure US20160235758A1-20160818-C00401
    47
    Figure US20160235758A1-20160818-C00402
    48
    Figure US20160235758A1-20160818-C00403
    ++++ + + + +
    49
    Figure US20160235758A1-20160818-C00404
    ++++++ + + ++ ++
    50
    Figure US20160235758A1-20160818-C00405
    ++++ + + ++ ++
    51
    Figure US20160235758A1-20160818-C00406
    ++++ + + ++ ++
    52
    Figure US20160235758A1-20160818-C00407
    ++ + + + ++
    53
    Figure US20160235758A1-20160818-C00408
    +++ + + +
    54
    Figure US20160235758A1-20160818-C00409
    +++++ + + +
    55
    Figure US20160235758A1-20160818-C00410
    ++ + + +
    56
    Figure US20160235758A1-20160818-C00411
    + + + +
    57
    Figure US20160235758A1-20160818-C00412
    +++++ + + +
    58
    Figure US20160235758A1-20160818-C00413
    + + + +
    59
    Figure US20160235758A1-20160818-C00414
    + + + +
    60
    Figure US20160235758A1-20160818-C00415
    +++ + + +++
    61
    Figure US20160235758A1-20160818-C00416
    +++++ + + + +
    62
    Figure US20160235758A1-20160818-C00417
    +++++++ + + + +++
    63
    Figure US20160235758A1-20160818-C00418
    +++++++ ++ + +++++ +++++
    64
    Figure US20160235758A1-20160818-C00419
    +++++ + + ++ ++
    65
    Figure US20160235758A1-20160818-C00420
    ++++++ ++++ + +++++ +++++
    66
    Figure US20160235758A1-20160818-C00421
    + + + + +
    67
    Figure US20160235758A1-20160818-C00422
    + + + + +
    68
    Figure US20160235758A1-20160818-C00423
    +++++++ ++ + ++++ +++++
    69
    Figure US20160235758A1-20160818-C00424
    +++++++ + + + ++
    70
    Figure US20160235758A1-20160818-C00425
    +++++++ ++ + +++ +++++
    71
    Figure US20160235758A1-20160818-C00426
    +++ + + + +
  • In Table 2 above, a +++++++ indicates an IC50 of 5 nM or less; a ++++++ indicates an IC50 of 10 nM or less; a +++++ indicates an IC50 of 25 nM or less; an ++++ indicates an IC50 of 50 nm or less, a +++ indicates an IC50 of 100 nM or less, a ++ indicates an IC50 of 500 nM or less, and a + indicates an IC50 of more than 500 nM.
  • Exemplary PI3Kα Inhibitor Compounds
  • In one aspect, the present invention provides a PI3Kα inhibitor which is a compound of Formula I:
  • Figure US20160235758A1-20160818-C00427
  • or its pharmaceutically acceptable salts thereof, wherein:
  • W1′ is N, NR3′, or CR3′; W2′ is N, NR4′, CR4′, or C═O; W3′ is N, NR5 or CR5; W4′ is N, wherein no more than two N atoms and no more than two C═O groups are adjacent;
  • W5′ is N;
  • W6′ is N or CR8′;
  • Wa′ and Wb′ are independently N or CR9′;
  • one of Wc′ and Wd′ is N, and the other is O, NR10′, or S;
  • R1′ and R2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety;
  • R5′, R6′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R9′ is alkyl or halo; and
  • R10′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • For example, the present invention provides a PI3Kα inhibitor which is a compound of Formula I:
  • Figure US20160235758A1-20160818-C00428
  • or its pharmaceutically acceptable salts thereof, wherein:
  • X is O or S or N;
  • W1′ is N, NR3′, CR3′, or C═O, W2′, is N, NR4′, or CR4′, or C═O, W3′ is N, NR5′, or CR5′, W4′ is N, C═O or CR6′, wherein no more than two N atoms and no more than two C═O groups are adjacent;
  • W5′ is N or CR7′;
  • W6′ is N or CR8′;
  • R1′ and R2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety; and
  • R5′, R6′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments, the compound of Formula II exists as a tautomer, and such tautomers are contemplated by the present invention.
  • In some embodiments, the compound of Formula II has the formula:
  • Figure US20160235758A1-20160818-C00429
  • For example, a compound of Formula II is:
  • Figure US20160235758A1-20160818-C00430
  • In some embodiments of the compound of Formula II, W1′ is CR3′, W2′ is CR4′, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′; W1 is N, W2 is CR4′, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′; or W1 is CR3′, W2′ is N, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′. Formulas for such embodiments are shown below:
  • Figure US20160235758A1-20160818-C00431
  • In some embodiments, X is O. In other embodiments, X is S.
  • In some embodiments, R1′ is hydrogen. In other embodiments, R1′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″, wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments, R2′ is hydrogen. In other embodiments, R2′ is, for example, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R2′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R2′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R2′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R2′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R2′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo[1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds wherein R2′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R2′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R2′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula II, R2′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R2′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R2′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R2′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R2′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R2′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R2′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3.
  • In some embodiments of the compound of Formula II, W1′ is CR3. R3′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R3′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R3′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R3′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R3′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R3′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo[1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds of Formula II wherein R3′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R3′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R3′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula II, R3′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R3′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R3′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R3′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R3′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R3′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R3′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3.
  • R3′ of the compounds of Formula II can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00432
  • The invention also provides compounds of Formula II, wherein when R3′ is a member of the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, and NR′R″ (wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety), then R3′ is optionally substituted with one or more of the following substituents: alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, heterocycloalkyloxy, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. Each of the above substituents may be further substituted with one or more substituents chosen from the group consisting of alkyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, oxo, phosphate, urea, and carbonate.
  • For example, the invention provides compounds wherein when R3′ is alkyl, the alkyl is substituted with NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety. The cyclic moiety so formed can be unsubstituted or substituted. Non-limiting exemplary cyclic moieties includes but are not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, and thiomorpholinyl. In other examples of the compounds of Formula II, when R3′ is alkyl, the alkyl is substituted with heterocycloalkyl, which includes oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolyl, tetrahydropyranyl, piperidinyl, morpholinyl, and piperazinyl. All of the above listed heterocycloaklyl substituents can be unsubstituted or substituted.
  • In yet other examples of the compounds of Formula II, when R3′ is alkyl, the alkyl is substituted with a 5, 6, 7, 8, 9, or 10 membered monocyclic or bicyclic heteroaryl, which is unsubstituted or substituted. The monocyclic heteroaryl includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. The bicyclic heteroaryl includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo[1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In other embodiments of the compound of Formula II, R3′ is —NHR3′, —N(CH3)R3′, —N(CH2CH3)R3′, —N(CH(CH3)2)R3′, or OR3′, wherein R3′ is unsubstituted or substituted heterocycloalkyl (nonlimiting examples thereof include 4-NH piperidin-1-yl, 4-methyl piperidin-1-yl, 4-ethyl piperidin-1-yl, 4-isopropyl-piperidin-1-yl, and pyrrolidin-3-yl), unsubstituted or substituted monocyclic aryl, or unsubstituted or substituted monocyclic heteroaryl (including but not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl). In one example, R3′ is O-aryl, i.e. phenoxy. In another example, R3′ is —O-(4-methyl)piperidin-1-yl or —O-(4-isopropyl)piperidin-1-yl.
  • In some embodiments of the compound of Formula II, R3′ is one of the following moieties:
  • Figure US20160235758A1-20160818-C00433
    Figure US20160235758A1-20160818-C00434
    Figure US20160235758A1-20160818-C00435
    Figure US20160235758A1-20160818-C00436
    Figure US20160235758A1-20160818-C00437
    Figure US20160235758A1-20160818-C00438
  • In some embodiments of the compound of Formula II, W1′ is NR3′, wherein R3′ is hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or unsubstituted or substituted C3-C7cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl). In other embodiments of the compound of Formula II, R3′ is unsubstituted or substituted heterocycloalkyl (which includes but is not limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, and piperazinyl), or unsubstituted or substituted C2-C10heteroalkyl (which includes but is not limited to methoxyethoxy, methoxymethyl, and diethylaminoethyl). Alternatively, R3′ is unsubstituted or substituted monocyclic heteroaryl (which includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl) or unsubstituted or substituted monocyclic aryl.
  • In still other embodiments, W1 is C═O.
  • In some embodiments of the compound of Formula II, W2′ is CR4′. R4′ can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R4′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R4′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R4′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R4′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R4′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • The present invention also provides compounds of Formula II wherein R4′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R4′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R4′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula II, R4′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R4′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R4′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R4′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In some embodiments, R4′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R4′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, or carbonate. Also contemplated are R4′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3.
  • R4′ of the compounds of Formula II, can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00439
  • The invention also provides compounds of Formula II, wherein when R4′ is a member of the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, and NR′R″ (wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety), then R4′ is optionally substituted with one or more of the following substituents: alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. Each of the above substituents may be further substituted with one or more substituents chosen from the group consisting of alkyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, oxo, phosphate, urea, and carbonate.
  • For example, the invention provides compounds wherein when R4′ is alkyl, the alkyl is substituted with NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety. The cyclic moiety so formed can be unsubstituted or substituted. Non-limiting exemplary cyclic moieties includes but are not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. In other examples of the compounds of Formula II, when R4′ is alkyl, the alkyl is substituted with heterocycloalkyl, which includes oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolyl, tetrahydropyranyl, piperidinyl, morpholinyl, and piperazinyl. All of the above listed heterocycloaklyl substituents can be unsubstituted or substituted.
  • In yet other examples of the compounds of Formula II, when R4′ is alkyl, the alkyl is substituted with a 5, 6, 7, 8, 9, or 10 membered monocyclic or bicyclic heteroaryl, which is unsubstituted or substituted. The monocyclic heteroaryl includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. The bicyclic heteroaryl includes but is not limited benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In some embodiments of the compound of Formula II, W2 is NR4′, wherein R4′ is hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or unsubstituted or substituted C3-C7cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl). In other embodiments of the compound of Formula II, R4′ is unsubstituted or substituted heterocycloalkyl (which includes but is not limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, and piperazinyl), or unsubstituted or substituted C2-C10heteroalkyl (which includes but is not limited to methoxyethoxy, methoxymethyl, and diethylaminoethyl). Alternatively, R4′ is unsubstituted or substituted monocyclic heteroaryl (which includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl) or unsubstituted or substituted monocyclic aryl.
  • In some embodiments R3′ and R4′ taken together form a cyclic moiety. Such a moiety may have, for example, from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted. In some embodiments, the substituent is C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or C3-C7cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl); heterocycloalkyl (which includes but is not limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, and piperazinyl), C2-C10heteroalkyl (which includes but is not limited to methoxyethoxy, methoxymethyl, and diethylaminoethyl); monocyclic heteroaryl (which includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl) or unsubstituted or substituted monocyclic aryl. The cyclic moiety may have one or more substituents, which may be the same or different.
  • In some embodiments, the cyclic moiety formed by R3′ and R4′ is substituted with at least one of the following substituents:
  • Figure US20160235758A1-20160818-C00440
    Figure US20160235758A1-20160818-C00441
    Figure US20160235758A1-20160818-C00442
    Figure US20160235758A1-20160818-C00443
    Figure US20160235758A1-20160818-C00444
    Figure US20160235758A1-20160818-C00445
  • In some embodiments of the compound of Formula II, W3′ is CR5′. R5′ can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In one embodiment, R5′ is H. In other embodiments, R5′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R5′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R5′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R5′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R5′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In some embodiments of the compound of Formula II, W3′ is N or NR5′, wherein R5′ is hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or unsubstituted or substituted C3-C7cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl). In other embodiments of the compound of Formula II, R5′ is unsubstituted or substituted heterocycloalkyl (which includes but is not limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, and piperazinyl), or unsubstituted or substituted C2-C10heteroalkyl (which includes but is not limited to methoxyethoxy, methoxymethyl, and diethylaminoethyl). Alternatively, R5′ is unsubstituted or substituted monocyclic heteroaryl (which includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl) or unsubstituted or substituted monocyclic aryl.
  • In some embodiments of the compound of Formula II, W4′ is CR6. R6 can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In one embodiment, R6′ is H. In other embodiments, R6′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R6 is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R6′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R6′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R6′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In some embodiments of the compound of Formula II, W4′ is N or NR6′, wherein R6′ is hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or unsubstituted or substituted C3-C7cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl). In other embodiments of the compound of Formula II, R6 is unsubstituted or substituted heterocycloalkyl (which includes but is not limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, and piperazinyl), or unsubstituted or substituted C2-C10heteroalkyl (which includes but is not limited to methoxyethoxy, methoxymethyl, and diethylaminoethyl). Alternatively, R6′ is unsubstituted or substituted monocyclic heteroaryl (which includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl) or unsubstituted or substituted monocyclic aryl.
  • In other embodiments, W4′ is C═O.
  • In some embodiments of the compound of Formula II, W5′ is N. In other embodiments of the compound of Formula II, W5′ is CR7′. R7′ can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In one embodiment, R7′ is H. In other embodiments, R7′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R7′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R7′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R7′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R7′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In some embodiments of the compound of Formula II, W6′ is N. In other embodiments of the compound of Formula II, W6′ is CR8′. R8 can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In one embodiment, R8 is H. In other embodiments, R8 is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R8 is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R8 is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R8 includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R8 includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In some embodiments, the compound of Formula II has the formula:
  • Figure US20160235758A1-20160818-C00446
  • In other embodiments, the compound of Formula II is:
  • Figure US20160235758A1-20160818-C00447
  • For example, the compound of Formula II is:
  • Figure US20160235758A1-20160818-C00448
  • In some embodiments, the compound of Formula II is:
  • Figure US20160235758A1-20160818-C00449
  • In another aspect, the invention provides compounds of Subformula IIa.
  • Figure US20160235758A1-20160818-C00450
  • In one embodiment, R1′, R3′, R4′, R5′, and R8′ are hydrogen. In another embodiment, R1′, R3′, R5′, and R8 are hydrogen and R4′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. R4′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R4′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R4′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R4′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R4′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R4′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • Figure US20160235758A1-20160818-C00451
  • In another aspect, the invention provides compounds of Subformula IIa′ and IIb′, where W1′ is CR3′, W2 is CR4′, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′. In one embodiment, R1′, R3′, R4′, R5′, R7 and R8 are hydrogen. In another embodiment, R1′, R4′, R5′, R7′ and R8′ are hydrogen and R3′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. R3′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R3′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R3′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R3′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R3′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R3′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds of Formula II wherein R3′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R3′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R3′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula II, R3′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R3′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R3′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R3′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R3′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R3′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R3′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3. In some embodiments R3′ can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00452
  • In another aspect, the invention provides compounds of Subformula IIb:
  • Figure US20160235758A1-20160818-C00453
  • In one embodiment, R1′, R4′, R5′ and R8 are hydrogen. In another embodiment, R1′, R5′, and R8′ are hydrogen and R4′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. R4′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R4′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R4′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R4′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R4′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R4′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In another aspect, the invention provides compounds of Subformula IIc and IId, where W1′ is N, W2′ is CR4′, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′:
  • Figure US20160235758A1-20160818-C00454
  • In one embodiment, R1′, R4′, R5′, R7′ and R8′ are hydrogen. In another embodiment, R1′, R5′, R7 and R8 are hydrogen and R4′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. R4′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R4′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R4′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R4′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R4′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R4′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds of Formula II wherein R4′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R4′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R4′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula II, R4′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R3′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R4′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R4′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R4′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R4′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R4′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3. In some embodiments R4′ can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00455
  • In another aspect, the invention provides compounds of Subformula IIe and IIf, where W1′ is CR3′, W2′ is N, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′:
  • Figure US20160235758A1-20160818-C00456
  • In one embodiment, R1′, R3′, R5′, R7′ and R8′ are hydrogen. In another embodiment, R1′, R5′, R7 and R8 are hydrogen and R3′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. R3′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R3′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R3′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R3′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R3′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R3′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds of Formula II wherein R3′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R3′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R3′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula II, R3′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R3′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R3′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R3′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R3′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R3′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R3′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3. In some embodiments R3′ can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00457
  • In some embodiments, the substituents R3′, R4′, R5′, R6′ or R8′ may be any of the substituents shown in Table 1:
  • TABLE 1
    R3′, R4′, R5′, R6′, R8′ moieties of the compounds of Formula II, each
    independently includes but is not limited to the following:
    Sub-
    class
    # R
    R-1
    Figure US20160235758A1-20160818-C00458
    R-2
    Figure US20160235758A1-20160818-C00459
    R-3
    Figure US20160235758A1-20160818-C00460
    R-4
    Figure US20160235758A1-20160818-C00461
    R-5
    Figure US20160235758A1-20160818-C00462
    R-6
    Figure US20160235758A1-20160818-C00463
    R-7
    Figure US20160235758A1-20160818-C00464
    R-8
    Figure US20160235758A1-20160818-C00465
    R-9 —CH(CH3)2
    R-10
    Figure US20160235758A1-20160818-C00466
    R-11
    Figure US20160235758A1-20160818-C00467
    R-12
    Figure US20160235758A1-20160818-C00468
    R-13
    Figure US20160235758A1-20160818-C00469
    R-14
    Figure US20160235758A1-20160818-C00470
    R-15
    Figure US20160235758A1-20160818-C00471
    R-16
    Figure US20160235758A1-20160818-C00472
    R-17
    Figure US20160235758A1-20160818-C00473
    R-18
    Figure US20160235758A1-20160818-C00474
    R-19
    Figure US20160235758A1-20160818-C00475
    R-20
    Figure US20160235758A1-20160818-C00476
    R-21
    Figure US20160235758A1-20160818-C00477
    R-22
    Figure US20160235758A1-20160818-C00478
    R-23
    Figure US20160235758A1-20160818-C00479
    R-24
    Figure US20160235758A1-20160818-C00480
    R-25
    Figure US20160235758A1-20160818-C00481
    R-26
    Figure US20160235758A1-20160818-C00482
    R-27
    Figure US20160235758A1-20160818-C00483
    R-28
    Figure US20160235758A1-20160818-C00484
    R-29
    Figure US20160235758A1-20160818-C00485
    R-30
    Figure US20160235758A1-20160818-C00486
    R-31
    Figure US20160235758A1-20160818-C00487
    R-32
    Figure US20160235758A1-20160818-C00488
    R-33
    Figure US20160235758A1-20160818-C00489
    R-34
    Figure US20160235758A1-20160818-C00490
    R-35
    Figure US20160235758A1-20160818-C00491
    R-36
    Figure US20160235758A1-20160818-C00492
    R-37
    Figure US20160235758A1-20160818-C00493
    R-38
    Figure US20160235758A1-20160818-C00494
    R-39
    Figure US20160235758A1-20160818-C00495
    R-40 H
    R-41
    Figure US20160235758A1-20160818-C00496
    R-42
    Figure US20160235758A1-20160818-C00497
    R-43
    Figure US20160235758A1-20160818-C00498
    R-44
    Figure US20160235758A1-20160818-C00499
    R-45
    Figure US20160235758A1-20160818-C00500
    R-46
    Figure US20160235758A1-20160818-C00501
    R-47
    Figure US20160235758A1-20160818-C00502
    R-48
    Figure US20160235758A1-20160818-C00503
    R-49
    Figure US20160235758A1-20160818-C00504
    R-50
    Figure US20160235758A1-20160818-C00505
    R-51
    Figure US20160235758A1-20160818-C00506
    R-52
    Figure US20160235758A1-20160818-C00507
    R-53
    Figure US20160235758A1-20160818-C00508
    R-54
    Figure US20160235758A1-20160818-C00509
    R-55
    Figure US20160235758A1-20160818-C00510
    R-56
    Figure US20160235758A1-20160818-C00511
    R-57
    Figure US20160235758A1-20160818-C00512
    R-58
    Figure US20160235758A1-20160818-C00513
    R-59
    Figure US20160235758A1-20160818-C00514
    R-60
    Figure US20160235758A1-20160818-C00515
    R-61
    Figure US20160235758A1-20160818-C00516
    R-62
    Figure US20160235758A1-20160818-C00517
    R-63
    Figure US20160235758A1-20160818-C00518
    R-64
    Figure US20160235758A1-20160818-C00519
    R-65
    Figure US20160235758A1-20160818-C00520
    R-66
    Figure US20160235758A1-20160818-C00521
    R-67
    Figure US20160235758A1-20160818-C00522
    R-68
    Figure US20160235758A1-20160818-C00523
    R-69
    Figure US20160235758A1-20160818-C00524
    R-70
    Figure US20160235758A1-20160818-C00525
    R-71
    Figure US20160235758A1-20160818-C00526
    R-72
    Figure US20160235758A1-20160818-C00527
    R-73
    Figure US20160235758A1-20160818-C00528
    R-74
    Figure US20160235758A1-20160818-C00529
    R-75
    Figure US20160235758A1-20160818-C00530
    R-76
    Figure US20160235758A1-20160818-C00531
    R-77
    Figure US20160235758A1-20160818-C00532
    R-78
    Figure US20160235758A1-20160818-C00533
    R-79
    Figure US20160235758A1-20160818-C00534
    R-80
    Figure US20160235758A1-20160818-C00535
    R-81
    Figure US20160235758A1-20160818-C00536
  • In another aspect, the invention provides a PI3Kα inhibitor which is a compound of Formula III:
  • Figure US20160235758A1-20160818-C00537
  • or its pharmaceutically acceptable salts thereof, where:
  • X is O or S or N;
  • W1 is S, N, NR3′ or CR3′, W2′ is N or CR4′, W3′ is S, N or CR5′, W4′ is N or C, and W7′ is N or C, wherein no more than two N atoms and no more than two C═O groups are adjacent;
  • W5′ is N or CR7′;
  • W6′ is N or CR8′;
  • R1′ and R2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety; and
  • R5′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments, the compound of Formula III exists as a tautomer, and such tautomers are contemplated by the present invention.
  • In some embodiments, the PI3Kα inhibitor is a compound of Formula III which has the Formula:
  • Figure US20160235758A1-20160818-C00538
  • In yet other embodiments, W1 is CR3′, W2′ is CR4′, W3′ is N, W4′ is N, W5′ is CR7′, and W6′ is CR8′. In other embodiments, W1 is CR3′, W2′ is CR4′, W3′ is N, W4′ is N, W5′ is CR7′, and W6′ is CR8′. In other embodiments, W1 is CR3′, W2′ is CR4′, W3′ is N, W4′ is N, W5′ is N, and W6′ is CR8′. In still other embodiments, W1 is NR3′, W2′ is CR4′, W3′ is N, W4′ is C, W5′ is CR7′, and W6′ is CR8′. In other embodiments, W1 is S, W2 is CR4′, W3′ is N, W4′ is C, W5′ is CR7′, and W6′ is CR8′. In other embodiments, W1 is CR3′, W2′ is CR4′, W3′ is S, W4′ is C, W5′ is N, and W6 is N.
  • In other embodiments, an inhibitor of Formula III is a compound according to one of the formulas:
  • Figure US20160235758A1-20160818-C00539
    Figure US20160235758A1-20160818-C00540
  • wherein for each of the above formulas, each respective R variable includes a ‘prime’ 0.
  • In some embodiments, X is O. In other embodiments, X is S.
  • In some embodiments, R1 is hydrogen. In other embodiments, R1 is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″, wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments, R2 is hydrogen. In other embodiments, R2 is, for example, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R2 is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R2′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R2′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R2′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R2′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds wherein R2′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R2′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R2′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula III, R2′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R2′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R2′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R2′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R2′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R2′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R2′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3.
  • In some embodiments of the compound of Formula III, W1′ is CR3. R3′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R3′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R3′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R3′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R3′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R3′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds of Formula III wherein R3′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R3′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R3′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula III, R3′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R3′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R3′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R3′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R3′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R3′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R3′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3.
  • R3′ of the compounds of Formula III, can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00541
  • The invention also provides compounds of Formula III, wherein when R3′ is a member of the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, and NR′R″ (wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety), then R3′ is optionally substituted with one or more of the following substituents: alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, heterocycloalkyloxy, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. Each of the above substituents may be further substituted with one or more substituents chosen from the group consisting of alkyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, oxo, phosphate, urea, and carbonate.
  • For example, the invention provides compounds wherein when R3′ is alkyl, the alkyl is substituted with NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety. The cyclic moiety so formed can be unsubstituted or substituted. Non-limiting exemplary cyclic moieties includes but are not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, and thiomorpholinyl. In other examples of the compounds of Formula III, when R3′ is alkyl, the alkyl is substituted with heterocycloalkyl, which includes oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolyl, tetrahydropyranyl, piperidinyl, morpholinyl, and piperazinyl. All of the above listed heterocycloaklyl substituents can be unsubstituted or substituted.
  • In yet other examples of the compounds of Formula III, when R3′ is alkyl, the alkyl is substituted with a 5, 6, 7, 8, 9, or 10 membered monocyclic or bicyclic heteroaryl, which is unsubstituted or substituted. The monocyclic heteroaryl includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. The bicyclic heteroaryl includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In other embodiments of the compound of Formula III, R3′ is NHR3′, —N(CH3)R3′, —N(CH2CH3)R3′, —N(CH(CH3)2)R3′, or OR3′, wherein R3′ is unsubstituted or substituted heterocycloalkyl (nonlimiting examples thereof include 4-NH piperidin-1-yl, 4-methyl piperidin-1-yl, 4-ethyl piperidin-1-yl, 4-isopropyl-piperidin-1-yl, and pyrrolidin-3-yl), unsubstituted or substituted monocyclic aryl, or unsubstituted or substituted monocyclic heteroaryl (including but not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl). In one example, R3′ is O-aryl, i.e. phenoxy. In another example, R3′ is —O-(4-methyl)piperidin-1-yl or —O-(4-isopropyl)piperidin-1-yl.
  • In some embodiments of the compound of Formula III, R3′ is one of the following moieties:
  • Figure US20160235758A1-20160818-C00542
    Figure US20160235758A1-20160818-C00543
    Figure US20160235758A1-20160818-C00544
    Figure US20160235758A1-20160818-C00545
    Figure US20160235758A1-20160818-C00546
    Figure US20160235758A1-20160818-C00547
    Figure US20160235758A1-20160818-C00548
  • In some embodiments of the compound of Formula III, W1′ is NR3′, wherein R3′ is hydrogen, unsubstituted or substituted C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or unsubstituted or substituted C3-C7cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl). In other embodiments of the compound of Formula III, R3′ is unsubstituted or substituted heterocycloalkyl (which includes but is not limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, and piperazinyl), or unsubstituted or substituted C2-C10heteroalkyl (which includes but is not limited to methoxyethoxy, methoxymethyl, and diethylaminoethyl). Alternatively, R3′ is unsubstituted or substituted monocyclic heteroaryl (which includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl) or unsubstituted or substituted monocyclic aryl.
  • In other embodiments, W1′ is N. In still other embodiments, W1′ is S.
  • In some embodiments of the compound of Formula III, W2′ is CR4′. R4′ can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R4′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R4′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R4′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R4′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R4′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • The present invention also provides compounds of Formula III wherein R4′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R4′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R4′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula III, R4′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R4′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R4′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R4′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In some embodiments, R4′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R4′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, or carbonate. Also contemplated are R4′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3. R4′ of the compounds of Formula III, can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00549
  • The invention also provides compounds of Formula III, wherein when R4′ is a member of the group consisting of alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, and NR′R″ (wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety), then R4′ is optionally substituted with one or more of the following substituents: alkyl, alkenyl, alkynyl, cycloalkyl, heteroalkyl, heterocycloalkyl, heterocycloalkyloxy, aryl, arylalkyl, heteroaryl, heteroarylalkyl, acyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. Each of the above substituents may be further substituted with one or more substituents chosen from the group consisting of alkyl, alkoxy, amido, amino, sulfonamido, acyloxy, alkoxycarbonyl, halo, cyano, hydroxy, nitro, oxo, phosphate, urea, and carbonate.
  • For example, the invention provides compounds wherein when R4′ is alkyl, the alkyl is substituted with NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety. The cyclic moiety so formed can be unsubstituted or substituted. Non-limiting exemplary cyclic moieties includes but are not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1, 2, dioxide, and thiomorpholinyl. In other examples of the compounds of Formula III, when R4′ is alkyl, the alkyl is substituted with heterocycloalkyl, which includes oxetanyl, azetidinyl, tetrahydrofuranyl, pyrrolyl, tetrahydropyranyl, piperidinyl, morpholinyl, and piperazinyl. All of the above listed heterocycloaklyl substituents can be unsubstituted or substituted.
  • In yet other examples of the compounds of Formula III, when R4′ is alkyl, the alkyl is substituted with a 5, 6, 7, 8, 9, or 10 membered monocyclic or bicyclic heteroaryl, which is unsubstituted or substituted. The monocyclic heteroaryl includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. The bicyclic heteroaryl includes but is not limited benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. In some embodiments of the compound of Formula III, W2′ is N.
  • In some embodiments R3′ and R4′ taken together form a cyclic moiety. Such a moiety may have, for example, from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted. In some embodiments, the substituent is C1-C10alkyl (which includes but is not limited to —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl), or C3-C7cycloalkyl (which includes but is not limited to cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl); heterocycloalkyl (which includes but is not limited to oxetanyl, tetrahydrofuranyl, pyrrolidinyl, tetrahydropyranyl, piperidinyl, and piperazinyl), C2-C10heteroalkyl (which includes but is not limited to methoxyethoxy, methoxymethyl, and diethylaminoethyl); monocyclic heteroaryl (which includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl) or unsubstituted or substituted monocyclic aryl. The cyclic moiety may have one or more substituents, which may be the same or different.
  • In some embodiments, the cyclic moiety formed by R3′ and R4′ is substituted with at least one of the following substituents:
  • Figure US20160235758A1-20160818-C00550
    Figure US20160235758A1-20160818-C00551
    Figure US20160235758A1-20160818-C00552
    Figure US20160235758A1-20160818-C00553
    Figure US20160235758A1-20160818-C00554
    Figure US20160235758A1-20160818-C00555
    Figure US20160235758A1-20160818-C00556
  • In some embodiments of the compound of Formula III, W3′ is CR5′. R5′ can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In one embodiment, R5′ is H. In other embodiments, R5′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R5′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R5′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R5′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R5′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. In some embodiments of the compound of Formula III, W3′ is N. In other embodiments, W3′ is S.
  • In some embodiments of the compound of Formula III, W4′ is C. In other embodiments, W4′ is N.
  • In some embodiments of the compound of Formula III, W5′ is N. In other embodiments of the compound of Formula III, W5′ is CR7′. R7 can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In one embodiment, R7 is H. In other embodiments, R7 is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R7 is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R7 is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R7 includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R7 includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In some embodiments of the compound of Formula III, W6′ is N. In other embodiments of the compound of Formula III, W6′ is CR8′. R8 can be, for example, hydrogen, or unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, and heptyl). In one embodiment, R8 is H. In other embodiments, R8 is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R8 is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R8 is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R8 includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R8 includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl.
  • In some embodiments of the compound of Formula III, W7′ is C. In other embodiments, W7′ is N.
  • The invention also provides compounds of Formula III which are defined as defined by the following subclasses:
  • Figure US20160235758A1-20160818-C00557
    Figure US20160235758A1-20160818-C00558
  • wherein for each of the above formulas, each respective R variable includes a ‘prime’ (′).
  • In some embodiments of compounds of Subclasses IIIa-IIIj, R1′ is hydrogen. In other embodiments of compounds of Subclasses IIIa-IIIl, R2′ is NH2 of NHCO(alkyl). In other embodiments of compounds of Subclasses IIIa-IIIl, R4′ is hydrogen. In other embodiments of compounds of Subclasses IIIc-IIIf and IIIi-IIIl, R7 is hydrogen. In other embodiments of compounds of Subclasses IIIa-IIIh and IIIk-IIIl, R8 is hydrogen.
  • In some embodiments of compounds of Subclasses IIIa through IIIl, R3′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety. R3′ can be, for example, hydrogen, unsubstituted or substituted alkyl (including but not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, ten-butyl, sec-butyl, pentyl, hexyl, and heptyl). In other embodiments, R3′ is unsubstituted or substituted alkenyl (including but not limited to unsubstituted or substituted C2-C5alkenyl such as, for example, vinyl, allyl, 1-methyl propen-1-yl, butenyl, or pentenyl) or unsubstituted or substituted alkynyl (including but not limited to unsubstituted or substituted C2-C5alkynyl such as acetylenyl, propargyl, butynyl, or pentynyl). Alternatively, R3′ is unsubstituted or substituted aryl (including but not limited to monocyclic or bicyclic aryl) or unsubstituted or substituted arylalkyl (including but not limited to monocyclic or bicyclic aryl linked to alkyl wherein alkyl includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl). In some other embodiments, R3′ is unsubstituted or substituted heteroaryl, including but not limited to monocyclic and bicyclic heteroaryl. Monocyclic heteroaryl R3′ includes but is not limited to pyrrolyl, thienyl, furyl, pyridinyl, pyranyl, pyrimidinyl, pyrazinyl, pyridazinyl, imidazolyl, thiazolyl, pyrazolyl, and oxazolyl. Bicyclic heteroaryl R3′ includes but is not limited to benzothiophenyl, benzofuryl, indolyl, quinolinyl, isoquinolinyl, benzimidazolyl, benzoxazolyl, benzothiazolyl, quinazolinyl, azaindolyl, pyrazolopyrimidinyl, purinyl, pyrrolo [1, 2-b]pyridazinyl, pyrrolopyrimidinyl, indazolyl, pyrazolylpyridinyl, imidazo[1, 2-a]pyridinyl, and pyrrolo[1, 2-f][1, 2, 4]triazinyl. The present invention also provides compounds of Formula II wherein R3′ is unsubstituted or substituted heteroarylalkyl, including but not limited to monocyclic and bicyclic heteroaryl as described above, that are linked to alkyl, which in turn includes but is not limited to CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, sec-butyl, and pentyl. In some embodiments, R3′ is unsubstituted or substituted cycloalkyl (including but not limited to cyclopropyl, cyclobutyl, and cyclopentyl) or unsubstituted or substituted heteroalkyl (non-limiting examples include ethoxymethyl, methoxymethyl, and diethylaminomethyl). In some further embodiments, R3′ is unsubstituted or substituted heterocycloalkyl which includes but is not limited to pyrrolidinyl, tetrahydrofuranyl, piperidinyl, tetrahydropyranyl, thiazolidinyl, imidazolidinyl, morpholinyl, and piperazinyl. In yet other embodiments of the compounds of Formula II, R3′ is unsubstituted or substituted alkoxy including but not limited to C1-C4alkoxy such as methoxy, ethoxy, propoxy or butoxy. R3′ can also be unsubstituted or substituted heterocycloalkyloxy, including but not limited to 4-NH piperidin-1-yl-oxy, 4-methyl piperidin-1-yl-oxy, 4-ethyl piperidin-1-yl-oxy, 4-isopropyl-piperidin-1-yl-oxy, and pyrrolidin-3-yl-oxy. In other embodiments, R3′ is unsubstituted or substituted amino, wherein the substituted amino includes but is not limited to dimethylamino, diethylamino, di-isopropyl amino, N-methyl N-ethyl amino, and dibutylamino. In some embodiments, R3′ is unsubstituted or substituted acyl, unsubstituted or substituted acyloxy, unsubstituted or substituted C1-C4acyloxy, unsubstituted or substituted alkoxycarbonyl, unsubstituted or substituted amido, or unsubstituted or substituted sulfonamido. In other embodiments, R3′ is halo, which is —I, —F, —Cl, or —Br. In some embodiments, R3′ is selected from the group consisting of cyano, hydroxy, nitro, phosphate, urea, and carbonate. Also contemplated are R3′ being —CH3, —CH2CH3, n-propyl, isopropyl, n-butyl, tert-butyl, sec-butyl, pentyl, hexyl, heptyl, —OCH3, —OCH2CH3, or —CF3. In some embodiments R3′ can also be NR′R″ wherein R′ and R″ are taken together with the nitrogen to form a cyclic moiety having from 3 to 8 ring atoms. The cyclic moiety so formed may further include one or more heteroatoms which are selected from the group consisting of S, O, and N. The cyclic moiety so formed is unsubstituted or substituted, including but not limited to morpholinyl, azetidinyl, pyrrolidinyl, piperidinyl, piperazinyl, isothiazolidinyl 1,2, dioxide, and thiomorpholinyl. Further non-limiting exemplary cyclic moieties are the following:
  • Figure US20160235758A1-20160818-C00559
  • The invention further provides a PI3Kα inhibitor which is a compound of Formula IV:
  • Figure US20160235758A1-20160818-C00560
  • or its pharmaceutically acceptable salts thereof, wherein
  • W1′ is CR3′, W2′ is C-benzoxazolyl substituted with R2 and W3′ is S;
  • W1′ is CR3′, W2′ is C-benzoxazolyl substituted with R2 and W3′ is CR5;
  • W1′ is N or NR3′, W2′ is CR4′, and W3′ is C-benzoxazolyl substituted with R2;
  • W1′ is CR3′, W2′ is CR4′, and W3′ is C-benzoxazolyl substituted with R2; or
  • W1′ is N or NR3′, W2′ is NR4′, and W3′ is C-benzoxazolyl substituted with R2;
  • X is N;
  • R1 and R2 are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R5′, R6′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments of the compound of Formula IV, the compound is:
  • Figure US20160235758A1-20160818-C00561
  • and wherein W1′ is CR3′ or NR3′ and W2 is CR4′.
  • In another aspect, the invention provides a PI3Kα inhibitor which is a compound of Formula V:
  • Figure US20160235758A1-20160818-C00562
  • or its pharmaceutically acceptable salts thereof, wherein:
  • W1′ is N3′, CR3′, or C═O; W2, NR4′, CR4′, or C═O; W3′ is N, NR5′, or CR5′; W4′ is N, C═O or CR6′, wherein no more than two N atoms and no more than two C═O groups are adjacent;
  • W5′ is N or CR7′;
  • W6′ is N or CR8′;
  • Wa′ and Wb′ are independently N or CR9′;
  • one of We′ and Wd′, is N, and the other is O, NR10′, or S;
  • R1′ and R2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety;
  • R5′, R6′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R9′ is alkyl or halo; and
  • R10′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments of the compound of Formula IV, W1′ is CR3′, W2′ is CR4′, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′; W1′ is N, W2′ is CR4′, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′; or W1′ is CR3′, W2′ is N, W3′ is CR5′, W4′ is N, W5′ is CR7′, and W6′ is CR8′. In some embodiments of the compound of Formula IV, Wb′ is N. In other embodiments, Wa′ is CR9′ and R9′ is alkyl.
  • The invention also provides a PI3Kα inhibitor which is a compound of Formula VI:
  • Figure US20160235758A1-20160818-C00563
  • or its pharmaceutically acceptable salts thereof, wherein
  • W1′ is S, N, NR3′ or CR3′, W2′ is N or CR4′, W3′ is S, N or CR5′, W4′ is N or C, and W7′ is N or C, wherein no more than two N atoms and no more than two C═O groups are adjacent;
  • W5′ is N or CR7′;
  • W6′ is N or CR8′;
  • Wa′ and Wb′ are independently N or CR9′;
  • one of Wc′ and Wd′ is N, and the other is O, NR10′, or S;
  • R1′ and R2′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety; R5′, R7′ and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R9′ is alkyl or halo; and
  • R10 is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments of the compound of Formula VI, Wb′ is CR3′, W2′ is CR4′, W3′ is N, W4′ is N, W5′ is CR7′, and W6′ is CR8′. In other embodiments, W1 is CR3′, W2′ is CR4′, W3′ is N, W4′ is N, W5′ is CR7′, and W6′ is CR8′. In other embodiments, W1 is CR3′, W2′ is CR4′, W3′ is N, W4′ is N, W5′ is N, and W6′ is CR8′. In still other embodiments, W1′ is NR3′, W2′ is CR4′, W3′ is N, W4′ is C, W5′ is CR7′, and W6 is CR8′. In other embodiments, W1 is S, W2 is CR4′, W3′ is N, W4′ is C, W5′ is CR7′, and W6′ is CR8′. In other embodiments, W1′ is CR3′, W2′ is CR4′, W3′ is S, W4′ is C, W5′ is N, and W6 is N.
  • In some embodiments of the compound of Formula VI, Wb′, is N. In other embodiments, Wa′ is CR9′ and R9′ is alkyl.
  • The invention further provides PI3Kα inhibitors which are compounds of Formula VI-A and VI-B:
  • Figure US20160235758A1-20160818-C00564
  • or its pharmaceutically acceptable salts thereof, wherein
  • W1′ is CR3′;
  • R1 and R2 are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • and R3′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • Also provided herein are PI3Kα inhibitors which are compounds of Formula VI-C and VI-D:
  • Figure US20160235758A1-20160818-C00565
  • or its pharmaceutically acceptable salts thereof, wherein
  • Wr is CR3′;
  • W5′ is N or CR7′;
  • Wa′ and Wb′ are independently N or CR9′;
  • one of Wc′ and Wd′, is N, and the other is O, NR10′, or S;
  • R1′ and R2 are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R7′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R9′ is alkyl or halo; and
  • R10′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments of the compound of Formula V-C or V-D, Wb′ is N. In other embodiments, Wa′ is CR9′ and R9′ is alkyl.
  • Also provided herein is a PI3Kα inhibitor which is a compound of Formula VII:
  • Figure US20160235758A1-20160818-C00566
  • or its pharmaceutically acceptable salts thereof, wherein
  • W1′ is CR3′; W2 is CR4′;
  • Wa′ is CH or N;
  • R1′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ is alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R4′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety; and
  • R10′ and R11′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • The invention further provides a PI3Kα inhibitor which is a compound of Formula VIII:
  • Figure US20160235758A1-20160818-C00567
  • or a pharmaceutically acceptable salt thereof, wherein
  • X1 is CR3′, NR3′, or S;
  • X2 is CR4′, NR4′, CR4′
    Figure US20160235758A1-20160818-P00001
    CR5 or CR4′
    Figure US20160235758A1-20160818-P00001
    NR5;
  • X3 and X4 are independently C or N;
  • X5 is CR6′, NR6, or S;
  • X4 is CR7 NR7 CR7′
    Figure US20160235758A1-20160818-P00001
    CR8′, or CR7′
    Figure US20160235758A1-20160818-P00001
    NR8′;
  • Wa′ and Wb′ are independently N or CR9′;
  • one of We′ and Wd′, is N, and the other is O, NR10′, or S;
  • R1′ and R2 are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R3′ and R4′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • or R3′ and R4′ taken together form a cyclic moiety;
  • R5′, R6′, R7′, and R8′ are independently hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety;
  • R9′ is alkyl or halo; and
  • R10′ is hydrogen, alkyl, heteroalkyl, alkenyl, alkynyl, cycloalkyl, heterocycloalkyl, aryl, arylalkyl, heteroaryl, heteroarylalkyl, alkoxy, heterocycloalkyloxy, amido, amino, acyl, acyloxy, alkoxycarbonyl, sulfonamido, halo, cyano, hydroxy, nitro, phosphate, urea, carbonate, or NR′R″ wherein R′ and R″ are taken together with nitrogen to form a cyclic moiety.
  • In some embodiments of the compound of Formula VIII, Wb′ is N. In other embodiments, Wa′ is CR9′ and R9′ is alkyl.
  • In some embodiments, the mTor inhibitor is a compound as described in U.S. Pat. No. 7,651,687 or U.S. Pat. No. 7,585,868; or as described International Patent Applications WO 2007/079164, WO 2007/061737, WO 2007/106503, WO 2007/134828 or WO2011/025889 which are hereby incorporated by reference in their entirety.
  • In other embodiments, the mTor inhibitor is NVP-BEZ235 (Novartis), BGT226 (Novartis), XL765 (Sanofi-Aventis, Exelixis), GDC0980 (Genentech), SF1126 (Semafore), PKI587 (Wyeth), PF04691502 (Pfizer), or GSK2126458 (GlaxoSmithKline). In still other embodiments, the mTor inhibitor is CC223 (Celgene), OSIO27 (OSI Pharmaceuticals), AZD8055 (Astra Zeneca), AZD2014 (Astra Zeneca), or Palomid 529 (Paloma Pharmaceuticals).
  • Structures of exemplary mTor inhibitors are shown below:
  • mTor inhibitor Structure
    NVP-BEZ235
    Figure US20160235758A1-20160818-C00568
    XL765
    Figure US20160235758A1-20160818-C00569
    GDC0980
    Figure US20160235758A1-20160818-C00570
    SF1126
    Figure US20160235758A1-20160818-C00571
    PKI587
    Figure US20160235758A1-20160818-C00572
    PF04691502
    Figure US20160235758A1-20160818-C00573
    GSK2126458
    Figure US20160235758A1-20160818-C00574
    OSI027
    Figure US20160235758A1-20160818-C00575
    AZD8055
    Figure US20160235758A1-20160818-C00576
    P529
    Figure US20160235758A1-20160818-C00577
  • Reaction Schemes—PI3Kα Inhibitor Compounds
  • In general, compounds of the invention may be prepared by the following reaction scheme:
  • Figure US20160235758A1-20160818-C00578
  • For example, compounds of the invention may be prepared by the following reaction schemes:
  • Figure US20160235758A1-20160818-C00579
  • Figure US20160235758A1-20160818-C00580
  • The compounds of the invention may be synthesized via a reaction scheme represented generally in Schemes A′, A″ and B′. The synthesis proceeds via coupling a compound of Formula A with a compound of Formula B to yield a compound of Formula C. The coupling step is typically catalyzed by using a palladium catalyst, including but not limited to palladium tetrakis (triphenylphosphine). The coupling is generally performed in the presence of a suitable base, a nonlimiting example being sodium carbonate. One example of a suitable solvent for the reaction is aqueous dioxane.
  • A compound of Formula A for use in Scheme A′ or A″ has a structure of Formula A, wherein T1 is halo including bromo, chloro, fluoro, and iodo, and wherein the remaining substituents are defined for Formulas I and II of compounds of the invention. For boronic acids and acid derivatives as depicted in Formula B, X is either O or S, and the benzoxazole or benzothiazole moiety can be attached at the 4-, 5-, 6- or 7-position.
  • For a compound of Formula B, G is hydrogen or RG1, wherein RG1 is alkyl, alkenyl, or aryl. Alternatively, B(OG)2 is taken together to form a 5- or 6-membered cyclic moiety. In some embodiments, the compound of Formula B is a compound having a structure of Formula E:
  • Figure US20160235758A1-20160818-C00581
  • wherein G is H or RG1; RG1 is alkyl, alkenyl, or aryl. Alternatively, B(OG)2 is taken together to form a 5- or 6-membered cyclic moiety; and RG2 is H, tert-butyl carbamate, or acyl.
  • Figure US20160235758A1-20160818-C00582
  • Scheme C′ depicts an exemplary scheme for synthesizing a compound of Formula B′ or, optionally, Formula B″ for use in Reaction Scheme C′. M is a heterocyclic moiety such as a benzoxazolyl or benzothiazolyl moiety as described by Formula B. This reaction proceeds via reacting a compound of Formula D with a trialkyl borate or a boronic acid derivative to produce a compound of Formula B′. The trialkyl borate includes but is not limited to triisopropyl borate and the boronic acid derivative includes but is not limited to bis(pinacolato)diboron. The reaction typically is run in the presence of a base, a nonlimiting example being potassium acetate. The reaction may be run in a solvent such as dioxane or tetrahydrofuran.
  • A compound of Formula D for use in Scheme C′ is a compound wherein T2 is halo or another leaving group, and M is as defined above. The compound of Formula B′ may further be converted to a compound of Formula B″ by treatment with an acid such as hydrochloric acid.
  • Some exemplary compounds of Formula B that can be synthesized via Scheme C′ include but are not limited to compounds of the following formulae:
  • Figure US20160235758A1-20160818-C00583
    Figure US20160235758A1-20160818-C00584
    Figure US20160235758A1-20160818-C00585
  • Where desired, deprotection of a substituent (e.g., removal of Boc protection from an amino substituent) on the benzoxazolyl moiety (i.e. M1 of Formula C) is performed after coupling the compound of Formula B to the compound of Formula A.
  • Some exemplary compounds with such protecting groups, include but are not limited to compounds of the following formulae:
  • Figure US20160235758A1-20160818-C00586
  • The following Reaction Schemes illustrate the preparation of several compounds of the invention.
  • Figure US20160235758A1-20160818-C00587
    Figure US20160235758A1-20160818-C00588
  • Figure US20160235758A1-20160818-C00589
    Figure US20160235758A1-20160818-C00590
  • Figure US20160235758A1-20160818-C00591
    Figure US20160235758A1-20160818-C00592
  • Figure US20160235758A1-20160818-C00593
  • Figure US20160235758A1-20160818-C00594
  • Figure US20160235758A1-20160818-C00595
  • Figure US20160235758A1-20160818-C00596
  • Figure US20160235758A1-20160818-C00597
  • Figure US20160235758A1-20160818-C00598
  • Figure US20160235758A1-20160818-C00599
  • Figure US20160235758A1-20160818-C00600
  • Figure US20160235758A1-20160818-C00601
  • Figure US20160235758A1-20160818-C00602
  • Figure US20160235758A1-20160818-C00603
  • Figure US20160235758A1-20160818-C00604
  • Figure US20160235758A1-20160818-C00605
  • Figure US20160235758A1-20160818-C00606
  • Figure US20160235758A1-20160818-C00607
  • Figure US20160235758A1-20160818-C00608
  • Figure US20160235758A1-20160818-C00609
  • Figure US20160235758A1-20160818-C00610
  • Figure US20160235758A1-20160818-C00611
  • Figure US20160235758A1-20160818-C00612
  • Figure US20160235758A1-20160818-C00613
    Figure US20160235758A1-20160818-C00614
  • Figure US20160235758A1-20160818-C00615
  • Figure US20160235758A1-20160818-C00616
  • Figure US20160235758A1-20160818-C00617
  • Figure US20160235758A1-20160818-C00618
  • Figure US20160235758A1-20160818-C00619
  • Figure US20160235758A1-20160818-C00620
  • Figure US20160235758A1-20160818-C00621
  • Figure US20160235758A1-20160818-C00622
    Figure US20160235758A1-20160818-C00623
  • Figure US20160235758A1-20160818-C00624
  • Table 3 shows exemplary PI3Kα inhibitors of the invention.
  • TABLE 3
    In Vitro IC50 data for selected compounds of the invention. The following symbols are used: +
    (greater than 10 microMolar), ++ (less than 10 microMolar), +++ (less than 1 microMolar), and ++++ (less
    than 100 nM).
    PC3 T47D
    mTO pro- pro-
    RC PI3K α PI3K β PI3K δ PI3K γ lifer- lifer- Mass
    IC50 IC50 IC50 IC50 IC50 ation ation Character-
    Structure (nM) (nM) (nM) (nM) (nM) (nM)* (nM) ization
    1
    Figure US20160235758A1-20160818-C00625
    +++ ++++ +++ ++++ ++++ +++ +++ Calcd: 388.1 Found: 389.0 [M + H]+
    2
    Figure US20160235758A1-20160818-C00626
    + +++ + + +++ ++ Calcd: 396.10 Found: 397.0 [M + H]+
    3
    Figure US20160235758A1-20160818-C00627
    +++ ++++ + +++ +++ Calcd: 354.09 Found: 355.0 [M + H]+
    4
    Figure US20160235758A1-20160818-C00628
    +++ ++++ ++++ ++++ ++++ ++++ Calcd: 338.12 Found: 339.0 [M + H]+
    5
    Figure US20160235758A1-20160818-C00629
    +++ +++ + ++ +++ Calcd: 348.1 Found: 349.0 [M + H]+
    6
    Figure US20160235758A1-20160818-C00630
    ++ ++++ ++ +++ ++++ ++ +++ Calcd: 327.11 Found: 328.0 [M + H]+
    7
    Figure US20160235758A1-20160818-C00631
    ++++ ++++ ++++ ++++ ++++ ++++ Calcd: 474.18 Found: 475.0 [M + H]+
    8
    Figure US20160235758A1-20160818-C00632
    + ++ + +++ Calcd: 349.13 Found: 350.0 [M + H]+
    9
    Figure US20160235758A1-20160818-C00633
    + +++ ++ +++ Calcd: 349.13 Found: 350.0 [M + H]+
    10
    Figure US20160235758A1-20160818-C00634
    +++ ++++ +++ ++++ +++ +++ Calcd: 328.11 Found: 329.0 [M + H]+
    11
    Figure US20160235758A1-20160818-C00635
    + ++++ ++ +++ + Calcd: 326.12 Found: 327.0 [M + H]+
    12
    Figure US20160235758A1-20160818-C00636
    + + + + Calcd: 354.09 Found: 355.0 [M + H]+
    13
    Figure US20160235758A1-20160818-C00637
    ++++ +++ +++ +++ Calcd: 354.09 Found: 355.0 [M + H]+
    14
    Figure US20160235758A1-20160818-C00638
    ++ + + ++ Calcd: 344.09 Found: 345.0 [M + H]+
    15
    Figure US20160235758A1-20160818-C00639
    ++++ ++++ ++++ +++ +++ Calcd: 338.12 Found: 339.0 [M + H]+
    16
    Figure US20160235758A1-20160818-C00640
    ++++ ++ ++ +++ +++ Calcd: 338.12 Found: 339.0 [M + H]+
    17
    Figure US20160235758A1-20160818-C00641
    ++++ +++ +++ ++++ Calcd: 354.09 Found: 355.0 [M + H]+
    18
    Figure US20160235758A1-20160818-C00642
    ++ + + ++ Calcd: 338.12 Found: 339.0 [M + H]+
    19
    Figure US20160235758A1-20160818-C00643
    + + + + Calcd: 379.14 Found: 380.0 [M + H]+
    20
    Figure US20160235758A1-20160818-C00644
    ++++ ++++ ++++ Calcd: 327.11 Found: 328.0 [M + H]+
    21
    Figure US20160235758A1-20160818-C00645
    +++ ++ ++ ++ Calcd: 359.17 Found: 360.0 [M + H]+
    22
    Figure US20160235758A1-20160818-C00646
    +++ ++++ ++++ ++++ ++++ Calcd: 347.14 Found: 348.0 [M + H]+
    23
    Figure US20160235758A1-20160818-C00647
    ++++ +++ ++ ++++ Calcd: 360.17 Found: 361.0 [M + H]+
    24
    Figure US20160235758A1-20160818-C00648
    ++++ +++ ++++ ++++ Calcd: 361.15 Found: 362.0 [M + H]+
    25
    Figure US20160235758A1-20160818-C00649
    ++ + + ++
    26
    Figure US20160235758A1-20160818-C00650
    +++ ++++ ++++ ++++ Calcd: 341.13 Found: 342.2 [M + H]+
    27
    Figure US20160235758A1-20160818-C00651
    +++ ++ ++++ Calcd: 347.17 Found: 375.2 [M + H]+
    28
    Figure US20160235758A1-20160818-C00652
    ++++ +++ ++++ Calcd: 346.14 Found: 347.2 [M + H]+
    29
    Figure US20160235758A1-20160818-C00653
    ++++ ++++ ++++ Calcd: 361.15 Found: 362.0 [M + H]+
    30
    Figure US20160235758A1-20160818-C00654
    ++++ ++++ ++++ Calcd: 436.20 Found: 437.2 [M + H]+
    31
    Figure US20160235758A1-20160818-C00655
    ++++ ++++ ++++ Calcd: 338.12 Found: 339.2 [M + H]+
    32
    Figure US20160235758A1-20160818-C00656
    ++ + ++ Calcd: 360.16 Found: 361.2 [M + H]+
    33
    Figure US20160235758A1-20160818-C00657
    ++++ +++ +++ Calcd: 389.19 Found: 390.2 [M + H]+
    34
    Figure US20160235758A1-20160818-C00658
    ++++ +++ ++++ Calcd: 375.7 Found: 376.0 [M + H]+
    35
    Figure US20160235758A1-20160818-C00659
    ++ ++++ ++++ ++++ ++ Calcd: 374.19 Found: 375.0 [M + H]+
    36
    Figure US20160235758A1-20160818-C00660
    ++ ++++ ++ + ++ Calcd: 375.17 Found: 376.0 [M + H]+
    37
    Figure US20160235758A1-20160818-C00661
    ++++ ++++ ++++ Calcd: 327.11 Found: 328.0 [M + H]+
    38
    Figure US20160235758A1-20160818-C00662
    ++++ ++++ ++++ Calcd: 316.11 Found: 317.0 [M + H]+
    39
    Figure US20160235758A1-20160818-C00663
    ++++ ++++ ++++ Calcd: 314.12 Found: 315.0 [M + H]+
    40
    Figure US20160235758A1-20160818-C00664
    ++ ++ + ++ Calcd: 250.09 Found: 251.0 [M + H]+
    41
    Figure US20160235758A1-20160818-C00665
    ++ ++++ +++ +++ ++ Calcd: 330.12 Found: 331.0 [M + H]+
    42
    Figure US20160235758A1-20160818-C00666
    + ++++ +++ +++ ++ Calcd: 425.20 Found: 426.0 [M + H]+
    43
    Figure US20160235758A1-20160818-C00667
    ++++ ++++ ++++
    44
    Figure US20160235758A1-20160818-C00668
    ++ ++++ ++ ++ Calcd: 376.16 Found: 377.0 [M + H]+
    45
    Figure US20160235758A1-20160818-C00669
    ++++ ++++ ++++ Calcd: 406.18 Found: 407.0 [M + H]+
    46
    Figure US20160235758A1-20160818-C00670
    ++++ ++++ ++++ ++++ Calcd: 363.11 Found: 364.0 [M + H]+
    47
    Figure US20160235758A1-20160818-C00671
    ++++ ++++ +++ +++ +++ +++ Calcd: 362.14 Found: 363.0 [M + H]+
    48
    Figure US20160235758A1-20160818-C00672
    ++ + + + Calcd: 334.14 Found: 335.0 [M + H]+
    49
    Figure US20160235758A1-20160818-C00673
    ++ ++++ ++ +++ ++ Calcd: 362.14 Found: 363.0 [M + H]+
    50
    Figure US20160235758A1-20160818-C00674
    + ++++ ++ ++++ ++ Calcd: 382.12 Found: 383.0 [M + H]+
    51
    Figure US20160235758A1-20160818-C00675
    ++++ +++ +++ +++ Calcd: 412.16 Found: 413.0 [M + H]+
    52
    Figure US20160235758A1-20160818-C00676
    ++ ++++ ++++ ++++ Calcd: 412.16 Found: 413.0 [M + H]+
    53
    Figure US20160235758A1-20160818-C00677
    ++ + + + Calcd: 335.14 Found: 336.0 [M + H]+
    54
    Figure US20160235758A1-20160818-C00678
    ++ ++++ ++ ++ ++ ++ Calcd: 363.13 Found: 364.2 [M + H]+
    55
    Figure US20160235758A1-20160818-C00679
    + +++ + + Calcd: 342.12 Found: 363.0 [M + H]+
    56
    Figure US20160235758A1-20160818-C00680
    +++ ++++ +++ + ++++ ++ Calcd: 342.12 Found: 343.0 [M + H]+
    57
    Figure US20160235758A1-20160818-C00681
    +++ +++ + ++ Calcd: 336.13 Found: 337.0 [M + H]+
    58
    Figure US20160235758A1-20160818-C00682
    ++++ ++++ ++++ ++++ Calcd: 328.11 Found: 329.0 [M + H]+
    59
    Figure US20160235758A1-20160818-C00683
    ++++ ++++ +++ ++++ Calcd: 317.10 Found: 318.0 [M + H]+
    60
    Figure US20160235758A1-20160818-C00684
    +++ ++++ ++ +++ ++++ Calcd: 402.18 Found: 403.0 [M + H]+
    61
    Figure US20160235758A1-20160818-C00685
    + + + + Calcd: 328.11 Found: 329.0 [M + H]+
    62
    Figure US20160235758A1-20160818-C00686
    ++++ ++++ ++++ ++++ ++++ Calcd: 328.11 Found: 329.0 [M + H]+
    63
    Figure US20160235758A1-20160818-C00687
    ++ + + + Calcd: 362.14 Found: 363.2 [M + H]+
    64
    Figure US20160235758A1-20160818-C00688
    ++ ++++ +++ + ++++ ++ Calcd: 347.14 Found: 348.0 [M + H]+
    65
    Figure US20160235758A1-20160818-C00689
    + ++++ ++++ ++ +++ ++ Calcd: 360.17 Found: 361.2 [M + H]+
    66
    Figure US20160235758A1-20160818-C00690
    ++ + + Calcd: 403.16 Found: 404.2 [M + H]+
    67
    Figure US20160235758A1-20160818-C00691
    +++ + + Calcd: 439.13 Found: 440.0 [M + H]+
    68
    Figure US20160235758A1-20160818-C00692
    ++++ ++ ++ +++ Calcd: 328.11 Found: 329.0 [M + H]+
    69
    Figure US20160235758A1-20160818-C00693
    ++++ ++ ++ +++ Calcd: 317.10 Found: 318.0 [M + H]+
    70
    Figure US20160235758A1-20160818-C00694
    ++++ ++++ ++++ ++++ Calcd: 354.09 Found: 355.0 [M + H]+
    71
    Figure US20160235758A1-20160818-C00695
    ++++ ++ +++ ++++ Calcd: 339.11 Found: 340.0 [M + H]+
    72
    Figure US20160235758A1-20160818-C00696
    +++ ++++ +++ + ++++ Calcd: 278.08 Found: 279.0 [M + H]+
    73
    Figure US20160235758A1-20160818-C00697
    +++ ++ + + Calcd: 389.19 Found: 390.2 [M + H]+
    74
    Figure US20160235758A1-20160818-C00698
    ++ ++++ ++ + ++ ++ Calcd: 403.20 Found: 404.2 [M + H]+
    75
    Figure US20160235758A1-20160818-C00699
    ++++ ++ + ++ Calcd: 361.15 Found: 362.0 [M + H]+
    76
    Figure US20160235758A1-20160818-C00700
    ++ ++++ + + +++ ++ Calcd: 339.11 Found: 340.0 [M + H]+
    77
    Figure US20160235758A1-20160818-C00701
    +++ Calcd: 320.05 Found: 321.0 [M + H]+
    78
    Figure US20160235758A1-20160818-C00702
    ++++ +++ +++ ++++ Calcd: 328.11 Found: 329.0 [M + H]+
    79
    Figure US20160235758A1-20160818-C00703
    ++ + + + Calcd: 390.18 Found: 391.2 [M + H]+
    80
    Figure US20160235758A1-20160818-C00704
    +++ + + + Calcd: 377.15 Found: 378.0 [M + H]+
    81
    Figure US20160235758A1-20160818-C00705
    ++++ +++ ++++ +++ Calcd: 374.19 Found: 375.2 [M + H]+
    82
    Figure US20160235758A1-20160818-C00706
    ++++ ++ ++++ ++++ Calcd: 402.18 Found: 403.2 [M + H]+
    83
    Figure US20160235758A1-20160818-C00707
    ++++ ++ ++++ ++++ Calcd: 377.15 Found: 378.0 [M + H]+
    84
    Figure US20160235758A1-20160818-C00708
    ++++ ++++ +++ ++++ ++ Calcd: 388.20 Found: 389.2 [M + H]+
    85
    Figure US20160235758A1-20160818-C00709
    ++++ ++++ ++++ ++++ Calcd: 388.16 Found: 389.0 [M + H]+
    86
    Figure US20160235758A1-20160818-C00710
    ++++ +++ +++ ++++ Calcd: 424.13 Found: 425.0 [M + H]+
    87
    Figure US20160235758A1-20160818-C00711
    ++++ +++ ++++ ++++ Calcd: 438.15 Found: 439.0 [M + H]+
    88
    Figure US20160235758A1-20160818-C00712
    ++++ ++++ ++ +++ ++ Calcd: 346.15 Found: 347.2 [M + H]+
    89
    Figure US20160235758A1-20160818-C00713
    ++++ ++ ++++ ++++ Calcd: 428.20 Found: 429.2 [M + H]+
    90
    Figure US20160235758A1-20160818-C00714
    ++++ +++ ++ +++ +++ Calcd: 452.16 Found: 453.0 [M + H]+
    91
    Figure US20160235758A1-20160818-C00715
    + + + + Calcd: 390.18 Found: 391.0 [M + H]+
    92
    Figure US20160235758A1-20160818-C00716
    + + + + Calcd: 377.15 Found: 378.0 [M + H]+
    93
    Figure US20160235758A1-20160818-C00717
    ++ +++ + + +++ Calcd: 286.09 Found: 287.0 [M + H]+
    94
    Figure US20160235758A1-20160818-C00718
    ++++ +++ ++ ++++ Calcd: 342.12 Found: 343.0 [M + H]+
    95
    Figure US20160235758A1-20160818-C00719
    ++++ ++ ++++ ++++ Calcd: 438.15 Found: 439.0 [M + H]+
    96
    Figure US20160235758A1-20160818-C00720
    ++++ ++ +++ ++++ +++ Calcd: 361.15 Found: 362.0 [M + H]+
    97
    Figure US20160235758A1-20160818-C00721
    ++ + + + Calcd: 374.14 Found: 375.0 [M + H]+
    98
    Figure US20160235758A1-20160818-C00722
    ++++ + + ++++ Calcd: 375.11 Found: 396.0 [M + H]+
    99
    Figure US20160235758A1-20160818-C00723
    ++++ ++ +++ +++ ++ Calcd: 361.15 Found: 362.2 [M + H]+
    100
    Figure US20160235758A1-20160818-C00724
    ++++ ++++ ++ +++ +++ Calcd: 390.18 Found: 391.0 [M + H]+
    101
    Figure US20160235758A1-20160818-C00725
    ++++ ++ +++ ++ ++ Calcd: 402.22 Found: 403.0 [M + H]+
    102
    Figure US20160235758A1-20160818-C00726
    ++++ +++ +++ ++++ Calcd: 420.17 Found: 421.0 [M + H]+
    103
    Figure US20160235758A1-20160818-C00727
    ++++ +++ +++ +++ ++ Calcd: 445.22 Found: 446.0 [M + H]+
    104
    Figure US20160235758A1-20160818-C00728
    ++ + + ++ Calcd: 377.16 Found: 378.0 [M + H]+
    105
    Figure US20160235758A1-20160818-C00729
    +++ ++ + +++ Calcd: 364.13 Found: 365.0 [M + H]+
    106
    Figure US20160235758A1-20160818-C00730
    ++ + + ++ Calcd: 391.16 Found: 392.0 [M + H]+
    107
    Figure US20160235758A1-20160818-C00731
    +++ + + ++ Calcd: 411.10 Found: 412.0 [M + H]+
    108
    Figure US20160235758A1-20160818-C00732
    ++++ ++ + ++++ +++ Calcd: 372.13 Found: 373.2 [M + H]+
    109
    Figure US20160235758A1-20160818-C00733
    + + + + Calcd: 399.10 Found: 400.0 [M + H]+
    110
    Figure US20160235758A1-20160818-C00734
    + + + + Calcd: 412.13 Found: 413.0 [M + H]+
    111
    Figure US20160235758A1-20160818-C00735
    ++++ ++ + + Calcd: 362.15 Found: 363.0 [M + H]+
    112
    Figure US20160235758A1-20160818-C00736
    ++++ + ++ +++ +++ Calcd: 440.13 Found: 441.0 [M + H]+
    113
    Figure US20160235758A1-20160818-C00737
    ++++ + + +++ ++ Calcd: 404.16 Found: 405.2 [M + H]+
    114
    Figure US20160235758A1-20160818-C00738
    ++++ + + +++ Calcd: 363.12 Found: 364.0 [M + H]+
    115
    Figure US20160235758A1-20160818-C00739
    ++++ ++++ + ++ ++ Calcd: 376.15 Found: 377.0 [M + H]+
    116
    Figure US20160235758A1-20160818-C00740
    ++++ +++ + +++ +++ Calcd: 363.12 Found: 364.0 [M + H]+
    117
    Figure US20160235758A1-20160818-C00741
    ++++ +++ + +++ ++ Calcd: 347.14 Found: 348.2 [M + H]+
    118
    Figure US20160235758A1-20160818-C00742
    +++ ++++ + ++ Calcd: 360.17 Found: 361.0 [M + H]+
    119
    Figure US20160235758A1-20160818-C00743
    ++ ++++ ++ ++ ++ ++ Calcd: 418.18 Found: 419.0 [M + H]+
    120
    Figure US20160235758A1-20160818-C00744
    ++++ +++ ++ + +++ Calcd: 417.19 Found: 418.0 [M + H]+
    121
    Figure US20160235758A1-20160818-C00745
    + ++++ ++++ ++++ ++++ +++ Calcd: 443.21 Found: 444.2 [M + H]+
    122
    Figure US20160235758A1-20160818-C00746
    + + + + Calcd: 364.13 Found: 365.0 [M + H]+
    123
    Figure US20160235758A1-20160818-C00747
    ++ + + + Calcd: 377.16 Found: 378.2 [M + H]+
    124
    Figure US20160235758A1-20160818-C00748
    +++ +++ + +++ Calcd: 363.13 Found: 364.0 [M + H]+
    125
    Figure US20160235758A1-20160818-C00749
    +++ + ++ +++ Calcd: 379.11 Found: 380.0 [M + H]+
    126
    Figure US20160235758A1-20160818-C00750
    +++ ++ + +++ Calcd: 379.11 Found: 380.0 [M + H]+
    127
    Figure US20160235758A1-20160818-C00751
    +++ ++++ + ++ Calcd: 376.15 Found: 377.2 [M + H]+
    128
    Figure US20160235758A1-20160818-C00752
    ++++ ++ ++++ ++++ Calcd: 375.17 Found: 376.2 [M + H]+
    129
    Figure US20160235758A1-20160818-C00753
    +++ ++++ +++ +++ ++++ +++ Calcd: 529.16 Found: 530.0 [M + H]+
    130
    Figure US20160235758A1-20160818-C00754
    + + + + Calcd: 587.14 Found: 588.0 [M + H]+
    131
    Figure US20160235758A1-20160818-C00755
    +++ +++ +++ +++ Calcd: 374.19 Found: 375.2 [M + H]+
    132
    Figure US20160235758A1-20160818-C00756
    +++ + + ++ Calcd: 375.17 Found: 376.2 [M + H]+
    133
    Figure US20160235758A1-20160818-C00757
    + ++++ +++ +++ +++ ++ Calcd: 390.18 Found: 390.2 [M]+
    134
    Figure US20160235758A1-20160818-C00758
    ++++ +++ +++ ++ +++ Calcd: 417.19 Found: 418.0 [M + H]+
    135
    Figure US20160235758A1-20160818-C00759
    + ++++ +++ + +++ +++ Calcd: 360.13 Found: 361.0 [M + H]+
    136
    Figure US20160235758A1-20160818-C00760
    +++ +++ + ++ Calcd: 374.19 Found: 375.2 [M + H]+
    137
    Figure US20160235758A1-20160818-C00761
    + ++++ +++ +++ +++ +++ Calcd: 416.20 Found: 417.2 [M + H]+
    138
    Figure US20160235758A1-20160818-C00762
    ++++ +++ ++++ +++ + Calcd: 457.22 Found: 458.2 [M + H]+
    139
    Figure US20160235758A1-20160818-C00763
    + ++++ +++ +++ +++ +++ Calcd: 431.21 Found: 432.2 [M + H]+
    140
    Figure US20160235758A1-20160818-C00764
    +++ ++ + + Calcd: 390.18 Found: 391.2 [M + H]+
    141
    Figure US20160235758A1-20160818-C00765
    +++ + + ++ Calcd: 390.18 Found: 391.2 [M + H]+
    142
    Figure US20160235758A1-20160818-C00766
    ++ + + + Calcd: 390.18 Found: 391.2 [M + H]+
    143
    Figure US20160235758A1-20160818-C00767
    + ++++ ++ ++++ ++++ +++ Calcd: 391.16 Found: 392.2 [M + H]+
    144
    Figure US20160235758A1-20160818-C00768
    +++ ++ + + Calcd: 392.14 Found: 393.0 [M + H]+
    145
    Figure US20160235758A1-20160818-C00769
    +++ ++ + ++ Calcd: 376.16 Found: 377.2 [M + H]+
    146
    Figure US20160235758A1-20160818-C00770
    ++ ++ + + Calcd: 392.14 Found: 393.0 [M + H]+
    147
    Figure US20160235758A1-20160818-C00771
    +++ ++ ++ ++ Calcd: 418.18 Found: 419.2 [M + H]+
    148
    Figure US20160235758A1-20160818-C00772
    ++++ + + ++ ++ Calcd: 406.18 Found: 407.2 [M + H]+
    149
    Figure US20160235758A1-20160818-C00773
    ++++ ++++ ++ +++ + Calcd: 360.17 Found: 361.0 [M + H]+
    150
    Figure US20160235758A1-20160818-C00774
    +++ + + + Calcd: 424.16 Found: 425.0 [M + H]+
    151
    Figure US20160235758A1-20160818-C00775
    ++++ ++ +++ ++ ++ Calcd: 388.20 Found: 389.0 [M + H]+
    152
    Figure US20160235758A1-20160818-C00776
    ++++ ++++ ++ +++ ++ Calcd: 374.19 Found: 375.0 [M + H]+
    153
    Figure US20160235758A1-20160818-C00777
    ++++ ++++ +++ +++ +++ Calcd: 400.20 Found: 401.0 [M + H]+
    154
    Figure US20160235758A1-20160818-C00778
    ++++ +++ ++ ++++ +++ Calcd: 403.18 Found: 404.0 [M + H]+
    155
    Figure US20160235758A1-20160818-C00779
    ++++ ++++ +++ ++++ Calcd: 388.20 Found: 389.0 [M + H]+
    156
    Figure US20160235758A1-20160818-C00780
    + ++++ +++ +++ +++ +++ Calcd: 443.21 Found: 444.0 [M + H]+
    157
    Figure US20160235758A1-20160818-C00781
    ++ + + + Calcd: 388.16 Found: 389.0 [M + H]+
    158
    Figure US20160235758A1-20160818-C00782
    ++ + + + Calcd: 375.13 Found: 376.0 [M + H]+
    159
    Figure US20160235758A1-20160818-C00783
    ++++ +++ ++++ +++ ++ Calcd: 390.18 Found: 391.0 [M + H]+
    160
    Figure US20160235758A1-20160818-C00784
    ++++ ++ + + ++ Calcd: 404.20 Found: 405.0 [M + H]+
    161
    Figure US20160235758A1-20160818-C00785
    +++ ++ ++ ++ Calcd: 416.20 Found: 417.0 [M + H]+
    162
    Figure US20160235758A1-20160818-C00786
    +++ + + ++ Calcd: 377.15 Found: 378.0 [M + H]+
    163
    Figure US20160235758A1-20160818-C00787
    +++ ++ ++ ++ Calcd: 446.21 Found: 447.0 [M + H]+
    164
    Figure US20160235758A1-20160818-C00788
    +++ +++ + + Calcd: 377.15 Found: 378.0 [M + H]+
    165
    Figure US20160235758A1-20160818-C00789
    +++ ++ ++ + +++ Calcd: 404.20 Found: 405.0 [M + H]+
    166
    Figure US20160235758A1-20160818-C00790
    +++ ++ + ++ Calcd: 404.20 Found: 405.2 [M + H]+
    167
    Figure US20160235758A1-20160818-C00791
    ++ + + + Calcd: 424.16 Found: 425.0 [M + H]+
    168
    Figure US20160235758A1-20160818-C00792
    ++++ +++ ++ + ++ Calcd: 360.17 Found: 361.0 [M + H]+
    169
    Figure US20160235758A1-20160818-C00793
    ++ ++++ +++ +++ +++ +++ Calcd: 430.18 Found: 431.0 [M + H]+
    170
    Figure US20160235758A1-20160818-C00794
    ++ ++++ +++ +++ ++ +++ Calcd: 443.21 Found: 444.2 [M + H]+
    171
    Figure US20160235758A1-20160818-C00795
    + ++++ +++ +++ +++ +++ Calcd: 430.21 Found: 431.0 [M + H]+
    172
    Figure US20160235758A1-20160818-C00796
    ++++ ++++ ++++ +++ Calcd: 388.20 Found: 389.0 [M + H]+
    173
    Figure US20160235758A1-20160818-C00797
    ++++ +++ ++ ++++ ++ Calcd: 361.15 Found: 362.0 [M + H]+
    174
    Figure US20160235758A1-20160818-C00798
    ++++ +++ ++ ++++ ++ Calcd: 342.12 Found: 343.0 [M + H]+
    175
    Figure US20160235758A1-20160818-C00799
    ++++ + + ++ + Calcd: 345.16 Found: 346.0 [M + H]+
    176
    Figure US20160235758A1-20160818-C00800
    ++++ +++ +++ ++++ +++ Calcd: 331.14 Found: 332.0 [M + H]+
    177
    Figure US20160235758A1-20160818-C00801
    ++++ + + +++ + Calcd: 343.12 Found: 343.0 [M]+
    178
    Figure US20160235758A1-20160818-C00802
    ++++ ++++ +++ +++ Calcd: 360.17 Found: 361.0 [M + H]+
    179
    Figure US20160235758A1-20160818-C00803
    ++++ +++ ++ ++ ++ Calcd: 360.17 Found: 361.0 [M + H]+
    180
    Figure US20160235758A1-20160818-C00804
    +++ + + + Calcd: 338.12 Found: 339.0 [M + H]+
    181
    Figure US20160235758A1-20160818-C00805
    +++ ++ +++ ++++ ++ Calcd: 446.18 Found: 467.0 [M + H]+
    182
    Figure US20160235758A1-20160818-C00806
    ++++ +++ +++ +++ ++ Calcd: 402.22 Found: 403.2 [M + H]+
    183
    Figure US20160235758A1-20160818-C00807
    ++++ ++ ++ ++++ ++ Calcd: 360.11 Found: 307.0 [M + H]+
    184
    Figure US20160235758A1-20160818-C00808
    ++++ ++ + +++ ++ Calcd: 322.11 Found: 323.0 [M + H]+
    185
    Figure US20160235758A1-20160818-C00809
    +++ + + + Calcd: 353.13 Found: 354.0 [M + H]+
    186
    Figure US20160235758A1-20160818-C00810
    ++++ ++ +++ ++++ ++ Calcd: 347.14 Found: 348.0 [M + H]+
    187
    Figure US20160235758A1-20160818-C00811
    ++++ ++ +++ ++++ +++ Calcd: 333.12 Found: 334.0 [M + H]+
    188
    Figure US20160235758A1-20160818-C00812
    ++++ +++ +++ ++++ +++ Calcd: 321.12 Found: 322.0 [M + H]+
    189
    Figure US20160235758A1-20160818-C00813
    ++++ +++ +++ +++ +++ Calcd: 337.12 Found: 338.0 [M + H]+
    190
    Figure US20160235758A1-20160818-C00814
    ++++ + +++ ++++ + Calcd: 370.12 Found: 371.0 [M + H]+
    191
    Figure US20160235758A1-20160818-C00815
    ++++ ++ ++ ++ Calcd: 418.21 Found: 419.0 [M + H]+
    192
    Figure US20160235758A1-20160818-C00816
    ++++ +++ +++ +++ Calcd: 403.18 Found: 404.0 [M + H]+
    193
    Figure US20160235758A1-20160818-C00817
    ++++ +++ ++ ++++ Calcd: 404.16 Found: 405.0 [M + H]+
    194
    Figure US20160235758A1-20160818-C00818
    ++++ + + ++++ Calcd: 353.13 Found: 354.0 [M + H]+
    195
    Figure US20160235758A1-20160818-C00819
    +++ ++ + ++++ Calcd: 339.11 Found: 340.0 [M + H]+
    196
    Figure US20160235758A1-20160818-C00820
    +++ ++ + ++ Calcd: 361.15 Found: 362.0 [M + H]+
    197
    Figure US20160235758A1-20160818-C00821
    ++++ ++ ++ ++++ Calcd: 333.12 Found: 334.0 [M + H]+
    198
    Figure US20160235758A1-20160818-C00822
    ++++ ++ ++ +++ Calcd: 468.16 Found: 469.0 [M + H]+
    199
    Figure US20160235758A1-20160818-C00823
    ++++ ++ ++++ ++++ Calcd: 482.17 Found: 483.0 [M + H]+
    200
    Figure US20160235758A1-20160818-C00824
    ++++ ++ ++ ++++ ++ Calcd: 349.12 Found: 350.0 [M + H]+
    201
    Figure US20160235758A1-20160818-C00825
    ++++ ++ + ++ ++ Calcd: 377.15 Found: 378.0 [M + H]+
    202
    Figure US20160235758A1-20160818-C00826
    ++++ ++ ++ +++ +++ Calcd: 363.13 Found: 364.0 [M + H]+
    203
    Figure US20160235758A1-20160818-C00827
    ++++ ++++ +++ ++++ Calcd: 391.18 Found: 392.0 [M + H]+
    204
    Figure US20160235758A1-20160818-C00828
    ++++ +++ + + Calcd: 376.16 Found: 377.0 [M + H]+
    205
    Figure US20160235758A1-20160818-C00829
    ++++ + + + Calcd: 332.13 Found: 333.0 [M + H]+
    206
    Figure US20160235758A1-20160818-C00830
    ++++ + + +++ Calcd: 320.13 Found: 321.0 [M + H]+
    207
    Figure US20160235758A1-20160818-C00831
    ++++ ++++ +++ ++++ +++ Calcd: 417.19 Found: 418.0 [M + H]+
    208
    Figure US20160235758A1-20160818-C00832
    ++++ +++ +++ ++ ++ Calcd: 417.19 Found: 418.0 [M + H]+
    209
    Figure US20160235758A1-20160818-C00833
    ++++ +++ ++ ++++ ++ Calcd: 347.14 Found: 348.0 [M + H]+
    210
    Figure US20160235758A1-20160818-C00834
    +++ + + + Calcd: 355.12 Found: 356.0 [M + H]+
    211
    Figure US20160235758A1-20160818-C00835
    ++++ ++++ ++++ ++++ +++ Calcd: 403.18 Found: 404.0 [M + H]+
    212
    Figure US20160235758A1-20160818-C00836
    ++++ +++ +++ +++ +++ Calcd: 375.18 Found: 376.0 [M + H]+
    213
    Figure US20160235758A1-20160818-C00837
    ++++ +++ +++ + +++ Calcd: 431.21 Found: 432.0 [M + H]+
    214
    Figure US20160235758A1-20160818-C00838
    ++++ ++++ ++ +++ Calcd: 374.15 Found: 375.0 [M + H]+
    215
    Figure US20160235758A1-20160818-C00839
    ++++ +++ ++ +++ Calcd: 400.16 Found: 401.0 [M + H]+
    216
    Figure US20160235758A1-20160818-C00840
    ++++ +++ ++ ++++ Calcd: 333.12 Found: 334.0 [M + H]+
    217
    Figure US20160235758A1-20160818-C00841
    ++++ +++ +++ +++ Calcd: 431.21 Found: 432.0 [M + H]+
    218
    Figure US20160235758A1-20160818-C00842
    +++ +++ +++ ++ Calcd: 376.20 Found: 377.2 [M + H]+
    219
    Figure US20160235758A1-20160818-C00843
    +++ +++ +++ +++ Calcd: 374.19 Found: 375.0 [M + H]+
    220
    Figure US20160235758A1-20160818-C00844
    ++++ +++ ++++ +++ Calcd: 390.22 Found: 391.2 [M + H]+
    221
    Figure US20160235758A1-20160818-C00845
    ++++ +++ ++++ ++++ Calcd: 388.20 Found: 389.0 [M + H]+
    222
    Figure US20160235758A1-20160818-C00846
    ++++ +++ ++++ +++ Calcd: 404.20 Found: 405.0 [M + H]+
    223
    Figure US20160235758A1-20160818-C00847
    ++++ + + +++ Calcd: 425.20 Found: 426.0 [M + H]+
    224
    Figure US20160235758A1-20160818-C00848
    ++ + + + Calcd: 377.19 Found: 378.0 [M + H]+
    225
    Figure US20160235758A1-20160818-C00849
    ++++ ++++ ++++ ++++ Calcd: 443.21 Found: 444.0 [M + H]+
    226
    Figure US20160235758A1-20160818-C00850
    +++ + ++ ++ Calcd: 429.19 Found: 430.2 [M + H]+
    227
    Figure US20160235758A1-20160818-C00851
    ++++ +++ ++++ ++ Calcd: 443.24 Found: 444.0 [M + H]+
    228
    Figure US20160235758A1-20160818-C00852
    Calcd: 347.14 Found: 348.0 [M + H]+
    229
    Figure US20160235758A1-20160818-C00853
    +++ +++ + ++ Calcd: 328.11 Found: 329.0 [M + H]+
    230
    Figure US20160235758A1-20160818-C00854
    ++ +++ ++ +++ Calcd: 328.11 Found: 329.0 [M + H]+
    231
    Figure US20160235758A1-20160818-C00855
    ++ ++++ +++ ++++ ++++ ++ +++ Calcd: 324.07 Found: 325.0 [M + H]+
    232
    Figure US20160235758A1-20160818-C00856
    ++ +++ +++ ++ ++++ Calcd: 282.06 Found: 283.0 [M + H]+
    233
    Figure US20160235758A1-20160818-C00857
    + + + + ++ Calcd: 227.08 Found: 228.0 [M + H]+
    234
    Figure US20160235758A1-20160818-C00858
    +++ ++++ +++ ++++ ++++ ++++ +++ Calcd: 307.11 Found: 308.0 [M + H]+
    235
    Figure US20160235758A1-20160818-C00859
    ++ ++ + ++ ++ Calcd: 274.05 Found: 275.0 [M + H]+
    236
    Figure US20160235758A1-20160818-C00860
    + + + + Calcd: 265.10 Found: 266.0 [M + H]+
    237
    Figure US20160235758A1-20160818-C00861
    +++ ++ ++ ++ Calcd: 285.12 Found: 286.0 [M + H]+
    238
    Figure US20160235758A1-20160818-C00862
    +++ +++ ++ +++ Calcd: 257.09 Found: 258.0 [M + H]+
    239
    Figure US20160235758A1-20160818-C00863
    ++ +++ ++ ++ Calcd: 339.09 Found: 340.0 [M + H]+
    240
    Figure US20160235758A1-20160818-C00864
    ++ +++ ++ ++++ Calcd: 266.08 Found: 267.0 [M + H]+
    241
    Figure US20160235758A1-20160818-C00865
    ++ +++ +++ ++++ Calcd: 266.08 Found: 267.0 [M + H]+
    242
    Figure US20160235758A1-20160818-C00866
    ++++ ++++ + ++++ Calcd: 324.07 Found: 325.0 [M + H]+
    243
    Figure US20160235758A1-20160818-C00867
    +++ ++++ + ++++ Calcd: 282.06 Found: 283.0 [M + H]+
    244
    Figure US20160235758A1-20160818-C00868
    + + + + Calcd: 308.09 Found: 309.0 [M + H]+
    245
    Figure US20160235758A1-20160818-C00869
    +++ ++ + +++ Calcd: 265.10 Found: 266.0
    246
    Figure US20160235758A1-20160818-C00870
    + + + + Calcd: 266.08 Found: 267.0 [M + H]+
    247
    Figure US20160235758A1-20160818-C00871
    + ++ + ++ Calcd: 265.10 Found: 266.2 [M + H]+
    248
    Figure US20160235758A1-20160818-C00872
    ++ +++ ++ +++ ++ Calcd: 282.06 Found: 283.0 [M + H]+
    249
    Figure US20160235758A1-20160818-C00873
    ++++ ++++ +++ ++++ Calcd: 282.06 Found: 283.0 [M + H]+
    250
    Figure US20160235758A1-20160818-C00874
    + + + ++++ Calcd: 324.07 Found: 325.0 [M + H]+
    251
    Figure US20160235758A1-20160818-C00875
    ++++ ++++ ++++ ++++ Calcd: 324.07 Found: 325.0 [M + H]+
    252
    Figure US20160235758A1-20160818-C00876
    + ++ ++ ++
    253
    Figure US20160235758A1-20160818-C00877
    + ++ ++ ++
    254
    Figure US20160235758A1-20160818-C00878
    +++ + + +++ Calcd: 318.11 Found: 319.0 [M + H]+
    255
    Figure US20160235758A1-20160818-C00879
    ++ ++ ++ ++ Calcd: 375.17 Found: 376.0 [M + H]+
    256
    Figure US20160235758A1-20160818-C00880
    ++ + + + Calcd: 346.14 Found: 347.0 [M + H]+
    257
    Figure US20160235758A1-20160818-C00881
    ++++ + + + ++ Calcd: 374.15 Found: 375.0 [M + H]+
    258
    Figure US20160235758A1-20160818-C00882
    ++++ ++++ + ++++ +++ Calcd: 390.14 Found: 391.0 [M + H]+
    259
    Figure US20160235758A1-20160818-C00883
    +++ + + ++ Calcd: 390.14 Found: 391.0 [M + H]+
    260
    Figure US20160235758A1-20160818-C00884
    +++ + + ++ Calcd: 390.14 Found: 391.0 [M + H]+
    261
    Figure US20160235758A1-20160818-C00885
    ++++ +++ + +++ ++ Calcd: 386.19 Found: 387.0 [M + H]+
    262
    Figure US20160235758A1-20160818-C00886
    ++ ++++ +++ + +++ ++ Calcd: 347.14 Found: 348.0 [M + H]+
    263
    Figure US20160235758A1-20160818-C00887
    ++++ ++ ++ ++ ++ Calcd: 402.18 Found: 403.0 [M + H]+
    264
    Figure US20160235758A1-20160818-C00888
    ++++ ++ ++ +++ +++ Calcd: 363.13 Found: 364.0 [M + H]+
    265
    Figure US20160235758A1-20160818-C00889
    ++++ ++ ++ ++++ +++ Calcd: 363.13 Found: 364.0 [M + H]+
    266
    Figure US20160235758A1-20160818-C00890
    ++++ ++ +++ +++ ++ Calcd: 418.18 Found: 419.2 [M + H]+
    267
    Figure US20160235758A1-20160818-C00891
    ++ ++++ ++ +++ +++ +++ Calcd: 443.21 Found: 444.2 [M + H]+
    268
    Figure US20160235758A1-20160818-C00892
    ++++ ++ ++ +++ +++ Calcd: 443.21 Found: 444.2 [M + H]+
    269
    Figure US20160235758A1-20160818-C00893
    ++++ +++ +++ ++ ++ Calcd: 445.22 Found: 446.2 [M + H]+
    270
    Figure US20160235758A1-20160818-C00894
    ++ ++++ +++ +++ +++ +++ Calcd: 445.22 Found: 446.2 [M + H]+
    271
    Figure US20160235758A1-20160818-C00895
    +++ ++ ++ + Calcd: 389.19 Found: 390.2 [M + H]+
    272
    Figure US20160235758A1-20160818-C00896
    +++ ++ + ++ Calcd: 336.12 Found: 337.0 [M + H]+
    273
    Figure US20160235758A1-20160818-C00897
    +++ ++++ + + ++ +++ Calcd: 402.18 Found: 403.0 [M + H]+
    274
    Figure US20160235758A1-20160818-C00898
    ++ ++++ ++ ++ ++ +++ Calcd: 415.21 Found: 416.2 [M + H]+
    275
    Figure US20160235758A1-20160818-C00899
    ++++ +++ +++ +++ +++ Calcd: 403.21 Found: 404.0 [M + H]+
    276
    Figure US20160235758A1-20160818-C00900
    ++ ++++ ++ +++ +++ +++ Calcd: 469.22 Found: 470.2 [M + H]+
    277
    Figure US20160235758A1-20160818-C00901
    ++ ++++ +++ ++ +++ +++ Calcd: 374.15 Found: 375.0 [M + H]+
    278
    Figure US20160235758A1-20160818-C00902
    ++++ +++ ++ +++ +++ Calcd: 457.22 Found: 458.2 [M + H]+
    279
    Figure US20160235758A1-20160818-C00903
    ++++ ++ ++ ++ +++ Calcd: 459.24 Found: 460.2 [M + H]+
    280
    Figure US20160235758A1-20160818-C00904
    ++ + + + Calcd: 404.20 Found: 405.2 [M + H]+
    281
    Figure US20160235758A1-20160818-C00905
    +++ + ++ + Calcd: 391.20 Found: 392.2 [M + H]+
    282
    Figure US20160235758A1-20160818-C00906
    + + + + + Calcd: 364.13 Found: 365.0 [M + H]+
    283
    Figure US20160235758A1-20160818-C00907
    ++++ + + +++ ++ Calcd: 457.19 Found: 458.0 [M + H]+
    284
    Figure US20160235758A1-20160818-C00908
    ++++ ++ ++ +++ ++ Calcd: 457.19 Found: 458.0 [M + H]+
    285
    Figure US20160235758A1-20160818-C00909
    ++++ ++ +++ +++ +++ Calcd: 471.24 Found: 472.2 [M + H]+
    286
    Figure US20160235758A1-20160818-C00910
    ++++ +++ +++ ++++ +++ Calcd: 443.21 Found: 444.2 [M + H]+
    287
    Figure US20160235758A1-20160818-C00911
    ++++ ++ +++ +++ ++ Calcd: 483.24 Found: 484.2 [M + H]+
    288
    Figure US20160235758A1-20160818-C00912
    +++ +++ +++ ++ ++ Calcd: 420.19 Found: 421.2 [M + H]+
    289
    Figure US20160235758A1-20160818-C00913
    +++ ++ + +++ Calcd: 391.16 Found: 392.0 [M + H]+
    290
    Figure US20160235758A1-20160818-C00914
    +++ + + + Calcd: 405.18 Found: 406.2 [M + H]+
    291
    Figure US20160235758A1-20160818-C00915
    +++ + + + Calcd: 418.21 Found: 419.0 [M + H]+
    292
    Figure US20160235758A1-20160818-C00916
    ++ + + + Calcd: 349.15 Found: 350.0 [M + H]+
    293
    Figure US20160235758A1-20160818-C00917
    ++++ +++ ++++ +++ +++ Calcd: 404.20 Found: 405.0 [M + H]+
    294
    Figure US20160235758A1-20160818-C00918
    ++++ + ++ ++++ ++ Calcd: 384.13 Found: 385.0 [M + H]+
    295
    Figure US20160235758A1-20160818-C00919
    +++ +++ +++ +++ Calcd: 392.20 Found: 393.0 [M + H]+
    296
    Figure US20160235758A1-20160818-C00920
    +++ ++ +++ + Calcd: 390.18 Found: 391.0 [M + H]+
    297
    Figure US20160235758A1-20160818-C00921
    +++ + + ++ Calcd: 441.19 Found: 442.0 [M + H]+
    298
    Figure US20160235758A1-20160818-C00922
    ++ ++++ +++ ++ ++ ++ Calcd: 433.19 Found: 434.0 [M + H]+
    299
    Figure US20160235758A1-20160818-C00923
    ++++ +++ ++++ ++++ +++ Calcd: 433.19 Found: 434.0 [M + H]+
    300
    Figure US20160235758A1-20160818-C00924
    ++ ++ + + Calcd: 328.11 Found: 329.0 [M + H]+
    301
    Figure US20160235758A1-20160818-C00925
    ++ ++ + + Calcd: 328.11 Found: 329.0 [M + H]+
    302
    Figure US20160235758A1-20160818-C00926
    +++ ++ ++++ ++ Calcd: 406.21 Found: 407.2 [M + H]+
    303
    Figure US20160235758A1-20160818-C00927
    ++ ++++ + ++ + +++ Calcd: 431.21 Found: 432.2 [M + H]+
    304
    Figure US20160235758A1-20160818-C00928
    +++ + + + Calcd: 404.20 Found: 405.2 [M + H]+
    305
    Figure US20160235758A1-20160818-C00929
    ++ + + + Calcd: 348.13 Found: 349.0 [M + H]+
    306
    Figure US20160235758A1-20160818-C00930
    ++++ +++ +++ +++ Calcd: 419.21 Found: 420.0 [M + H]+
    307
    Figure US20160235758A1-20160818-C00931
    +++ +++ + + Calcd: 376.15 Found: 377.0 [M + H]+
    308
    Figure US20160235758A1-20160818-C00932
    +++ +++ + ++ Calcd: 376.15 Found: 377.0 [M + H]+
    309
    Figure US20160235758A1-20160818-C00933
    +++ +++ + ++ Calcd: 360.17 Found: 361.0 [M + H]+
    310
    Figure US20160235758A1-20160818-C00934
    + + + + Calcd: 360.17 Found: 361.2 [M + H]+
    311
    Figure US20160235758A1-20160818-C00935
    +++ + + + Calcd: 386.12 Found: 387.0 [M + H]+
    312
    Figure US20160235758A1-20160818-C00936
    +++ ++ +++ ++++ Calcd: 288.07 Found: 289.0 [M + H]+
    313
    Figure US20160235758A1-20160818-C00937
    ++ ++++ ++ ++ ++ Calcd: 441.23 Found: 442.2 [M + H]+
    314
    Figure US20160235758A1-20160818-C00938
    +++ ++ ++ ++ Calcd: 386.19 Found: 387.2 [M + H]+
    315
    Figure US20160235758A1-20160818-C00939
    +++ ++++ ++ ++ ++ Calcd: 402.18 Found: 403.2 [M + H]+
    316
    Figure US20160235758A1-20160818-C00940
    +++ ++ + ++ Calcd: 381.14 Found: 382.0 [M + H]+
    317
    Figure US20160235758A1-20160818-C00941
    + + + + Calcd: 363.13 Found: 364.0 [M + H]+
    318
    Figure US20160235758A1-20160818-C00942
    +++ ++++ ++ + +++ Calcd: 383.15 Found: 384.2 [M + H]+
    319
    Figure US20160235758A1-20160818-C00943
    ++++ +++ ++ +++ Calcd: 383.15 Found: 384.2 [M + H]+
    320
    Figure US20160235758A1-20160818-C00944
    + ++++ ++ ++ ++ Calcd: 397.17 Found: 398.0 [M + H]+
    321
    Figure US20160235758A1-20160818-C00945
    + + + + Calcd: 376.15 Found: 377.0 [M + H]+
    322
    Figure US20160235758A1-20160818-C00946
    + ++++ +++ ++ +++ Calcd: 402.18 Found: 403.2 [M + H]+
    323
    Figure US20160235758A1-20160818-C00947
    + ++++ ++ ++ +++ Calcd: 429.19 Found: 430.2 [M + H]+
    324
    Figure US20160235758A1-20160818-C00948
    + ++++ ++ ++ +++ Calcd: 388.20 Found: 389.2 [M + H]+
    325
    Figure US20160235758A1-20160818-C00949
    ++++ +++ ++ ++++ Calcd: 415.18 Found: 416.2 [M + H]+
    326
    Figure US20160235758A1-20160818-C00950
    + ++++ +++ +++ +++ Calcd: 402.20 Found: 402.2 [M + H]+
    327
    Figure US20160235758A1-20160818-C00951
    + ++++ ++ +++ +++ Calcd: 443.21 Found: 444.2 [M + H]+
    328
    Figure US20160235758A1-20160818-C00952
    + ++++ ++ ++ ++ Calcd: 479.17 Found: 480.2 [M + H]+
    329
    Figure US20160235758A1-20160818-C00953
    + ++++ +++ +++ ++ Calcd: 429.23 Found: 430.2 [M + H]+
    330
    Figure US20160235758A1-20160818-C00954
    +++ +++ ++ + + Calcd: 478.21 Found: 479.2 [M + H]+
    331
    Figure US20160235758A1-20160818-C00955
    + ++++ ++ ++ ++ Calcd: 443.24 Found: 444.2 [M + H]+
    332
    Figure US20160235758A1-20160818-C00956
    +++ ++++ ++ +++ Calcd: 374.19 Found: 375.2 [M + H]+
    333
    Figure US20160235758A1-20160818-C00957
    + ++++ ++ +++ ++ +++ Calcd: 445.22 Found: 446.2 [M + H]+
    334
    Figure US20160235758A1-20160818-C00958
    ++++ ++ ++ ++ +++ Calcd: 450.15 Found: 451.0 [M + H]+
    335
    Figure US20160235758A1-20160818-C00959
    + ++++ ++ ++ + +++ Calcd: 455.24 Found: 456.2 [M + H]+
    336
    Figure US20160235758A1-20160818-C00960
    +++ +++ ++ + Calcd: 374.19 Found: 375.2 [M + H]+
    337
    Figure US20160235758A1-20160818-C00961
    ++++ + + +++ Calcd: 411.18 Found: 412.2 [M + H]+
    338
    Figure US20160235758A1-20160818-C00962
    ++++ ++ ++ ++ ++ Calcd: 425.20 Found: 426.2 [M + H]+
    339
    Figure US20160235758A1-20160818-C00963
    ++++ +++ +++ +++ Calcd: 397.17 Found: 398.2 [M + H]+
    340
    Figure US20160235758A1-20160818-C00964
    +++ + + +++ Calcd: 319.11 Found: 320.2 [M + H]+
    341
    Figure US20160235758A1-20160818-C00965
    +++ + ++ ++ ++ Calcd: 416.20 Found: 417.2 [M + H]+
    342
    Figure US20160235758A1-20160818-C00966
    + + + + Calcd: 360.17 Found: 361.2 [M + H]+
    343
    Figure US20160235758A1-20160818-C00967
    + ++ + + Calcd: 360.17 Found: 361.2 [M + H]+
    344
    Figure US20160235758A1-20160818-C00968
    ++++ ++ +++ ++ Calcd: 425.20 Found: 426.2 [M + H]+
    345
    Figure US20160235758A1-20160818-C00969
    ++ ++++ +++ +++ +++ Calcd: 411.18 Found: 412.2 [M + H]+
    346
    Figure US20160235758A1-20160818-C00970
    ++++ ++ + +++ Calcd: 347.14 Found: 348.2 [M + H]+
    347
    Figure US20160235758A1-20160818-C00971
    +++ ++ + +++ Calcd: 363.13 Found: 364.2 [M + H]+
    348
    Figure US20160235758A1-20160818-C00972
    +++ +++ ++++ ++ +++ Calcd: 420.20 Found: 421.2 [M + H]+
    349
    Figure US20160235758A1-20160818-C00973
    + + + + Calcd: 377.15 Found: 378.2 [M + H]+
    350
    Figure US20160235758A1-20160818-C00974
    + ++++ ++ ++ + Calcd: 415.21 Found: 416.2 [M + H]+
    351
    Figure US20160235758A1-20160818-C00975
    ++ +++ + + Calcd: 374.19 Found: 375.0 [M + H]+
    352
    Figure US20160235758A1-20160818-C00976
    + + + + Calcd: 377.15 Found: 378.2 [M + H]+
    353
    Figure US20160235758A1-20160818-C00977
    + ++ + + + Calcd: 416.20 Found: 417.2 [M + H]+
    354
    Figure US20160235758A1-20160818-C00978
    ++ ++++ ++ ++ ++ +++ Calcd: 397.17 Found: 398.0 [M + H]+
    355
    Figure US20160235758A1-20160818-C00979
    ++++ ++ +++ ++ +++ Calcd: 415.21 Found: 416.2 [M + H]+
    356
    Figure US20160235758A1-20160818-C00980
    ++++ ++++ ++++ ++++ +++ Calcd: 327.11 Found: 328.2 [M + H]+
    357
    Figure US20160235758A1-20160818-C00981
    +++ ++ ++ +++ Calcd: 377.15 Found: 378.2 [M + H]+
    358
    Figure US20160235758A1-20160818-C00982
    ++++ +++ +++ +++ Calcd: 436.14 Found: 437.0 [M + H]+
    359
    Figure US20160235758A1-20160818-C00983
    ++++ +++ ++ ++++ Calcd: 397.09 Found: 398.0 [M + H]+
    360
    Figure US20160235758A1-20160818-C00984
    ++++ +++ ++ ++++ Calcd: 381.12 Found: 382.0 [M + H]+
    361
    Figure US20160235758A1-20160818-C00985
    ++++ +++ ++ +++ Calcd: 420.17 Found: 421.0 [M + H]+
    362
    Figure US20160235758A1-20160818-C00986
    + + + + Calcd: 416.20 Found: 417.0 [M + H]+
    363
    Figure US20160235758A1-20160818-C00987
    + + + +
    364
    Figure US20160235758A1-20160818-C00988
    +++ ++ + +++ Calcd: 377.15 Found: 378.0 [M + H]+
    365
    Figure US20160235758A1-20160818-C00989
    ++ ++ ++ ++ Calcd: 416.20 Found: 417.0 [M + H]+
    366
    Figure US20160235758A1-20160818-C00990
    ++++ ++ + ++ Calcd: 360.17 Found: 361.0 [M + H]+
    367
    Figure US20160235758A1-20160818-C00991
    +++ +++ ++ +++ Calcd: 332.14 Found: 333.0 [M + H]+
    368
    Figure US20160235758A1-20160818-C00992
    ++++ ++ +++ +++ Calcd: 361.15 Found: 362.0 [M + H]+
    369
    Figure US20160235758A1-20160818-C00993
    + + + + Calcd: 377.15 Found: 378.0 [M + H]+
    370
    Figure US20160235758A1-20160818-C00994
    ++ + + + Calcd: 397.09 Found: 398.0 [M + H]+
    371
    Figure US20160235758A1-20160818-C00995
    +++ ++ + + Calcd: 436.14 Found: 437.0 [M + H]+
    372
    Figure US20160235758A1-20160818-C00996
    +++ ++ ++ ++ Calcd: 381.12 Found: 382.2 [M + H]+
    373
    Figure US20160235758A1-20160818-C00997
    + ++++ ++ ++ ++ Calcd: 420.17 Found: 421.2 [M + H]+
    374
    Figure US20160235758A1-20160818-C00998
    +++ ++++ ++ + Calcd: 374.19 Found: 375.2 [M + H]+
    375
    Figure US20160235758A1-20160818-C00999
    +++ ++ +++ +++ Calcd: 416.20 Found: 417.2 [M + H]+
    376
    Figure US20160235758A1-20160818-C01000
    ++++ ++ ++ ++++ Calcd: 416.20 Found: 417.2 [M + H]+
    377
    Figure US20160235758A1-20160818-C01001
    ++ ++++ ++ ++ +++ Calcd: 507.21 Found: 508.2 [M + H]+
    378
    Figure US20160235758A1-20160818-C01002
    +++ ++++ ++++ ++ ++ Calcd: 374.19 Found: 375.2 [M + H]+
    379
    Figure US20160235758A1-20160818-C01003
    ++ ++++ +++ ++ + Calcd: 429.23 Found: 430.2 [M + H]+
    380
    Figure US20160235758A1-20160818-C01004
    +++ ++ + ++ Calcd: 376.16 Found: 377.0 [M + H]+
    381
    Figure US20160235758A1-20160818-C01005
    ++++ +++ ++ ++ Calcd: 429.23 Found: 430.2 [M + H]+
    382
    Figure US20160235758A1-20160818-C01006
    ++++ +++ ++ +++ Calcd: 374.19 Found: 375.2 [M + H]+
    383
    Figure US20160235758A1-20160818-C01007
    +++ + + + Calcd: 416.20 Found: 417.2 [M + H]+
    384
    Figure US20160235758A1-20160818-C01008
    +++ ++++ +++ ++ +++ Calcd: 414.14 Found: 415.2 [M + H]+
    385
    Figure US20160235758A1-20160818-C01009
    ++++ ++ + + Calcd: 359.17 Found: 360.2 [M + H]+
    386
    Figure US20160235758A1-20160818-C01010
    ++++ + + Calcd: 415.21 Found: 416.2 [M + H]+
    387
    Figure US20160235758A1-20160818-C01011
    ++++ +++ + +++ Calcd: 404.20 Found: 405.2 [M + H]+
    388
    Figure US20160235758A1-20160818-C01012
    ++++ ++++ +++ +++ +++ Calcd: 429.23 Found: 430.2 [M + H]+
    389
    Figure US20160235758A1-20160818-C01013
    +++ + + ++ Calcd: 360.13 Found: 361.0 [M + H]+
    390
    Figure US20160235758A1-20160818-C01014
    ++ + Calcd: 374.15 Found: 375.2 [M + H]+
    391
    Figure US20160235758A1-20160818-C01015
    ++ + + + Calcd: 388.16 Found: 389.2 [M + H]+
    392
    Figure US20160235758A1-20160818-C01016
    + + + + Calcd: 402.18 Found: 403.2 [M + H]+
    393
    Figure US20160235758A1-20160818-C01017
    ++ + + + Calcd: 414.18 Found: 415.2 [M + H]+
    394
    Figure US20160235758A1-20160818-C01018
    ++++ ++++ +++ ++++ Calcd: 383.15 Found: 384.2 [M + H]+
    395
    Figure US20160235758A1-20160818-C01019
    ++++ + + ++++ Calcd: 428.16 Found: 429.2 [M + H]+
    396
    Figure US20160235758A1-20160818-C01020
    ++++ ++++ + ++ Calcd: 359.17 Found: 360.2 [M + H]+
    397
    Figure US20160235758A1-20160818-C01021
    ++++ +++ +++ ++++ Calcd: 341.13 Found: 342.2 [M + H]+
    398
    Figure US20160235758A1-20160818-C01022
    +++ ++ ++ ++ Calcd: 446.21 Found: 447.2 [M + H]+
    399
    Figure US20160235758A1-20160818-C01023
    + + + + Calcd: 365.13 Found: 366.2 [M + H]+
    400
    Figure US20160235758A1-20160818-C01024
    ++++ ++ + ++ Calcd: 364.13 Found: 365.2 [M + H]+
    401
    Figure US20160235758A1-20160818-C01025
    + + + ++ Calcd: 325.10 Found: 326.0 [M + H]+
    402
    Figure US20160235758A1-20160818-C01026
    + + + + Calcd: 345.07 Found: 346.0 [M + H]+
    403
    Figure US20160235758A1-20160818-C01027
    ++ + + + Calcd: 375.18 Found: 376.2 [M + H]+
    404
    Figure US20160235758A1-20160818-C01028
    + + + + Calcd: 417.19 Found: 418.2 [M + H]+
    405
    Figure US20160235758A1-20160818-C01029
    + + + + Calcd: 441.11 Found: 442.0 [M + H]+
    406
    Figure US20160235758A1-20160818-C01030
    ++ ++ + + Calcd: 377.15 Found: 378.2 [M + H]+
    407
    Figure US20160235758A1-20160818-C01031
    + ++ + + Calcd: 439.16 Found: 440.2 [M + H]+
    408
    Figure US20160235758A1-20160818-C01032
    + + + + Calcd: 460.22 Found: 461.2 [M + H]+
    409
    Figure US20160235758A1-20160818-C01033
    ++ + + + Calcd: 403.16 Found: 404.2 [M + H]+
    410
    Figure US20160235758A1-20160818-C01034
    ++ + + + Calcd: 391.13 Found: 392.0 [M + H]+
    411
    Figure US20160235758A1-20160818-C01035
    ++++ ++ ++ +++ Calcd: 436.15 Found: 437.2 [M + H]+
    412
    Figure US20160235758A1-20160818-C01036
    ++ + + + Calcd: 364.13 Found: 365.0 [M + H]+
    413
    Figure US20160235758A1-20160818-C01037
    ++++ +++ ++ +++ Calcd: 396.15 Found: 397.2 [M + H]+
    414
    Figure US20160235758A1-20160818-C01038
    ++ + + + Calcd: 353.15 Found: 354.2 [M + H]+
    415
    Figure US20160235758A1-20160818-C01039
    ++ + + + Calcd: 357.10 Found: 358.0 [M + H]+
    416
    Figure US20160235758A1-20160818-C01040
    ++++ ++ ++ +++ Calcd: 431.13 Found: 432.0 [M + H]+
    417
    Figure US20160235758A1-20160818-C01041
    +++ ++++ ++++ ++++ ++++ Calcd: 529.12 Found: 530.0 [M + H]+
    418
    Figure US20160235758A1-20160818-C01042
    ++++ ++++ ++++ ++++ ++++ Calcd: 435.08 Found: 436.0 [M + H]+
    419
    Figure US20160235758A1-20160818-C01043
    +++ ++++ ++++ ++++ ++++ Calcd: 533.07 Found: 534.0 [M + H]+
    420
    Figure US20160235758A1-20160818-C01044
    ++++ + +++ +++ Calcd: 493.14 Found: 494.2 [M + H]+
    421
    Figure US20160235758A1-20160818-C01045
    ++++ +++ ++++ ++++ Calcd: 511.13 Found: 512.0 [M + H]+
    422
    Figure US20160235758A1-20160818-C01046
    ++++ ++ +++ ++++ Calcd: 511.13 Found: 512.0 [M + H]+
    423
    Figure US20160235758A1-20160818-C01047
    ++++ +++ ++++ ++++ Calcd: 511.13 Found: 512.0 [M + H]+
    424
    Figure US20160235758A1-20160818-C01048
    ++++ + ++ +++ Calcd: 463.51 Found: 464.0 [M + H]+
    425
    Figure US20160235758A1-20160818-C01049
    ++++ + +++ ++++ Calcd: 499.49 Found: 500.0 [M + H]+
    426
    Figure US20160235758A1-20160818-C01050
    + + + + Calcd: 307.11 Found: 308.0 [M + H]+
    427
    Figure US20160235758A1-20160818-C01051
    + + + + Calcd: 265.27 Found: 266.0 [M + H]+
    428
    Figure US20160235758A1-20160818-C01052
    ++ ++ + +++ Calcd: 265.10 Found: 266.0 [M + H]+
    429
    Figure US20160235758A1-20160818-C01053
    + + + + Calcd: 385.15 Found: 386.2 [M + H]+
    430
    Figure US20160235758A1-20160818-C01054
    ++ ++ + ++ Calcd: 307.11 Found: 308.0 [M + H]+
    431
    Figure US20160235758A1-20160818-C01055
    + + + + Calcd: 307.11 Found: 308.0 [M + H]+
    432
    Figure US20160235758A1-20160818-C01056
    ++ +++ + ++++ Calcd: 284.07 Found: 285.0 [M + H]+
    433
    Figure US20160235758A1-20160818-C01057
    +++ +++ ++ ++++ Calcd: 284.07 Found: 285.2 [M + H]+
    434
    Figure US20160235758A1-20160818-C01058
    ++++ ++++ ++++ ++++ Calcd: 342.06 Found: 343.0 [M + H]+
    435
    Figure US20160235758A1-20160818-C01059
    ++ ++ + ++++ Calcd: 342.06 Found: 343.0 [M + H]+
    436
    Figure US20160235758A1-20160818-C01060
    ++++ ++++ +++ ++++ Calcd: 300.05 Found: 301.1 [M + H]+
    437
    Figure US20160235758A1-20160818-C01061
    ++ +++ + +++ Calcd: 300.05 Found: 301.2 [M + H]+
    438
    Figure US20160235758A1-20160818-C01062
    ++ + + + Calcd: 381.12 Found: 382.2 [M + H]+
    439
    Figure US20160235758A1-20160818-C01063
    ++ + + + Calcd: 381.12 Found: 382.2 [M + H]+
    440
    Figure US20160235758A1-20160818-C01064
    + + + + Calcd: 462.14 Found: 463.0 [M + H]+
    441
    Figure US20160235758A1-20160818-C01065
    + + + +
    442
    Figure US20160235758A1-20160818-C01066
    + + + + Calcd: 400.12 Found: 401.0 [M + H]+
    443
    Figure US20160235758A1-20160818-C01067
    + + + + Calcd: 400.12 Found: 401.2 [M + H]+
    444
    Figure US20160235758A1-20160818-C01068
    ++ + + + Calcd: 492.15 Found: 493.0 [M + H]+
    445
    Figure US20160235758A1-20160818-C01069
    + + + + Calcd: 430.13 Found: 431.0 [M + H]+
    446
    Figure US20160235758A1-20160818-C01070
    +++ + + + Calcd: 376.16 Found: 377.2 [M + H]+
    447
    Figure US20160235758A1-20160818-C01071
    ++ + + + Calcd: 463.13 Found: 464.0 [M + H]+
    448
    Figure US20160235758A1-20160818-C01072
    +++ + + ++ Calcd: 377.40 Found: 378.2 [M + H]+
    449
    Figure US20160235758A1-20160818-C01073
    +++ +++ + ++++ Calcd: 377.15 Found: 378.2 [M + H]+
    450
    Figure US20160235758A1-20160818-C01074
    ++ + + + Calcd: 401.12 Found: 402.2 [M + H]+
    451
    Figure US20160235758A1-20160818-C01075
    ++++ +++ ++++ ++++ +++ Calcd: 497.95 Found: 498.0 [M]+
    452
    Figure US20160235758A1-20160818-C01076
    ++++ +++ ++++ ++++ ++ Calcd: 515.08 Found: 516.0 [M + H]+
    453
    Figure US20160235758A1-20160818-C01077
    ++ + + ++ Calcd: 462.14 Found: 463.0 [M + H]+
    454
    Figure US20160235758A1-20160818-C01078
    ++ + ++ ++ Calcd: 498.12 Found: 499.0 [M + H]+
    455
    Figure US20160235758A1-20160818-C01079
    ++ + + ++ Calcd: 492.15 Found: 493.2 [M + H]+
    456
    Figure US20160235758A1-20160818-C01080
    + + + + Calcd: 430.13 Found: 431.0 [M + H]+
    457
    Figure US20160235758A1-20160818-C01081
    ++++ ++++ ++++ ++++ +++ Calcd: 515.08 Found: 516.0 [M + H]+
    458
    Figure US20160235758A1-20160818-C01082
    ++++ ++++ ++++ ++++ +++ Calcd: 515.08 Found: 516.0 [M + H]+
    459
    Figure US20160235758A1-20160818-C01083
    +++ + + +++ Calcd: 496.10 Found: 497.0 [M + H]+
    460
    Figure US20160235758A1-20160818-C01084
    ++ + + + Calcd: 434.08 Found: 435.0 [M + H]+
    461
    Figure US20160235758A1-20160818-C01085
    +++ ++ ++ +++ Calcd: 532.08 Found: 533.0 [M + H]+
    462
    Figure US20160235758A1-20160818-C01086
    ++++ ++++ ++++ ++++ +++ Calcd: 494.14 Found: 495.0 [M + H]+
    463
    Figure US20160235758A1-20160818-C01087
    ++++ ++ ++ +++ Calcd: 377.15 Found: 378.2 [M + H]+
    464
    Figure US20160235758A1-20160818-C01088
    ++++ + + +++ ++ Calcd: 377.15 Found: 378.2 [M + H]+
    465
    Figure US20160235758A1-20160818-C01089
    ++++ + + +++ Calcd: 432.19 Found: 433.2 [M + H]+
    466
    Figure US20160235758A1-20160818-C01090
    +++ ++ ++ +++ Calcd: 432.19 Found: 433.2 [M + H]+
    467
    Figure US20160235758A1-20160818-C01091
    ++++ ++ ++ ++++ +++ Calcd: 363.13 Found: 364.0 [M + H]+
    468
    Figure US20160235758A1-20160818-C01092
    ++++ ++ + ++++ Calcd: 351.11 Found: 352.2 [M + H]+
    469
    Figure US20160235758A1-20160818-C01093
    ++++ ++ ++ +++ ++ Calcd: 418.18 Found: 419.2 [M + H]+
    470
    Figure US20160235758A1-20160818-C01094
    ++++ ++ ++ +++ ++ Calcd: 418.18 Found: 419.2 [M + H]+
    471
    Figure US20160235758A1-20160818-C01095
    ++++ + + ++++ ++ Calcd: 383.12 Found: 384.2 [M + H]+
    472
    Figure US20160235758A1-20160818-C01096
    ++++ ++ ++ +++ ++ Calcd: 391.16 Found: 392.2 [M + H]+
    473
    Figure US20160235758A1-20160818-C01097
    ++++ + + ++++ +++ Calcd: 369.10 Found: 370.0 [M + H]+
    474
    Figure US20160235758A1-20160818-C01098
    ++ + + + Calcd: 379.11 Found: 380.2 [M + H]+
    475
    Figure US20160235758A1-20160818-C01099
    ++++ + ++ ++++ Calcd: 365.13 Found: 366.2 [M + H]+
    476
    Figure US20160235758A1-20160818-C01100
    ++++ + + ++++ Calcd: 365.13 Found: 366.2 [M + H]+
    477
    Figure US20160235758A1-20160818-C01101
    ++++ + ++ +++ Calcd: 413.16 Found: 414.2 [M + H]+
    478
    Figure US20160235758A1-20160818-C01102
    ++++ ++ +++ +++ +++ Calcd: 413.16 Found: 414.2 [M + H]+
    479
    Figure US20160235758A1-20160818-C01103
    ++++ + ++ +++ Calcd: 399.14 Found: 400.2 [M + H]+
    480
    Figure US20160235758A1-20160818-C01104
    ++++ + ++ +++ +++ Calcd: 389.15 Found: 390.2 [M + H]+
    481
    Figure US20160235758A1-20160818-C01105
    ++++ ++ ++ +++ Calcd: 361.15 Found: 362.0 [M + H]+
    482
    Figure US20160235758A1-20160818-C01106
    ++++ + ++ +++ Calcd: 361.15 Found: 362.2 [M + H]+
    483
    Figure US20160235758A1-20160818-C01107
    ++++ ++ + ++ ++ Calcd: 377.15 Found: 378.2 [M + H]+
    484
    Figure US20160235758A1-20160818-C01108
    +++ + + + Calcd: 363.13 Found: 364.2 [M + H]+
    485
    Figure US20160235758A1-20160818-C01109
    ++ + + + Calcd: 379.11 Found: 380.2 [M + H]+
    486
    Figure US20160235758A1-20160818-C01110
    ++ + + ++ Calcd: 377.15 Found: 378.2 [M + H]+
    487
    Figure US20160235758A1-20160818-C01111
    + + + + Calcd: 362.15 Found: 363.2 [M + H]+
    488
    Figure US20160235758A1-20160818-C01112
    + + + + Calcd: 363.13 Found: 364.2 [M + H]+
    489
    Figure US20160235758A1-20160818-C01113
    ++ + + + Calcd: 379.11 Found: 380.0 [M + H]+
    490
    Figure US20160235758A1-20160818-C01114
    + + + + Calcd: 362.15 Found: 363.0 [M + H]+
    491
    Figure US20160235758A1-20160818-C01115
    + + + + Calcd: 363.13 Found: 364.2 [M + H]+
    492
    Figure US20160235758A1-20160818-C01116
    +++ ++ ++ ++ Calcd: 348.13 Found: 349.2 [M + H]+
    493
    Figure US20160235758A1-20160818-C01117
    ++++ +++ + ++ Calcd: 399.14 Found: 400.0 [M + H]+
    494
    Figure US20160235758A1-20160818-C01118
    ++++ ++ + +++ Calcd: 391.13 Found: 392.0 [M + H]+
    495
    Figure US20160235758A1-20160818-C01119
    ++++ ++ + +++ Calcd: 377.15 Found: 378.2 [M + H]+
    496
    Figure US20160235758A1-20160818-C01120
    +++ ++ + ++ Calcd: 363.13 Found: 364.2 [M + H]+
    497
    Figure US20160235758A1-20160818-C01121
    + + + + Calcd: 393.13 Found: 394.0 [M + H]+
    498
    Figure US20160235758A1-20160818-C01122
    +++ + + ++ Calcd: 438.16 Found: 439.0 [M + H]+
    499
    Figure US20160235758A1-20160818-C01123
    +++ + + ++ Calcd: 405.14 Found: 406.0 [M + H]+
    500
    Figure US20160235758A1-20160818-C01124
    +++ + ++ ++ Calcd: 452.18 Found: 452.2 [M + H]+
    501
    Figure US20160235758A1-20160818-C01125
    +++ + + ++ Calcd: 397.14 Found: 398.0 [M + H]+
    502
    Figure US20160235758A1-20160818-C01126
    ++++ + ++ +++ Calcd: 438.16 Found: 439.2 [M + H]+
    503
    Figure US20160235758A1-20160818-C01127
    +++ + + ++++ Calcd: 379.14 Found: 380.2 [M + H]+
    504
    Figure US20160235758A1-20160818-C01128
    +++ ++ + ++++ Calcd: 389.15 Found: 390.2
    505
    Figure US20160235758A1-20160818-C01129
    + + + + Calcd: 455.14 Found: 456.0 [M + H]+
    506
    Figure US20160235758A1-20160818-C01130
    ++++ + + + Calcd: 348.13 Found: 349.2 [M + H]+
    507
    Figure US20160235758A1-20160818-C01131
    + + + + Calcd: 377.15 Found: 378.2 [M + H]+
    508
    Figure US20160235758A1-20160818-C01132
    ++++ ++ +++ ++ +++ Calcd: 445.22 Found: 446.2 [M + H]+
    509
    Figure US20160235758A1-20160818-C01133
    + + + + Calcd: 439.16 Found: 440.0 [M + H]+
    510
    Figure US20160235758A1-20160818-C01134
    ++++ ++ +++ ++ ++ Calcd: 431.21 Found: 432.2 [M + H]+
    511
    Figure US20160235758A1-20160818-C01135
    ++++ ++ +++ ++ ++ Calcd: 431.21 Found: 432.2 [M + H]+
    512
    Figure US20160235758A1-20160818-C01136
    ++++ ++ +++ ++ +++ Calcd: 445.22 Found: 446.2 [M + H]+
    513
    Figure US20160235758A1-20160818-C01137
    +++ + + ++ Calcd: 391.16 Found: 392.2 [M + H]+
    514
    Figure US20160235758A1-20160818-C01138
    +++ + + + Calcd: 391.16 Found: 392.2 [M + H]+
    515
    Figure US20160235758A1-20160818-C01139
    ++++ ++ ++++ +++ +++ Calcd: 445.22 Found: 446.2 [M + H]+
    516
    Figure US20160235758A1-20160818-C01140
    +++ + + + Calcd: 382.09 Found: 383.2 [M + H]+
    517
    Figure US20160235758A1-20160818-C01141
    +++ + ++ ++ Calcd: 378.14 Found: 379.2 [M + H]+
    518
    Figure US20160235758A1-20160818-C01142
    +++ + + + Calcd: 376.16 Found: 377.2 [M + H]+
    519
    Figure US20160235758A1-20160818-C01143
    +++ ++ ++ +++ Calcd: 539.18 Found: 540.2 [M + H]+
    520
    Figure US20160235758A1-20160818-C01144
    + + + + Calcd: 441.18 Found: 442.2 [M + H]+
    521
    Figure US20160235758A1-20160818-C01145
    ++++ ++++ ++++ +++ Calcd: 537.16 Found: 538.0 [M + H]+
    522
    Figure US20160235758A1-20160818-C01146
    ++++ ++++ ++++ ++++ +++ Calcd: 533.21 Found: 534.2 [M + H]+
    523
    Figure US20160235758A1-20160818-C01147
    ++++ +++ ++++ ++++ ++ Calcd: 475.14 Found: 476.2 [M + H]+
    524
    Figure US20160235758A1-20160818-C01148
    ++++ ++++ ++++ +++ +++ Calcd: 505.18 Found: 506.2 [M + H]+
    525
    Figure US20160235758A1-20160818-C01149
    ++++ +++ ++++ ++ Calcd: 443.16 Found: 444.2 [M + H]+
    526
    Figure US20160235758A1-20160818-C01150
    ++++ ++ +++ + ++ Calcd: 447.11 Found: 448.0 [M + H]+
    527
    Figure US20160235758A1-20160818-C01151
    ++++ ++++ ++++ +++ Calcd: 509.13 Found: 510.2 [M + H]+
    528
    Figure US20160235758A1-20160818-C01152
    +++ ++ + + Calcd: 347.14 Found: 348.2 [M + H]+
    529
    Figure US20160235758A1-20160818-C01153
    +++ + + ++ Calcd: 371.18 Found: 372.2 [M + H]+
    530
    Figure US20160235758A1-20160818-C01154
    ++++ + ++ +
    531
    Figure US20160235758A1-20160818-C01155
    ++++ ++ + ++++
    532
    Figure US20160235758A1-20160818-C01156
    + ++ + + +
    533
    Figure US20160235758A1-20160818-C01157
    + ++++ +++ +++ +++
    534
    Figure US20160235758A1-20160818-C01158
    ++ + + +
    535
    Figure US20160235758A1-20160818-C01159
    ++ + + +
    536
    Figure US20160235758A1-20160818-C01160
    ++ + + +
    537
    Figure US20160235758A1-20160818-C01161
    ++ + + ++
    538
    Figure US20160235758A1-20160818-C01162
    + ++++ + ++ +
    539
    Figure US20160235758A1-20160818-C01163
    + + + +
    540
    Figure US20160235758A1-20160818-C01164
    + + + + Calcd: 389.15 Found: 390.2 [M + H]+
    541
    Figure US20160235758A1-20160818-C01165
    + + + Calcd: 363.13 Found: 364.2 [M + H]+
    542
    Figure US20160235758A1-20160818-C01166
    + + + + Calcd: 349.17 Found: 350.20 [M + H]+
    543
    Figure US20160235758A1-20160818-C01167
    + ++ + + Calcd: 342.12 Found: 343.2 [M + H]+
    544
    Figure US20160235758A1-20160818-C01168
    +++ + + + Calcd: 362.15 Found: 363.20 [M + H]+
    545
    Figure US20160235758A1-20160818-C01169
    + + + + Calcd: 377.20 Found: 378.20 [M + H]+
    546
    Figure US20160235758A1-20160818-C01170
    + + + + Calcd: 363.18 Found: 364.2 [M + H]+
    547
    Figure US20160235758A1-20160818-C01171
    + + + + Calcd: 363.18 Found: 364.2 [M + H]+
    548
    Figure US20160235758A1-20160818-C01172
    + + + + Calcd: 391.2 Found: 392.2 [M + H]+
    549
    Figure US20160235758A1-20160818-C01173
    + + + + Calcd: 377.2 Found: 378.2 [M + H]+
    550
    Figure US20160235758A1-20160818-C01174
    +++ ++ ++ +++ Calcd: 381.1 Found: 382.2 [M + H]+
    551
    Figure US20160235758A1-20160818-C01175
    ++++ +++ ++ ++++ Calcd: 395.1 Found: 396.0 [M + H]+
    *Starting with compound 438, proliferation data was obtained using an MDA-MB-361 cell line.
    Figure US20160235758A1-20160818-C01176

    Table 4 shows additional exemplary PI3K a inhibitors of the invention.
  • TABLE 4
    In Vitro IC50 data for selected compounds of the invention. The following symbols are used: +
    (greater than 10 microMolar), ++ (less than 10 microMolar), +++ (less than 1 microMolar), and ++++ (less
    than 100 nM).
    MDA-
    mTO MB-361 Mass
    RC PI3K α PI3K β PI3K δ PI3K γ prolifera- Char-
    IC50 IC50 IC50 IC50 IC50 tion acteri-
    Structure (nM) (nM) (nM) (nM) (nM) (nM)* zation
    552
    Figure US20160235758A1-20160818-C01177
    +++ ++++ ++ ++ ++ Calcd: 361.17 Found: 362.2 [M + H]+
    553
    Figure US20160235758A1-20160818-C01178
    ++++ +++ + ++++ +++ Calcd: 348.13 Found: 349.2 [M + H]+
    554
    Figure US20160235758A1-20160818-C01179
    +++ ++ + +++ Calcd: 340.11 Found: 341.0 [M + H]+
    555
    Figure US20160235758A1-20160818-C01180
    +++ +++ ++ ++ ++ Calcd: 361.17 Found: 362.2 [M + H]+
    556
    Figure US20160235758A1-20160818-C01181
    ++ ++++ + ++ ++ ++ Calcd: 403.18 Found: 404.2 [M + H]+
    557
    Figure US20160235758A1-20160818-C01182
    ++++ +++ +++ +++ +++ Calcd: 419.17 Found: 420.2 [M + H]+
    558
    Figure US20160235758A1-20160818-C01183
    + + + + Calcd: 348.13 Found: 349.2 [M + H]+
    559
    Figure US20160235758A1-20160818-C01184
    ++++ + ++ +++ ++ Calcd: 480.17 Found: 481.0 [M + H]+
    560
    Figure US20160235758A1-20160818-C01185
    ++++ ++ ++ ++++ +++ Calcd: 416.16 Found: 417.2 [M + H]+
    561
    Figure US20160235758A1-20160818-C01186
    ++ ++++ + ++ ++ ++ Calcd: 416.21 Found: 417.2 [M + H]+
    562
    Figure US20160235758A1-20160818-C01187
    ++++ ++ + ++++ Calcd: 345.12 Found: 346.0 [M + H]+
    563
    Figure US20160235758A1-20160818-C01188
    + ++++ + ++ + ++ Calcd: 416.21 Found: 417.0 [M + H]+
    564
    Figure US20160235758A1-20160818-C01189
    ++++ + + ++++ ++ Calcd: 382.14 Found: 383.2 [M + H]+
    565
    Figure US20160235758A1-20160818-C01190
    ++++ ++ ++ ++++ +++ Calcd: 362.15 Found: 363.2 [M + H]+
    566
    Figure US20160235758A1-20160818-C01191
    ++++ ++ + ++++ +++ Calcd: 362.15 Found: 363.0 [M + H]+
    567
    Figure US20160235758A1-20160818-C01192
    ++++ ++ ++ ++++ + Calcd: 339.11 Found: 340.0 [M + H]+
    568
    Figure US20160235758A1-20160818-C01193
    +++ + + ++ Calcd: 431.21 Found: 432.2 [M + H]+
    569
    Figure US20160235758A1-20160818-C01194
    +++ + + ++ Calcd: 417.19 Found: 418.0 [M + H]+
    570
    Figure US20160235758A1-20160818-C01195
    + ++++ + + ++++ +++ Calcd: 374.15 Found: 375.0 [M + H]+
    571
    Figure US20160235758A1-20160818-C01196
    +++ + + ++ Calcd: 429.19 Found: 430.2 [M + H]+
    572
    Figure US20160235758A1-20160818-C01197
    ++++ + ++ + ++ Calcd: 430.22 Found: 431.0 [M + H]+
    573
    Figure US20160235758A1-20160818-C01198
    ++++ +++ + ++++ ++ Calcd: 347.14 Found: 348.0 [M + H]+
    574
    Figure US20160235758A1-20160818-C01199
    ++++ ++ + ++++ Calcd: 373.15 Found: 374.2 [M + H]+
    575
    Figure US20160235758A1-20160818-C01200
    ++++ ++++ ++ +++ ++ Calcd: 360.17 Found: 361.2 [M + H]+
    576
    Figure US20160235758A1-20160818-C01201
    ++++ + ++ ++ ++ Calcd: 430.22 Found: 431.2 [M + H]+
    577
    Figure US20160235758A1-20160818-C01202
    +++ + ++ +++ Calcd: 334.15 Found: 335.2 [M + H]+
    578
    Figure US20160235758A1-20160818-C01203
    ++++ ++++ ++ +++ ++ Calcd: 361.17 Found: 362.2 [M + H]+
    579
    Figure US20160235758A1-20160818-C01204
    ++++ + + +++ +++ Calcd: 374.15 Found: 375.2 [M + H]+
    580
    Figure US20160235758A1-20160818-C01205
    ++++ +++ ++ ++++ +++ Calcd: 361.13 Found: 362.0 [M + H]+
    581
    Figure US20160235758A1-20160818-C01206
    ++++ ++++ ++++ ++++ Calcd: 418.19 Found: 419.2 [M + H]+
    582
    Figure US20160235758A1-20160818-C01207
    ++++ ++ ++ +++ ++ Calcd: 418.91 Found: 419.2 [M + H]+
    583
    Figure US20160235758A1-20160818-C01208
    ++++ +++ ++ +++ ++ Calcd: 389.45 Found: 390.2 [M + H]+
    584
    Figure US20160235758A1-20160818-C01209
    ++++ ++ ++ ++ + Calcd: 404.43 Found: 405.2 [M + H]+
    585
    Figure US20160235758A1-20160818-C01210
    +++ + + + + Calcd: 402.19 Found: 403.0 [M + H]+
    586
    Figure US20160235758A1-20160818-C01211
    + ++++ ++ ++ ++ ++ Calcd: 456.24 Found: 457.2 [M + H]+
    587
    Figure US20160235758A1-20160818-C01212
    ++++ ++ + ++++ +++ Calcd: 418.18 Found: 419.0 [M + H]+
    588
    Figure US20160235758A1-20160818-C01213
    + ++++ +++ +++ +++ ++ Calcd: 403.18 Found: 404.2 [M + H]+
    589
    Figure US20160235758A1-20160818-C01214
    ++++ +++ +++ +++ ++ Calcd: 417.19 Found: 418.0 [M + H]+
    590
    Figure US20160235758A1-20160818-C01215
    ++++ ++++ +++ +++ ++ Calcd: 417.19 Found: 418.2 [M + H]+
    591
    Figure US20160235758A1-20160818-C01216
    + ++++ ++ ++ ++ ++ Calcd: 432.20 Found: 433.2 [M + H]+
    592
    Figure US20160235758A1-20160818-C01217
    ++ ++++ ++ ++ ++++ ++ Calcd: 430.19 Found: 431.0 [M + H]+
    593
    Figure US20160235758A1-20160818-C01218
    ++++ +++ +++ ++++ +++ Calcd: 429.19 Found: 430.0 [M + H]+
    594
    Figure US20160235758A1-20160818-C01219
    + ++++ ++ +++ +++ +++ Calcd: 431.21 Found: 432.2 [M + H]+
    595
    Figure US20160235758A1-20160818-C01220
    + ++++ +++ +++ +++ ++ Calcd: 388.20 Found: 389.0 [M + H]+
    596
    Figure US20160235758A1-20160818-C01221
    ++++ ++ + ++++ +++ Calcd: 362.15 Found: 363.0 [M + H]+
    597
    Figure US20160235758A1-20160818-C01222
    + ++++ ++ ++ ++ ++ Calcd: 446.22 Found: 447.2 [M + H]+
    598
    Figure US20160235758A1-20160818-C01223
    + ++++ ++++ ++ +++ ++ Calcd: 360.17 Found: 361.2 [M + H]+
    599
    Figure US20160235758A1-20160818-C01224
    + ++++ +++ ++ ++ ++ Calcd: 360.17 Found: 361.2 [M + H]+
    600
    Figure US20160235758A1-20160818-C01225
    ++ + + ++ Calcd: 417.19 Found: 418.2 [M + H]+
    601
    Figure US20160235758A1-20160818-C01226
    + ++++ ++ ++ ++ ++ Calcd: 398.16 Found: 399.2 [M + H]+
    602
    Figure US20160235758A1-20160818-C01227
    ++ + + + Calcd: 429.19 Found: 430.2 [M + H]+
    603
    Figure US20160235758A1-20160818-C01228
    + ++++ ++ +++ ++ +++ Calcd: 430.22 Found: 431.2 [M + H]+
    604
    Figure US20160235758A1-20160818-C01229
    + ++++ ++ ++ +++ ++ Calcd: 402.18 Found: 403.2 [M + H]+
    605
    Figure US20160235758A1-20160818-C01230
    + ++++ ++ +++ ++ +++ Calcd: 415.21 Found: 416.2 [M + H]+
    606
    Figure US20160235758A1-20160818-C01231
    + ++++ ++ +++ ++ +++ Calcd: 429.23 Found: 430.2 [M + H]+
    607
    Figure US20160235758A1-20160818-C01232
    + ++++ ++ +++ +++ +++ Calcd: 415.21 Found: 416.2 [M + H]+
    608
    Figure US20160235758A1-20160818-C01233
    + ++++ ++ +++ ++ +++ Calcd: 429.23 Found: 430.2 [M + H]+
    609
    Figure US20160235758A1-20160818-C01234
    + ++++ ++ +++ +++ +++ Calcd: 429.23 Found: 430.0 [M + H]+
    610
    Figure US20160235758A1-20160818-C01235
    ++++ + + ++++ +++ Calcd: 373.15 Found: 374.0 [M + H]+
    611
    Figure US20160235758A1-20160818-C01236
    + ++++ ++ +++ ++ +++ Calcd: 455.24 Found: 456.2 [M + H]+
    612
    Figure US20160235758A1-20160818-C01237
    + ++++ + ++ ++ + Calcd: 417.19 Found: 418.0 [M + H]+
    613
    Figure US20160235758A1-20160818-C01238
    ++++ ++ ++ +++ ++ Calcd: 361.15 Found: 362.0 [M + H]+
    614
    Figure US20160235758A1-20160818-C01239
    + ++++ ++ +++ ++++ ++ Calcd: 397.17 Found: 398.2 [M + H]+
    615
    Figure US20160235758A1-20160818-C01240
    ++ ++++ ++ +++ ++++ +++ Calcd: 429.19; Found: 430.2 [M + H]+
    616
    Figure US20160235758A1-20160818-C01241
    + ++++ ++ +++ +++ +++ Calcd: 445.22 Found: 446.2 [M + H]+
    617
    Figure US20160235758A1-20160818-C01242
    ++++ ++ +++ ++++ ++ Calcd: 416.20 Found: 417.2 [M + H]+
    618
    Figure US20160235758A1-20160818-C01243
    ++++ + ++ +++ ++ Calcd: 428.49 Found: 429.2 [M + H]+
    619
    Figure US20160235758A1-20160818-C01244
    ++++ + + ++ ++ Calcd: 416.20 Found: 417.2 [M + H]+
    620
    Figure US20160235758A1-20160818-C01245
    ++++ ++ ++ ++ ++ Calcd: 442.22 Found: 443.2 [M + H]+
    621
    Figure US20160235758A1-20160818-C01246
    ++++ ++ ++ +++ ++ Calcd: 441.23 Found: 442.2 [M + H]+
    622
    Figure US20160235758A1-20160818-C01247
    + ++++ ++ +++ + ++ Calcd: 416.21 Found: 417.2 [M + H]+
    623
    Figure US20160235758A1-20160818-C01248
    + ++++ +++ +++ ++ +++ Calcd: 443.24 Found: 444.2 [M + H]+
    624
    Figure US20160235758A1-20160818-C01249
    ++ + + ++ Calcd: 347.14 Found: 348.2 [M + H]+
    625
    Figure US20160235758A1-20160818-C01250
    + ++++ +++ +++ +++ +++ Calcd: 415.21 Found: 416.2 [M + H]+
    626
    Figure US20160235758A1-20160818-C01251
    + ++++ +++ +++ ++ +++ Calcd: 429.23 Found: 430.2 [M + H]+
    627
    Figure US20160235758A1-20160818-C01252
    ++ ++++ ++ ++ ++ Calcd: 430.22 Found: 431.2 [M + H]+
    628
    Figure US20160235758A1-20160818-C01253
    ++ + + + Calcd: 444.24 Found: 445.2 [M + H]+
    629
    Figure US20160235758A1-20160818-C01254
    + ++++ +++ +++ ++ ++ Calcd: 387.18 Found: 388.2 [M + H]+
    630
    Figure US20160235758A1-20160818-C01255
    + ++++ +++ +++ +++ ++ Calcd: 386.19 Found: 387.2 [M + H]+
    631
    Figure US20160235758A1-20160818-C01256
    ++ ++ + + Calcd: 360.17 Found: 361.2 [M + H]+
    632
    Figure US20160235758A1-20160818-C01257
    ++ + + ++ Calcd: 373.15 Found: 374.2 [M + H]+
    633
    Figure US20160235758A1-20160818-C01258
    ++ + + ++ Calcd: 431.21 Found: 432.2 [M + H]+
  • In some embodiments, the invention provides a combination treatment comprising an mTor inhibitor, which can be a compound as provided herein and a PI3Kα inhibitor also as provided herein. In some embodiments, the mTor inhibitor is a compound of Formula I, Formula I-A, Formula I-B1, Formula I-C, Formula I-C1a, or a compound of Table 1 or Table 2, and the PI3Kα inhibitor is a compound of Formula II, Subformula IIa, Subformula IIb, Formula III, Subclass Inc, Subclass IIId, Formula VI-B, Formula VI-C, Formula VI-D, or Table 3. For example, the mTor inhibitor is a compound of Formula I where M1 is a bicyclic heteroaryl system, including, for instance, benzothiazolyl, quinolinyl, quinazolinyl, benzoxazolyl, and benzoimidazolyl, and the PI3Kα inhibitor is a compound of Formula II, Subformula IIa, Subformula II, Formula III, Subclass Inc, Subclass IIId, Formula VI-B, Formula VI-C, Formula VI-D, or Table 3. In other embodiments, the mTor inhibitor is a compound of Formula I where M1 is of formula M1-A, M1-B, M1-C or M1-D, and the PI3Kα inhibitor is a compound of Formula II, Subformula IIa, Subformula IIb, Formula III, Subclass Inc, Subclass IIId, Formula VI-B, Formula VI-C, Formula VI-D, or Table 3. In yet other embodiments, the mTor inhibitor is of Formula I-B1 and M1 is of formula M1-F1, and the PI3Kα inhibitor is a compound of Formula II, Subformula IIa, Subformula IIb, Formula III, Subclass IIIc, Subclass IIId, Formula VI-B, Formula VI-C, Formula VI-D, or Table 3. In still other embodiments, the mTor inhibitor is of Formula I-C, and the PI3Kα inhibitor is a compound of Formula II, Subformula IIa, Subformula IIb, Formula III, Subclass IIIc, Subclass IIId, Formula VI-B, Formula VI-C, Formula VI-D, or Table 3. In still other embodiments, the mTor inhibitor is of Formula I-C1a, and the PI3Kα inhibitor is a compound of Formula II, Subformula IIa, Subformula IIb, Formula III, Subclass IIIc, Subclass IIId, Formula VI-B, Formula VI-C, Formula VI-D, or Table 3.
  • In some embodiments, the mTor inhibitor is a compound of Formula I, Formula I-A, Formula I-B1, Formula I-C, Formula I-C1a, or a compound of Table 1 or Table 2, and the PI3Kα inhibitor is a compound of Formula II or Formula III. In other embodiments, the mTor inhibitor is a compound of Formula I, Formula I-A, Formula I-B1, Formula I-C, Formula I-C1a, or a compound of Table 1 or Table 2, and the PI3Kα inhibitor is a compound of Subformula Ha or Subformula IIb. In still other embodiments, the mTor inhibitor is a compound of Formula I, Formula I-A, Formula I-B1, Formula I-C, Formula I-C1a, or a compound of Table 1 or Table 2, and the PI3Kα inhibitor is a compound of Formula III, Subclass IIIc or Subclass IIId. In still other embodiments, the mTor inhibitor is a compound of Formula I, Formula I-A, Formula I-B1, Formula I-C, Formula I-C1a, or a compound of Table 1 or Table 2, and the PI3Kα inhibitor is a compound of Formula VI-B, VI-C, VI-D or of Table 3.
  • In some embodiments, the mTor inhibitor is a compound of Formula I where M1 is of formula M1-A, M1-B, M1-C or M1-D, and the PI3Kα inhibitor is a compound of Subclass Mc or Subclass IIId. In other embodiments, the mTor inhibitor is a compound of Formula I-C and the PI3Kα inhibitor is a compound of Subclass Mc or Subclass IIId.
  • In some embodiments, the mTor inhibitor is a compound of Table 1 or 2 and the PI3Kα inhibitor is a compound of Table 3.
  • Pharmaceutical Compositions and Administration
  • The invention provides, in one aspect, a combination treatment utilizing a PI3Kα inhibitor and an mTor inhibitor. The therapeutic agents (including compounds) that are provided for use in the combination therapies of the invention can be administered simultaneously or separately. This administration in combination includes, for example, simultaneous administration of two agents in the same dosage form, simultaneous administration in separate dosage forms, and separate administration. For example, multiple therapeutic agents can be formulated together in the same dosage form and administered simultaneously. Alternatively multiple therapeutic agents can be simultaneously administered, wherein both the agents are present in separate formulations. In another alternative, an inhibitor of the present invention can be administered just followed by and any of the agents described above, or vice versa. In the separate administration protocol, an inhibitor of the present invention and any of the agents described above may be administered a few minutes apart, or a few hours apart, or a few days apart. The term “combination treatments” also embraces the administration of the therapeutic agents as described herein in further combination with other biologically active compounds or ingredients and non-drug therapies (e.g., surgery or radiation treatment).
  • Administration of the compounds of the present invention can be effected by any method that enables delivery of the compounds to the site of action. An effective amount of an inhibitor of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, as an inhalant, or via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer. Sequential or substantially simultaneous administration of each inhibitor or therapeutic agent can be effected by any appropriate route as noted above and including, but not limited to, oral routes, intravenous routes, intramuscular routes, and direct absorption through mucous membrane tissues. The therapeutic agents can be administered by the same route or by different routes. For example, a first therapeutic agent of the combination selected may be administered by intravenous injection while the other therapeutic agents of the combination may be administered orally. Alternatively, for example, all therapeutic agents may be administered orally or all therapeutic agents may be administered by intravenous injection. The sequence in which the therapeutic agents are administered is not narrowly critical.
  • In some embodiments, administration of the inhibitors of the invention can be effected in one dose, continuously or intermittently throughout the course of treatment. Methods of determining the most effective means and dosage of administration are well known to those of skill in the art and will vary with the composition used for therapy, the purpose of the therapy, the target cell or tissue being treated, and the subject being treated. Single or multiple administrations can be carried out with the dose level and pattern being selected by the treating physician.
  • The amount of each inhibitor or compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. However, an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g., by dividing such larger doses into several small doses for administration throughout the day.
  • In some embodiments, a combination treatment of the invention is administered in a single dose comprising at least a PI3Kα inhibitor and an mTor inhibitor. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes may be used as appropriate. A single dose of a combination treatment of the invention may also be used for treatment of an acute condition.
  • In some embodiments, a combination treatment of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment a PI3Kα inhibitor and an mTor inhibitor are administered together about once per day to about 6 times per day. In another embodiment the administration of a a PI3Kα inhibitor and an mTor inhibitor continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • Administration of the combination treatments of the invention may continue as long as necessary. In some embodiments, an agent of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, an agent of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, an agent of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • When a combination treatment of the invention is administered as a composition that comprises one or more compounds, and one compound has a shorter half-life than another compound, the unit dose forms may be adjusted accordingly.
  • In some embodiments, combination treatments of the invention are tested to estimate pharmacokinetic properties and expected side effect profile. Various assays are known in the art for this purpose. For example, oral availability can be estimated during early stages of drug development by performing a Caco-2 permeability assay. Further, oral pharmacokinetics in humans can be approximated by extrapolating from the results of assays in mice, rats or monkey. In some embodiments, compounds of the invention show good oral availability across multiple species of organisms.
  • Other assays examine the effect of an inhibitor on liver function and metabolism. Cytochrome P450 (CYP) proteins are the main enzyme involved in metabolizing drugs administered to mammalian organisms. As such, undesired interaction of a drug candidate can be a significant source of adverse drug interactions. Generally, it is desirable for a drug to not interact with CYP isozymes such as CYP1A2, CYP2C9, CYP2C19, CYP2D6, or CYP3A4. In some embodiments, an inhibitor of the invention exhibits an IC50 of greater than 10 μM for CYP1A2, CYP2C9, CYP2C19, CYP2D6, or CYP3A4. Additionally, liver microsome and hepatocyte metabolism assays using human preparations can be used to estimate the in-vitro half life of a drug candidate.
  • Cardiac toxicity is also an important consideration in evaluating compounds. For example, hERG is the gene coding for the Kv11.1 potassium ion channel, a protein is involved in mediating repolarizing current in the cardiac action potential in the heart Inhibition of the hERG gene product by a drug candidate can lead to an increase in the risk of sudden death and is therefore an undesirable property. In some embodiments, an inhibitor of the invention exhibits less than 10% hERG inhibition when administered at a suitable concentration.
  • Mutagenicity of compounds can be assayed via an Ames test or a modified Ames test using e.g., the liver S9 system. In some embodiments, compounds show negative activity in such a test.
  • Other undesired interactions of an inhibitor can also be ascertained via a receptor panel screen. In some embodiments, no significant interactions are detected for combination treatments of the invention. The subject pharmaceutical compositions can be formulated to provide a therapeutically effective amount of a combination of therapeutic agents of the present invention, or pharmaceutically acceptable salts, esters, prodrugs, solvates, hydrates or derivatives thereof. Where desired, the pharmaceutical compositions contain pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including inert solid diluents and fillers, diluents, including sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • The subject pharmaceutical compositions can be administered as a combination of a PI3Kα inhibitor and an mTor inhibitor, or in further combination with one or more other agents, which are also typically administered in the form of pharmaceutical compositions. Where desired, the subject combinations and other agent(s) may be mixed into a preparation or both components may be formulated into separate preparations to use them in combination separately or at the same time.
  • In some embodiments, the concentration of one or more of the compounds provided in the pharmaceutical compositions of the present invention is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%,14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v or v/v.
  • In some embodiments, the concentration of one or more of the compounds of the present invention is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25% 13%, 12.75%, 12.50%, 12.25% 12%, 11.75%, 11.50%, 11.25% 11%, 10.75%, 10.50%, 10.25% 10%, 9.75%, 9.50%, 9.25% 9%, 8.75%, 8.50%, 8.25% 8%, 7.75%, 7.50%, 7.25% 7%, 6.75%, 6.50%, 6.25% 6%, 5.75%, 5.50%, 5.25% 5%, 4.75%, 4.50%, 4.25%, 4%, 3.75%, 3.50%, 3.25%, 3%, 2.75%, 2.50%, 2.25%, 2%, 1.75%, 1.50%, 125%, 1%, 0.5%, 0.4%, 0.3%, 0.2%, 0.1%, 0.09%, 0.08%, 0.07%, 0.06%, 0.05%, 0.04%, 0.03%, 0.02%, 0.01%, 0.009%, 0.008%, 0.007%, 0.006%, 0.005%, 0.004%, 0.003%, 0.002%, 0.001%, 0.0009%, 0.0008%, 0.0007%, 0.0006%, 0.0005%, 0.0004%, 0.0003%, 0.0002%, or 0.0001% w/w, w/v, or v/v.
  • In some embodiments, the concentration of one or more of the compounds of the present invention is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40%, approximately 0.01% to approximately 30%, approximately 0.02% to approximately 29%, approximately 0.03% to approximately 28%, approximately 0.04% to approximately 27%, approximately 0.05% to approximately 26%, approximately 0.06% to approximately 25%, approximately 0.07% to approximately 24%, approximately 0.08% to approximately 23%, approximately 0.09% to approximately 22%, approximately 0.1% to approximately 21%, approximately 0.2% to approximately 20%, approximately 0.3% to approximately 19%, approximately 0.4% to approximately 18%, approximately 0.5% to approximately 17%, approximately 0.6% to approximately 16%, approximately 0.7% to approximately 15%, approximately 0.8% to approximately 14%, approximately 0.9% to approximately 12%, approximately 1% to approximately 10% w/w, w/v or v/v. v/v.
  • In some embodiments, the concentration of one or more of the compounds of the present invention is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v.
  • In some embodiments, the amount of one or more of the compounds of the present invention is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g, 0.01 g, 0.009 g, 0.008 g, 0.007 g, 0.006 g, 0.005 g, 0.004 g, 0.003 g, 0.002 g, 0.001 g, 0.0009 g, 0.0008 g, 0.0007 g, 0.0006 g, 0.0005 g, 0.0004 g, 0.0003 g, 0.0002 g, or 0.0001 g.
  • In some embodiments, the amount of one or more of the compounds of the present invention is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g, 0.065 g, 0.07 g, 0.075 g, 0.08 g, 0.085 g, 0.09 g, 0.095 g, 0.1 g, 0.15 g, 0.2 g, 0.25 g, 0.3 g, 0.35 g, 0.4 g, 0.45 g, 0.5 g, 0.55 g, 0.6 g, 0.65 g, 0.7 g, 0.75 g, 0.8 g, 0.85 g, 0.9 g, 0.95 g, 1 g, 1.5 g, 2 g, 2.5, 3 g, 3.5, 4 g, 4.5 g, 5 g, 5.5 g, 6 g, 6.5 g, 7 g, 7.5 g, 8 g, 8.5 g, 9 g, 9.5 g, or 10 g.
  • In some embodiments, the amount of one or more of the compounds of the present invention is in the range of 0.0001-10 g, 0.0005-9 g, 0.001-8 g, 0.005-7 g, 0.01-6 g, 0.05-5 g, 0.1-4 g, 0.5-4 g, or 1-3 g.
  • The combination treatments according to the invention are effective over a wide dosage range. For example, in the treatment of adult humans, dosages from 0.01 to 1000 mg, from 0.5 to 100 mg, from 1 to 50 mg per day, and from 5 to 40 mg per day are examples of dosages that may be used. An exemplary dosage is 10 to 30 mg per day. The exact dosage will depend upon the route of administration, the form in which the compound is administered, the subject to be treated, the body weight of the subject to be treated, and the preference and experience of the attending physician.
  • A pharmaceutical composition of the present invention typically contains an active ingredient (e.g., an inhibitor of the present invention or a pharmaceutically acceptable salt and/or coordination complex thereof, and one or more pharmaceutically acceptable excipients, carriers, including but not limited inert solid diluents and fillers, diluents, sterile aqueous solution and various organic solvents, permeation enhancers, solubilizers and adjuvants.
  • Described below are non-limiting exemplary pharmaceutical compositions and methods for preparing the same.
  • Pharmaceutical Compositions for Oral Administration
  • In some embodiments, the invention provides a pharmaceutical composition for oral administration containing at least one therapeutic agent, and a pharmaceutical excipient suitable for oral administration.
  • In some embodiments, the invention provides a solid pharmaceutical composition for oral administration containing: (i) a compound which is a PI3Kα inhibitor; (ii) a second compound which is an mTor inhibitor; and (iii) a pharmaceutical excipient suitable for oral administration. In some embodiments, the composition further contains: (iv) a third agent or even a fourth agent. In some embodiments, each compound or agent is present in a therapeutically effective amount. In other embodiments, one or more compounds or agents is present in a sub-therapeutic amount, and the compounds or agents act synergystically to provide a therapeutically effective pharmaceutical composition.
  • In some embodiments, the invention provides for a pharmaceutical composition comprising a combination of a PI3-kinase α inhibitor and an mTOR inhibitor. The PI3-kinase α inhibitor and the mTOR inhibitor can be packaged as a single oral dosage form. In other embodiments, the PI3-kinase α inhibitor and the mTOR inhibitor can be packaged as separate dosage forms, such as a tablet.
  • In one embodiment, the present invention provides an oral dosage form comprising 100 mg to 1.5 g of an inhibitor of the invention. The oral dosage form can be a tablet, formulated in form of liquid, in immediate or sustained release format.
  • In some embodiments, the pharmaceutical composition may be a liquid pharmaceutical composition suitable for oral consumption. Pharmaceutical compositions of the invention suitable for oral administration can be presented as discrete dosage forms, such as capsules, cachets, or tablets, or liquids or aerosol sprays each containing a predetermined amount of an active ingredient as a powder or in granules, a solution, or a suspension in an aqueous or non-aqueous liquid, an oil-in-water emulsion, or a water-in-oil liquid emulsion, including liquid dosage forms (e.g., a suspension or slurry), and oral solid dosage forms (e.g., a tablet or bulk powder). As used herein the term “tablet” refers generally to tablets, caplets, capsules, including soft gelatin capsules, and lozenges. Oral dosage forms may be formulated as tablets, pills, dragees, capsules, emulsions, lipophilic and hydrophilic suspensions, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by an individual or a patient to be treated. Such dosage forms can be prepared by any of the methods of pharmacy, but all methods include the step of bringing the active ingredient into association with the carrier, which constitutes one or more necessary ingredients. In one embodiment, the inhibitor of the invention is contained in capsules. Capsules suitable for oral administration include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. Optionally, the inventive composition for oral use can be obtained by mixing the inhibitor with a solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). In general, the compositions are prepared by uniformly and intimately admixing the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product into the desired presentation. For example, a tablet can be prepared by compression or molding, optionally with one or more accessory ingredients. Compressed tablets can be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as powder or granules, optionally mixed with an excipient such as, but not limited to, a binder, a lubricant, an inert diluent, and/or a surface active or dispersing agent. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising an active ingredient, since water can facilitate the degradation of some compounds. For example, water may be added (e.g., 5%) in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms of the invention which contain lactose can be made anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected. An anhydrous pharmaceutical composition may be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions may be packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastic or the like, unit dose containers, blister packs, and strip packs.
  • An active ingredient can be combined in an intimate admixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier can take a wide variety of forms depending on the form of preparation desired for administration. In preparing the compositions for an oral dosage form, any of the usual pharmaceutical media can be employed as carriers, such as, for example, water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like in the case of oral liquid preparations (such as suspensions, solutions, and elixirs) or aerosols; or carriers such as starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents can be used in the case of oral solid preparations, in some embodiments without employing the use of lactose. For example, suitable carriers include powders, capsules, and tablets, with the solid oral preparations. If desired, tablets can be coated by standard aqueous or nonaqueous techniques.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose), polyvinyl pyrrolidone, methyl cellulose, pre-gelatinized starch, hydroxypropyl methyl cellulose, microcrystalline cellulose, and mixtures thereof.
  • Examples of suitable fillers for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • Disintegrants may be used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Too much of a disintegrant may produce tablets which may disintegrate in the bottle. Too little may be insufficient for disintegration to occur and may thus alter the rate and extent of release of the active ingredient(s) from the dosage form. Thus, a sufficient amount of disintegrant that is neither too little nor too much to detrimentally alter the release of the active ingredient(s) may be used to form the dosage forms of the compounds disclosed herein. The amount of disintegrant used may vary based upon the type of formulation and mode of administration, and may be readily discernible to those of ordinary skill in the art. About 0.5 to about 15 weight percent of disintegrant, or about 1 to about 5 weight percent of disintegrant, may be used in the pharmaceutical composition. Disintegrants that can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, other starches, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums or mixtures thereof.
  • Lubricants which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethylaureate, agar, or mixtures thereof. Additional lubricants include, for example, a syloid silica gel, a coagulated aerosol of synthetic silica, or mixtures thereof. A lubricant can optionally be added, in an amount of less than about 1 weight percent of the pharmaceutical composition.
  • Lubricants can be also be used in conjunction with tissue barriers which include, but are not limited to, polysaccharides, polyglycans, seprafilm, interceed and hyaluronic acid.
  • When aqueous suspensions and/or elixirs are desired for oral administration, the essential active ingredient therein may be combined with various sweetening or flavoring agents, coloring matter or dyes and, if so desired, emulsifying and/or suspending agents, together with such diluents as water, ethanol, propylene glycol, glycerin and various combinations thereof.
  • The tablets can be uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time delay material such as glyceryl monostearate or glyceryl distearate can be employed. Formulations for oral use can also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example, calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example, peanut oil, liquid paraffin or olive oil.
  • Surfactant which can be used to form pharmaceutical compositions and dosage forms of the invention include, but are not limited to, hydrophilic surfactants, lipophilic surfactants, and mixtures thereof. That is, a mixture of hydrophilic surfactants may be employed, a mixture of lipophilic surfactants may be employed, or a mixture of at least one hydrophilic surfactant and at least one lipophilic surfactant may be employed.
  • A suitable hydrophilic surfactant may generally have an HLB value of at least 10, while suitable lipophilic surfactants may generally have an HLB value of or less than about 10. An empirical parameter used to characterize the relative hydrophilicity and hydrophobicity of non-ionic amphiphilic compounds is the hydrophilic-lipophilic balance (“HLB” value). Surfactants with lower HLB values are more lipophilic or hydrophobic, and have greater solubility in oils, while surfactants with higher HLB values are more hydrophilic, and have greater solubility in aqueous solutions. Hydrophilic surfactants are generally considered to be those compounds having an HLB value greater than about 10, as well as anionic, cationic, or zwitterionic compounds for which the HLB scale is not generally applicable. Similarly, lipophilic (i.e., hydrophobic) surfactants are compounds having an HLB value equal to or less than about 10. However, HLB value of a surfactant is merely a rough guide generally used to enable formulation of industrial, pharmaceutical and cosmetic emulsions.
  • Hydrophilic surfactants may be either ionic or non-ionic. Suitable ionic surfactants include, but are not limited to, alkylammonium salts; fusidic acid salts; fatty acid derivatives of amino acids, oligopeptides, and polypeptides; glyceride derivatives of amino acids, oligopeptides, and polypeptides; lecithins and hydrogenated lecithins; lysolecithins and hydrogenated lysolecithins; phospholipids and derivatives thereof; lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • Within the aforementioned group, ionic surfactants include, by way of example: lecithins, lysolecithin, phospholipids, lysophospholipids and derivatives thereof; carnitine fatty acid ester salts; salts of alkylsulfates; fatty acid salts; sodium docusate; acylactylates; mono- and di-acetylated tartaric acid esters of mono- and di-glycerides; succinylated mono- and di-glycerides; citric acid esters of mono- and di-glycerides; and mixtures thereof.
  • Ionic surfactants may be the ionized forms of lecithin, lysolecithin, phosphatidylcholine, phosphatidylethanolamine, phosphatidylglycerol, phosphatidic acid, phosphatidylserine, lysophosphatidylcholine, lysophosphatidylethanolamine, lysophosphatidylglycerol, lysophosphatidic acid, lysophosphatidylserine, PEG-phosphatidylethanolamine, PVP-phosphatidylethanolamine, lactylic esters of fatty acids, stearoyl-2-lactylate, stearoyl lactylate, succinylated monoglycerides, mono/diacetylated tartaric acid esters of mono/diglycerides, citric acid esters of mono/diglycerides, cholylsarcosine, caproate, caprylate, caprate, laurate, myristate, palmitate, oleate, ricinoleate, linoleate, linolenate, stearate, lauryl sulfate, teracecyl sulfate, docusate, lauroyl carnitines, palmitoyl carnitines, myristoyl carnitines, and salts and mixtures thereof.
  • Hydrophilic non-ionic surfactants may include, but not limited to, alkylglucosides; alkylmaltosides; alkylthioglucosides; lauryl macrogolglycerides; polyoxyalkylene alkyl ethers such as polyethylene glycol alkyl ethers; polyoxyalkylene alkylphenols such as polyethylene glycol alkyl phenols; polyoxyalkylene alkyl phenol fatty acid esters such as polyethylene glycol fatty acids monoesters and polyethylene glycol fatty acids diesters; polyethylene glycol glycerol fatty acid esters; polyglycerol fatty acid esters; polyoxyalkylene sorbitan fatty acid esters such as polyethylene glycol sorbitan fatty acid esters; hydrophilic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids, and sterols; polyoxyethylene sterols, derivatives, and analogues thereof; polyoxyethylated vitamins and derivatives thereof; polyoxyethylene-polyoxypropylene block copolymers; and mixtures thereof; polyethylene glycol sorbitan fatty acid esters and hydrophilic transesterification products of a polyol with at least one member of the group consisting of triglycerides, vegetable oils, and hydrogenated vegetable oils. The polyol may be glycerol, ethylene glycol, polyethylene glycol, sorbitol, propylene glycol, pentaerythritol, or a saccharide.
  • Other hydrophilic-non-ionic surfactants include, without limitation, PEG-10 laurate, PEG-12 laurate, PEG-20 laurate, PEG-32 laurate, PEG-32 dilaurate, PEG-12 oleate, PEG-15 oleate, PEG-20 oleate, PEG-20 dioleate, PEG-32 oleate, PEG-200 oleate, PEG-400 oleate, PEG-15 stearate, PEG-32 distearate, PEG-40 stearate, PEG-100 stearate, PEG-20 dilaurate, PEG-25 glyceryl trioleate, PEG-32 dioleate, PEG-20 glyceryl laurate, PEG-30 glyceryl laurate, PEG-20 glyceryl stearate, PEG-20 glyceryl oleate, PEG-30 glyceryl oleate, PEG-30 glyceryl laurate, PEG-40 glyceryl laurate, PEG-40 palm kernel oil, PEG-50 hydrogenated castor oil, PEG-40 castor oil, PEG-35 castor oil, PEG-60 castor oil, PEG-40 hydrogenated castor oil, PEG-60 hydrogenated castor oil, PEG-60 corn oil, PEG-6 caprate/caprylate glycerides, PEG-8 caprate/caprylate glycerides, polyglyceryl-10 laurate, PEG-30 cholesterol, PEG-25 phyto sterol, PEG-30 soya sterol, PEG-20 trioleate, PEG-40 sorbitan oleate, PEG-80 sorbitan laurate, polysorbate 20, polysorbate 80, POE-9 lauryl ether, POE-23 lauryl ether, POE-10 oleyl ether, POE-20 oleyl ether, POE-20 stearyl ether, tocopheryl PEG-100 succinate, PEG-24 cholesterol, polyglyceryl-10 oleate, Tween 40, Tween 60, sucrose monostearate, sucrose monolaurate, sucrose monopalmitate, PEG 10-100 nonyl phenol series, PEG 15-100 octyl phenol series, and poloxamers.
  • Suitable lipophilic surfactants include, by way of example only: fatty alcohols; glycerol fatty acid esters; acetylated glycerol fatty acid esters; lower alcohol fatty acids esters; propylene glycol fatty acid esters; sorbitan fatty acid esters; polyethylene glycol sorbitan fatty acid esters; sterols and sterol derivatives; polyoxyethylated sterols and sterol derivatives; polyethylene glycol alkyl ethers; sugar esters; sugar ethers; lactic acid derivatives of mono- and di-glycerides; hydrophobic transesterification products of a polyol with at least one member of the group consisting of glycerides, vegetable oils, hydrogenated vegetable oils, fatty acids and sterols; oil-soluble vitamins/vitamin derivatives; and mixtures thereof. Within this group, preferred lipophilic surfactants include glycerol fatty acid esters, propylene glycol fatty acid esters, and mixtures thereof, or are hydrophobic transesterification products of a polyol with at least one member of the group consisting of vegetable oils, hydrogenated vegetable oils, and triglycerides.
  • In one embodiment, the composition may include a solubilizer to ensure good solubilization and/or dissolution of the compound of the present invention and to minimize precipitation of the compound of the present invention. This can be especially important for compositions for non-oral use, e.g., compositions for injection. A solubilizer may also be added to increase the solubility of the hydrophilic drug and/or other components, such as surfactants, or to maintain the composition as a stable or homogeneous solution or dispersion.
  • Examples of suitable solubilizers include, but are not limited to, the following: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether (glycofurol) or methoxy PEG; amides and other nitrogen-containing compounds such as 2-pyrrolidone, 2-piperidone, .epsilon.-caprolactam, N-alkylpyrrolidone, N-hydroxyalkylpyrrolidone, N-alkylpiperidone, N-alkylcaprolactam, dimethylacetamide and polyvinylpyrrolidone; esters such as ethyl propionate, tributylcitrate, acetyl triethylcitrate, acetyl tributyl citrate, triethylcitrate, ethyl oleate, ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate, propylene glycol diacetate, ε-caprolactone and isomers thereof, δ-valerolactone and isomers thereof, β-butyrolactone and isomers thereof; and other solubilizers known in the art, such as dimethyl acetamide, dimethyl isosorbide, N-methyl pyrrolidones, monooctanoin, diethylene glycol monoethyl ether, and water.
  • Mixtures of solubilizers may also be used. Examples include, but not limited to, triacetin, triethylcitrate, ethyl oleate, ethyl caprylate, dimethylacetamide, N-methylpyrrolidone, N-hydroxyethylpyrrolidone, polyvinylpyrrolidone, hydroxypropyl methylcellulose, hydroxypropyl cyclodextrins, ethanol, polyethylene glycol 200-100, glycofurol, transcutol, propylene glycol, and dimethyl isosorbide. Particularly preferred solubilizers include sorbitol, glycerol, triacetin, ethyl alcohol, PEG-400, glycofurol and propylene glycol.
  • The amount of solubilizer that can be included is not particularly limited. The amount of a given solubilizer may be limited to a bioacceptable amount, which may be readily determined by one of skill in the art. In some circumstances, it may be advantageous to include amounts of solubilizers far in excess of bioacceptable amounts, for example to maximize the concentration of the drug, with excess solubilizer removed prior to providing the composition to a subject using conventional techniques, such as distillation or evaporation. Thus, if present, the solubilizer can be in a weight ratio of 10%, 25%, 50%, 100%, or up to about 200% by weight, based on the combined weight of the drug, and other excipients. If desired, very small amounts of solubilizer may also be used, such as 5%, 2%, 1% or even less. Typically, the solubilizer may be present in an amount of about 1% to about 100%, more typically about 5% to about 25% by weight.
  • The composition can further include one or more pharmaceutically acceptable additives and excipients. Such additives and excipients include, without limitation, detackifiers, anti-foaming agents, buffering agents, polymers, antioxidants, preservatives, chelating agents, viscomodulators, tonicifiers, flavorants, colorants, odorants, opacifiers, suspending agents, binders, fillers, plasticizers, lubricants, and mixtures thereof.
  • In addition, an acid or a base may be incorporated into the composition to facilitate processing, to enhance stability, or for other reasons. Examples of pharmaceutically acceptable bases include amino acids, amino acid esters, ammonium hydroxide, potassium hydroxide, sodium hydroxide, sodium hydrogen carbonate, aluminum hydroxide, calcium carbonate, magnesium hydroxide, magnesium aluminum silicate, synthetic aluminum silicate, synthetic hydrocalcite, magnesium aluminum hydroxide, diisopropylethylamine, ethanolamine, ethylenediamine, triethanolamine, triethylamine, triisopropanolamine, trimethylamine, tris(hydroxymethyl)aminomethane (TRIS) and the like. Also suitable are bases that are salts of a pharmaceutically acceptable acid, such as acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acid, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid, and the like. Salts of polyprotic acids, such as sodium phosphate, disodium hydrogen phosphate, and sodium dihydrogen phosphate can also be used. When the base is a salt, the cation can be any convenient and pharmaceutically acceptable cation, such as ammonium, alkali metals, alkaline earth metals, and the like. Example may include, but not limited to, sodium, potassium, lithium, magnesium, calcium and ammonium.
  • Suitable acids are pharmaceutically acceptable organic or inorganic acids. Examples of suitable inorganic acids include hydrochloric acid, hydrobromic acid, hydriodic acid, sulfuric acid, nitric acid, boric acid, phosphoric acid, and the like. Examples of suitable organic acids include acetic acid, acrylic acid, adipic acid, alginic acid, alkanesulfonic acids, amino acids, ascorbic acid, benzoic acid, boric acid, butyric acid, carbonic acid, citric acid, fatty acids, formic acid, fumaric acid, gluconic acid, hydroquinosulfonic acid, isoascorbic acid, lactic acid, maleic acid, methanesulfonic acid, oxalic acid, para-bromophenylsulfonic acid, propionic acid, p-toluenesulfonic acid, salicylic acid, stearic acid, succinic acid, tannic acid, tartaric acid, thioglycolic acid, toluenesulfonic acid, uric acid and the like.
  • Pharmaceutical Compositions for Injection.
  • In some embodiments, the invention provides a pharmaceutical composition for injection containing at least one compound of the present invention and a pharmaceutical excipient suitable for injection. For example a pharmaceutical composition for injection is provided comprising at least one PI3Kα inhibitor and an mTor inhibitor. Also provided are pharmaceutical compositions comprising a PI3Kα inhibitor, and pharmaceutical compositions comprising an mTor inhibitor, where the PI3Kα inhibitor is administered separately or together with the mTor inhibitor. The PI3K a inhibitor and the mTor inhibitor may be formulated separately, and may further include a third therapeutic agent. Components and amounts of agents in the compositions are as described herein.
  • The forms in which the novel compositions of the present invention may be incorporated for administration by injection include aqueous or oil suspensions, or emulsions, with sesame oil, corn oil, cottonseed oil, or peanut oil, as well as elixirs, mannitol, dextrose, or a sterile aqueous solution, and similar pharmaceutical vehicles.
  • Aqueous solutions in saline are also conventionally used for injection. Ethanol, glycerol, propylene glycol, liquid polyethylene glycol, and the like (and suitable mixtures thereof), cyclodextrin derivatives, and vegetable oils may also be employed. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, for the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like.
  • Sterile injectable solutions are prepared by incorporating the compound of the present invention in the required amount in the appropriate solvent with various other ingredients as enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, certain desirable methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • Pharmaceutical Compositions for Topical (e.g., Transdermal) Delivery.
  • In some embodiments, the invention provides a pharmaceutical composition for transdermal delivery containing at least one compound of the present invention and a pharmaceutical excipient suitable for transdermal delivery. For example a pharmaceutical composition for topical delivery is provided comprising at least one PI3Kα inhibitor and an mTor inhibitor. Also provided are pharmaceutical compositions for topical delivery comprising a PI3Kα inhibitor, and pharmaceutical compositions for topical delivery comprising an mTor inhibitor, where the PI3Kα inhibitor is administered separately or together with the mTor inhibitor. The PI3K a inhibitor and the mTor inhibitor may be formulated separately, and may further include a third therapeutic agent.
  • Compositions of the present invention can be formulated into preparations in solid, semi-solid, or liquid forms suitable for local or topical administration, such as gels, water soluble jellies, creams, lotions, suspensions, foams, powders, slurries, ointments, solutions, oils, pastes, suppositories, sprays, emulsions, saline solutions, dimethylsulfoxide (DMSO)-based solutions. In general, carriers with higher densities are capable of providing an area with a prolonged exposure to the active ingredients. In contrast, a solution formulation may provide more immediate exposure of the active ingredient to the chosen area.
  • The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients, which are compounds that allow increased penetration of, or assist in the delivery of, therapeutic molecules across the stratum corneum permeability barrier of the skin. There are many of these penetration-enhancing molecules known to those trained in the art of topical formulation. Examples of such carriers and excipients include, but are not limited to, humectants (e.g., urea), glycols (e.g., propylene glycol), alcohols (e.g., ethanol), fatty acids (e.g., oleic acid), surfactants (e.g., isopropyl myristate and sodium lauryl sulfate), pyrrolidones, glycerol monolaurate, sulfoxides, terpenes (e.g., menthol), amines, amides, alkanes, alkanols, water, calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • Another exemplary formulation for use in the methods of the present invention employs transdermal delivery devices (“patches”). Such transdermal patches may be used to provide continuous or discontinuous infusion of an inhibitor of the present invention in controlled amounts, either with or without another agent.
  • The construction and use of transdermal patches for the delivery of pharmaceutical agents is well known in the art. See, e.g., U.S. Pat. Nos. 5,023,252, 4,992,445 and 5,001,139. Such patches may be constructed for continuous, pulsatile, or on demand delivery of pharmaceutical agents.
  • Pharmaceutical Compositions for Inhalation.
  • Compositions for inhalation or insufflation include solutions and suspensions in pharmaceutically acceptable, aqueous or organic solvents, or mixtures thereof, and powders. The liquid or solid compositions may contain suitable pharmaceutically acceptable excipients as described supra. Preferably the compositions are administered by the oral or nasal respiratory route for local or systemic effect. Compositions in preferably pharmaceutically acceptable solvents may be nebulized by use of inert gases. Nebulized solutions may be inhaled directly from the nebulizing device or the nebulizing device may be attached to a face mask tent, or intermittent positive pressure breathing machine. Solution, suspension, or powder compositions may be administered, preferably orally or nasally, from devices that deliver the formulation in an appropriate manner. For example a pharmaceutical composition for topical delivery is provided comprising at least one PI3Kα inhibitor and an mTor inhibitor. Also provided are pharmaceutical compositions for topical delivery comprising a PI3Kα inhibitor, and pharmaceutical compositions for topical delivery comprising an mTor inhibitor, where the PI3Kα inhibitor is administered separately or together with the mTor inhibitor. Compositions comprising a PI3K a inhibitor and an mTor inhibitor may be formulated separately, and may further include a third therapeutic agent.
  • Other Pharmaceutical Compositions.
  • Pharmaceutical compositions may also be prepared from compositions described herein and one or more pharmaceutically acceptable excipients suitable for sublingual, buccal, rectal, intraosseous, intraocular, intranasal, epidural, or intraspinal administration. Preparations for such pharmaceutical compositions are well-known in the art. See, e.g., See, e.g., Anderson, Philip O.; Knoben, James E.; Troutman, William G, eds., Handbook of Clinical Drug Data, Tenth Edition, McGraw-Hill, 2002; Pratt and Taylor, eds., Principles of Drug Action, Third Edition, Churchill Livingston, N.Y., 1990; Katzung, ed., Basic and Clinical Pharmacology, Ninth Edition, McGraw Hill, 20037ybg; Goodman and Gilman, eds., The Pharmacological Basis of Therapeutics, Tenth Edition, McGraw Hill, 2001; Remingtons Pharmaceutical Sciences, 20th Ed., Lippincott Williams & Wilkins., 2000; Martindale, The Extra Pharmacopoeia, Thirty-Second Edition (The Pharmaceutical Press, London, 1999); all of which are incorporated by reference herein in their entirety.
  • Administration of each compounds or pharmaceutical composition of the present invention can be effected by any method that enables delivery of the compounds to the site of action. These methods include oral routes, intraduodenal routes, parenteral injection (including intravenous, intraarterial, subcutaneous, intramuscular, intravascular, intraperitoneal or infusion), topical (e.g., transdermal application), rectal administration, via local delivery by catheter or stent or through inhalation. Compounds can also be administered intraadiposally or intrathecally.
  • The amount of each compound administered will be dependent on the mammal being treated, the severity of the disorder or condition, the rate of administration, the disposition of the compound and the discretion of the prescribing physician. However, an effective dosage is in the range of about 0.001 to about 100 mg per kg body weight per day, preferably about 1 to about 35 mg/kg/day, in single or divided doses. For a 70 kg human, this would amount to about 0.05 to 7 g/day, preferably about 0.05 to about 2.5 g/day. In some instances, dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, e.g., by dividing such larger doses into several small doses for administration throughout the day.
  • In some embodiments, an inhibitor of the invention is administered in a single dose. Typically, such administration will be by injection, e.g., intravenous injection, in order to introduce the agent quickly. However, other routes may be used as appropriate. A single dose of an inhibitor of the invention may also be used for treatment of an acute condition.
  • In some embodiments, an inhibitor of the invention is administered in multiple doses. Dosing may be about once, twice, three times, four times, five times, six times, or more than six times per day. Dosing may be about once a month, once every two weeks, once a week, or once every other day. In another embodiment an inhibitor of the invention and another agent are administered together about once per day to about 6 times per day. In another embodiment the administration of an inhibitor of the invention and an agent continues for less than about 7 days. In yet another embodiment the administration continues for more than about 6, 10, 14, 28 days, two months, six months, or one year. In some cases, continuous dosing is achieved and maintained as long as necessary.
  • Administration of the agents of the invention may continue as long as necessary. In some embodiments, an agent of the invention is administered for more than 1, 2, 3, 4, 5, 6, 7, 14, or 28 days. In some embodiments, an agent of the invention is administered for less than 28, 14, 7, 6, 5, 4, 3, 2, or 1 day. In some embodiments, an agent of the invention is administered chronically on an ongoing basis, e.g., for the treatment of chronic effects.
  • An effective amount of an inhibitor of the invention may be administered in either single or multiple doses by any of the accepted modes of administration of agents having similar utilities, including rectal, buccal, intranasal and transdermal routes, by intra-arterial injection, intravenously, intraperitoneally, parenterally, intramuscularly, subcutaneously, orally, topically, or as an inhalant.
  • The compositions of the invention may also be delivered via an impregnated or coated device such as a stent, for example, or an artery-inserted cylindrical polymer. Such a method of administration may, for example, aid in the prevention or amelioration of restenosis following procedures such as balloon angioplasty. Without being bound by theory, compounds of the invention may slow or inhibit the migration and proliferation of smooth muscle cells in the arterial wall which contribute to restenosis. An inhibitor of the invention may be administered, for example, by local delivery from the struts of a stent, from a stent graft, from grafts, or from the cover or sheath of a stent. In some embodiments, an inhibitor of the invention is admixed with a matrix. Such a matrix may be a polymeric matrix, and may serve to bond the compound to the stent. Polymeric matrices suitable for such use, include, for example, lactone-based polyesters or copolyesters such as polylactide, polycaprolactonglycolide, polyorthoesters, polyanhydrides, polyaminoacids, polysaccharides, polyphosphazenes, poly(ether-ester) copolymers (e.g., PEO-PLLA); polydimethylsiloxane, poly(ethylene-vinylacetate), acrylate-based polymers or copolymers (e.g., polyhydroxyethyl methylmethacrylate, polyvinyl pyrrolidinone), fluorinated polymers such as polytetrafluoroethylene and cellulose esters. Suitable matrices may be nondegrading or may degrade with time, releasing the compound or compounds. Compounds of the invention may be applied to the surface of the stent by various methods such as dip/spin coating, spray coating, dip-coating, and/or brush-coating. The compounds may be applied in a solvent and the solvent may be allowed to evaporate, thus forming a layer of compound onto the stent. Alternatively, the compound may be located in the body of the stent or graft, for example in microchannels or micropores. When implanted, the compound diffuses out of the body of the stent to contact the arterial wall. Such stents may be prepared by dipping a stent manufactured to contain such micropores or microchannels into a solution of the compound of the invention in a suitable solvent, followed by evaporation of the solvent. Excess drug on the surface of the stent may be removed via an additional brief solvent wash. In yet other embodiments, compounds of the invention may be covalently linked to a stent or graft. A covalent linker may be used which degrades in vivo, leading to the release of the compound of the invention. Any bio-labile linkage may be used for such a purpose, such as ester, amide or anhydride linkages. Compounds of the invention may additionally be administered intravascularly from a balloon used during angioplasty. Extravascular administration of the compounds via the pericard or via advential application of formulations of the invention may also be performed to decrease restenosis.
  • A variety of stent devices which may be used as described are disclosed, for example, in the following references, all of which are hereby incorporated by reference: U.S. Pat. No. 5,451,233; U.S. Pat. No. 5,040,548; U.S. Pat. No. 5,061,273; U.S. Pat. No. 5,496,346; U.S. Pat. No. 5,292,331; U.S. Pat. No. 5,674,278; U.S. Pat. No. 3,657,744; U.S. Pat. No. 4,739,762; U.S. Pat. No. 5,195,984; U.S. Pat. No. 5,292,331; U.S. Pat. No. 5,674,278; U.S. Pat. No. 5,879,382; U.S. Pat. No. 6,344,053.
  • The compounds of the invention may be administered in dosages. It is known in the art that due to intersubject variability in compound pharmacokinetics, individualization of dosing regimen is necessary for optimal therapy. Dosing for an inhibitor of the invention may be found by routine experimentation in light of the instant disclosure.
  • The subject pharmaceutical composition may, for example, be in a form suitable for oral administration as a tablet, capsule, pill, powder, sustained release formulations, solution, suspension, for parenteral injection as a sterile solution, suspension or emulsion, for topical administration as an ointment or cream or for rectal administration as a suppository. The pharmaceutical composition may be in unit dosage forms suitable for single administration of precise dosages. The pharmaceutical composition will include a conventional pharmaceutical carrier or excipient and an inhibitor according to the invention as an active ingredient. In addition, it may include other medicinal or pharmaceutical agents, carriers, adjuvants, etc.
  • Exemplary parenteral administration forms include solutions or suspensions of active compound in sterile aqueous solutions, for example, aqueous propylene glycol or dextrose solutions. Such dosage forms can be suitably buffered, if desired.
  • The invention also provides kits. The kits include an inhibitor or compounds of the present invention as described herein, in suitable packaging, and written material that can include instructions for use, discussion of clinical studies, listing of side effects, and the like. Such kits may also include information, such as scientific literature references, package insert materials, clinical trial results, and/or summaries of these and the like, which indicate or establish the activities and/or advantages of the composition, and/or which describe dosing, administration, side effects, drug interactions, or other information useful to the health care provider. Such information may be based on the results of various studies, for example, studies using experimental animals involving in vivo models and studies based on human clinical trials. The kit may further contain another agent. In some embodiments, the compound of the present invention and the agent are provided as separate compositions in separate containers within the kit. In some embodiments, the compound of the present invention and the agent are provided as a single composition within a container in the kit. Suitable packaging and additional articles for use (e.g., measuring cup for liquid preparations, foil wrapping to minimize exposure to air, and the like) are known in the art and may be included in the kit. Kits described herein can be provided, marketed and/or promoted to health providers, including physicians, nurses, pharmacists, formulary officials, and the like. Kits may also, in some embodiments, be marketed directly to the consumer.
  • In some embodiments, the subject is a human in need of treatment for cancer, or a precancerous condition or lesion, wherein the cancer is preferably NSCL, breast, colon or pancreatic cancer. Subjects that can be treated with combination treatments of the present invention, or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivatives of the therapeutic agents, according to the methods of this invention include, for example, subjects that have been diagnosed as having psoriasis; restenosis; atherosclerosis; BPH; breast cancer such as a ductal carcinoma in duct tissue in a mammary gland, medullary carcinomas, colloid carcinomas, tubular carcinomas, and inflammatory breast cancer; ovarian cancer, including epithelial ovarian tumors such as adenocarcinoma in the ovary and an adenocarcinoma that has migrated from the ovary into the abdominal cavity; uterine cancer; cervical cancer such as adenocarcinoma in the cervix epithelial including squamous cell carcinoma and adenocarcinomas; prostate cancer, such as a prostate cancer selected from the following: an adenocarcinoma or an adenocarinoma that has migrated to the bone; pancreatic cancer such as epitheliod carcinoma in the pancreatic duct tissue and an adenocarcinoma in a pancreatic duct; bladder cancer such as a transitional cell carcinoma in urinary bladder, urothelial carcinomas (transitional cell carcinomas), tumors in the urothelial cells that line the bladder, squamous cell carcinomas, adenocarcinomas, and small cell cancers; leukemia such as acute myeloid leukemia (AML), acute lymphocytic leukemia, chronic lymphocytic leukemia, chronic myeloid leukemia, hairy cell leukemia, myelodysplasia, myeloproliferative disorders, acute myelogenous leukemia (AML), chronic myelogenous leukemia (CML), mastocytosis, chronic lymphocytic leukemia (CLL), multiple myeloma (MM), and myelodysplastic syndrome (MDS); bone cancer; lung cancer such as non-small cell lung cancer (NSCLC), which is divided into squamous cell carcinomas, adenocarcinomas, and large cell undifferentiated carcinomas, and small cell lung cancer; skin cancer such as basal cell carcinoma, melanoma, squamous cell carcinoma and actinic keratosis, which is a skin condition that sometimes develops into squamous cell carcinoma; eye retinoblastoma; cutaneous or intraocular (eye) melanoma; primary liver cancer (cancer that begins in the liver); kidney cancer; thyroid cancer such as papillary, follicular, medullary and anaplastic; AIDS-related lymphoma such as diffuse large B-cell lymphoma, B-cell immunoblastic lymphoma and small non-cleaved cell lymphoma; Kaposi's Sarcoma; viral-induced cancers including hepatitis B virus (HBV), hepatitis C virus (HCV), and hepatocellular carcinoma; human lymphotropic virus-type 1 (HTLV-1) and adult T-cell leukemia/lymphoma; and human papilloma virus (HPV) and cervical cancer; central nervous system cancers (CNS) such as primary brain tumor, which includes gliomas (astrocytoma, anaplastic astrocytoma, or glioblastoma multiforme), Oligodendroglioma, Ependymoma, Meningioma, Lymphoma, Schwannoma, and Medulloblastoma; peripheral nervous system (PNS) cancers such as acoustic neuromas and malignant peripheral nerve sheath tumor (MPNST) including neurofibromas and schwannomas, malignant fibrous cytoma, malignant fibrous histiocytoma, malignant meningioma, malignant mesothelioma, and malignant mixed Mullerian tumor; oral cavity and oropharyngeal cancer such as, hypopharyngeal cancer, laryngeal cancer, nasopharyngeal cancer, and oropharyngeal cancer; stomach cancer such as lymphomas, gastric stromal tumors, and carcinoid tumors; testicular cancer such as germ cell tumors (GCTs), which include seminomas and nonseminomas, and gonadal stromal tumors, which include Leydig cell tumors and Sertoli cell tumors; thymus cancer such as to thymomas, thymic carcinomas, Hodgkin disease, non-Hodgkin lymphomas carcinoids or carcinoid tumors; rectal cancer; and colon cancer.
  • The invention also relates to a method of treating diabetes in a mammal that comprises administering to said mammal a therapeutically effective amount of a combination treatment of the present invention.
  • In addition, the combination treatments described herein may be used to treat acne.
  • In addition, the combination treatments described herein may be used for the treatment of arteriosclerosis, including atherosclerosis. Arteriosclerosis is a general term describing any hardening of medium or large arteries. Atherosclerosis is a hardening of an artery specifically due to an atheromatous plaque.
  • Further the combination treatments described herein may be used for the treatment of glomerulonephritis. Glomerulonephritis is a primary or secondary autoimmune renal disease characterized by inflammation of the glomeruli. It may be asymptomatic, or present with hematuria and/or proteinuria. There are many recognized types, divided in acute, subacute or chronic glomerulonephritis. Causes are infectious (bacterial, viral or parasitic pathogens), autoimmune or paraneoplastic.
  • Additionally, the combination treatments described herein may be used for the treatment of bursitis, lupus, acute disseminated encephalomyelitis (ADEM), addison's disease, antiphospholipid antibody syndrome (APS), aplastic anemia, autoimmune hepatitis, coeliac disease, crohn's disease, diabetes mellitus (type 1), goodpasture's syndrome, graves' disease, guillain-barre syndrome (GBS), hashimoto's disease, inflammatory bowel disease, lupus erythematosus, myasthenia gravis, opsoclonus myoclonus syndrome (OMS), optic neuritis, ord's thyroiditis, ostheoarthritis, uveoretinitis, pemphigus, polyarthritis, primary biliary cirrhosis, reiter's syndrome, takayasu's arteritis, temporal arteritis, warm autoimmune hemolytic anemia, wegener's granulomatosis, alopecia universalis, chagas' disease, chronic fatigue syndrome, dysautonomia, endometriosis, hidradenitis suppurativa, interstitial cystitis, neuromyotonia, sarcoidosis, scleroderma, ulcerative colitis, vitiligo, vulvodynia, appendicitis, arteritis, arthritis, blepharitis, bronchiolitis, bronchitis, cervicitis, cholangitis, cholecystitis, chorioamnionitis, colitis, conjunctivitis, cystitis, dacryoadenitis, dermatomyositis, endocarditis, endometritis, enteritis, enterocolitis, epicondylitis, epididymitis, fasciitis, fibrositis, gastritis, gastroenteritis, gingivitis, hepatitis, hidradenitis, ileitis, iritis, laryngitis, mastitis, meningitis, myelitis, myocarditis, myositis, nephritis, omphalitis, oophoritis, orchitis, osteitis, otitis, pancreatitis, parotitis, pericarditis, peritonitis, pharyngitis, pleuritis, phlebitis, pneumonitis, proctitis, prostatitis, pyelonephritis, rhinitis, salpingitis, sinusitis, stomatitis, synovitis, tendonitis, tonsillitis, uveitis, vaginitis, vasculitis, or vulvitis.
  • The invention also relates to a method of treating a cardiovascular disease in a mammal that comprises administering to said mammal a therapeutically effective amount of a combination treatment of the present invention. Examples of cardiovascular conditions include, but are not limited to, atherosclerosis, restenosis, vascular occlusion, carotid obstructive disease, or ischemic conditions.
  • In another aspect, the present invention provides methods of disrupting the function of a leukocyte or disrupting a function of an osteoclast. The method includes contacting the leukocyte or the osteoclast with a function disrupting amount of a combination treatment of the invention.
  • In another aspect of the present invention, methods are provided for treating ophthalmic disease by applying one or more of the subject combination treatments to the eye of a subject. Methods are further provided for administering the combination treatments of the present invention via eye drop, intraocular injection, intravitreal injection, topically, or through the use of a drug eluting device, microcapsule, implant, or microfluidic device. In some cases, combination treatments are administered with a carrier or excipient that increases the intraocular penetrance of the compound such as an oil and water emulsion with colloid particles having an oily core surrounded by an interfacial film.
  • In some cases, the colloid particles include at least one cationic agent and at least one non-ionic sufactant such as a poloxamer, tyloxapol, a polysorbate, a polyoxyethylene castor oil derivative, a sorbitan ester, or a polyoxyl stearate. In some cases, the cationic agent is an alkylamine, a tertiary alkyl amine, a quarternary ammonium compound, a cationic lipid, an amino alcohol, a biguanidine salt, a cationic compound or a mixture thereof. In some cases the cationic agent is a biguanidine salt such as chlorhexidine, polyaminopropyl biguanidine, phenformin, alkylbiguanidine, or a mixture thereof. In some cases, the quaternary ammonium compound is a benzalkonium halide, lauralkonium halide, cetrimide, hexadecyltrimethylammonium halide, tetradecyltrimethylammonium halide, dodecyltrimethylammonium halide, cetrimonium halide, benzethonium halide, behenalkonium halide, cetalkonium halide, cetethyldimonium halide, cetylpyridinium halide, benzododecinium halide, chlorallyl methenamine halide, rnyristylalkonium halide, stearalkonium halide or a mixture of two or more thereof. In some cases, cationic agent is a benzalkonium chloride, lauralkonium chloride, benzododecinium bromide, benzethenium chloride, hexadecyltrimethylammonium bromide, tetradecyltrimethylammonium bromide, dodecyltrimethylammonium bromide or a mixture of two or more thereof. In some cases, the oil phase is mineral oil and light mineral oil, medium chain triglycerides (MCT), coconut oil; hydrogenated oils comprising hydrogenated cottonseed oil, hydrogenated palm oil, hydrogenate castor oil or hydrogenated soybean oil; polyoxyethylene hydrogenated castor oil derivatives comprising poluoxyl-40 hydrogenated castor oil, polyoxyl-60 hydrogenated castor oil or polyoxyl-100 hydrogenated castor oil.
  • The invention further provides methods of modulating a PI3K and/or mTOR kinase activity by contacting the kinase with an effective amount of a composition comprising a PI3Kα inhibitor and an mTor inhibitor. Modulate can be inhibiting or activating kinase activity. In some embodiments, the invention provides methods of inhibiting kinase activity by contacting the kinase with an effective amount of a composition comprising a PI3Kα inhibitor and an mTor inhibitor in solution. In some embodiments, the invention provides methods of inhibiting the kinase activity by contacting a cell, tissue, or organ that expresses the kinase of interest. In some embodiments, the invention provides methods of inhibiting kinase activity in subject including but not limited to rodents and mammal (e.g., human) by administering into the subject an effective amount of a composition comprising a PI3Kα inhibitor and an mTor inhibitor. In some embodiments, the percentage of inhibiting exceeds 50%, 60%, 70%, 80%, or 90%.
  • Further Combination Therapies
  • The present invention also provides methods for further combination therapies in which, in addition to a PI3Kα inhibitor and an mTor inhibitor, an agent known to modulate other pathways, or other components of the same pathway, or even overlapping sets of target enzymes is used or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof. In one aspect, such therapy includes but is not limited to the combination of the composition comprising a PI3Kα inhibitor and an mTor inhibitor with chemotherapeutic agents, therapeutic antibodies, and radiation treatment, to provide, where desired, a synergistic or additive therapeutic effect.
  • For treatment of autoimmune diseases, the subject compounds or pharmaceutical compositions can be used in combination with commonly prescribed drugs including but not limited to Enbrel®, Remicade®, Humira®, Avonex®, and Rebie. For treatment of respiratory diseaseses, the subject compounds or pharmaceutical compositions can be administered in combination with commonly prescribed drugs including but not limited to Xolair®, Advair®, Singulair®, and Spiriva®.
  • The compounds of the invention may be formulated or administered in conjunction with other agents that act to relieve the symptoms of inflammatory conditions such as encephalomyelitis, asthma, and the other diseases described herein. These agents include non-steroidal anti-inflammatory drugs (NSAIDs), e.g., acetylsalicylic acid; ibuprofen; naproxen; indomethacin; nabumetone; tolmetin; etc. Corticosteroids are used to reduce inflammation and suppress activity of the immune system. The most commonly prescribed drug of this type is Prednisone. Chloroquine (Aralen) or hydroxychloroquine (Plaquenil) may also be very useful in some individuals with lupus. They are most often prescribed for skin and joint symptoms of lupus. Azathioprine (Imuran) and cyclophosphamide (Cytoxan) suppress inflammation and tend to suppress the immune system. Other agents, e.g., methotrexate and cyclosporin are used to control the symptoms of lupus. Anticoagulants are employed to prevent blood from clotting rapidly. They range from aspirin at very low dose which prevents platelets from sticking, to heparin/coumadin.
  • In another one aspect, this invention also relates to methods and pharmaceutical compositions for inhibiting abnormal cell growth in a mammal which comprises an amount of an inhibitor of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, in combination with an amount of an anti-cancer agent (e.g., a chemotherapeutic agent). Many chemotherapeutics are presently known in the art and can be used in combination with the compounds of the invention.
  • In some embodiments, the chemotherapeutic is selected from the group consisting of mitotic inhibitors, alkylating agents, anti-metabolites, intercalating antibiotics, growth factor inhibitors, cell cycle inhibitors, enzymes, topoisomerase inhibitors, biological response modifiers, anti-hormones, angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, and anti-androgens. Non-limiting examples are chemotherapeutic agents, cytotoxic agents, and non-peptide small molecules such as Gleevec (Imatinib Mesylate), Velcade (bortezomib), Casodex (bicalutamide), Iressa (gefitinib), and Adriamycin as well as a host of chemotherapeutic agents. Non-limiting examples of chemotherapeutic agents include alkylating agents such as thiotepa and cyclosphosphamide (CYTOXAN™); alkyl sulfonates such as busulfan, improsulfan and piposulfan; aziridines such as benzodopa, carboquone, meturedopa, and uredopa; ethylenimines and methylamelamines including altretamine, triethylenemelamine, trietylenephosphoramide, triethylenethiophosphaoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, chlornaphazine, cholophosphamide, estramustine, ifosfamide, mechlorethamine, mechlorethamine oxide hydrochloride, melphalan, novembichin, phenesterine, prednimustine, trofosfamide, uracil mustard; nitrosoureas such as carmustine, chlorozotocin, fotemustine, lomustine, nimustine, ranimustine; oxazaphosphorines; nitrosoureas; triazenes; antibiotics such as anthracyclins, actinomycins and bleomycins including aclacinomysins, actinomycin, anthramycin, azaserine, bleomycins, cactinomycin, calicheamicin, carabicin, carminomycin, carzinophilin, Casodex™, chromomycins, dactinomycin, daunorubicin, detorubicin, 6-diazo-5-oxo-L-norleucine, doxorubicin, epirubicin, esorubicin, idarubicin, marcellomycin, mitomycins, mycophenolic acid, nogalamycin, olivomycins, peplomycin, potfiromycin, puromycin, quelamycin, rodorubicin, streptonigrin, streptozocin, tubercidin, ubenimex, zinostatin, zorubicin; anti-metabolites such as methotrexate and 5-fluorouracil (5-FU); folic acid analogues such as denopterin, methotrexate, pteropterin, trimetrexate; purine analogs such as fludarabine, 6-mercaptopurine, thiamiprine, thioguanine; pyrimidine analogs such as ancitabine, azacitidine, 6-azauridine, carmofur, cytarabine, dideoxyuridine, doxifluridine, enocitabine, floxuridine, androgens such as calusterone, dromostanolone propionate, epitiostanol, mepitiostane, testolactone; anti-adrenals such as aminoglutethimide, mitotane, trilostane; folic acid replenisher such as frolinic acid; aceglatone; aldophosphamide glycoside; aminolevulinic acid; amsacrine; bestrabucil; bisantrene; edatraxate; defofamine; demecolcine; diaziquone; elfomithine; elliptinium acetate; etoglucid; gallium nitrate; hydroxyurea; lentinan; lonidamine; mitoguazone; mitoxantrone; mopidamol; nitracrine; pentostatin; phenamet; pirarubicin; podophyllinic acid; 2-ethylhydrazide; procarbazine; PSK.R™; razoxane; sizofiran; spirogermanium; tenuazonic acid; triaziquone; 2,2′,2″-trichlorotriethylamine; urethan; vindesine; dacarbazine; mannomustine; mitobronitol; mitolactol; pipobroman; gacytosine; arabinoside (“Ara-C”); cyclophosphamide; thiotepa; taxanes, e.g., paclitaxel (TAXOL™, Bristol-Myers Squibb Oncology, Princeton, N.J.) and docetaxel (TAXOTERE™, Rhone-Poulenc Rorer, Antony, France); retinoic acid; esperamicins; capecitabine; gemcitabine and pharmaceutically acceptable salts, acids or derivatives of any of the above. Also included as suitable chemotherapeutic cell conditioners are anti-hormonal agents that act to regulate or inhibit hormone action on tumors such as anti-estrogens including for example tamoxifen (Nolvadex™), raloxifene, aromatase inhibiting 4(5)-imidazoles, 4-hydroxytamoxifen, trioxifene, keoxifene, LY 117018, onapristone, and toremifene (Fareston); and anti-androgens such as flutamide, nilutamide, bicalutamide, leuprolide, and goserelin; chlorambucil; gemcitabine; 6-thioguanine; mercaptopurine; methotrexate; platinum or platinum analogs and complexes such as cisplatin and carboplatin; anti-microtubule such as diterpenoids, including paclitaxel and docetaxel, or Vinca alkaloids including vinblastine, vincristine, vinflunine, vindesine, and vinorelbine; etoposide (VP-16); ifosfamide; mitomycin C; mitoxantrone; vincristine; vinorelbine; navelbine; novantrone; teniposide; daunomycin; aminopterin; xeloda; ibandronate; topoisomerase I and II inhibitors including camptothecins (e.g., camptothecin-11), topotecan, irinotecan, and epipodophyllotoxins; topoisomerase inhibitor RFS 2000; epothilone A or B; difluoromethylornithine (DMFO); histone deacetylase inhibitors; compounds which induce cell differentiation processes; gonadorelin agonists; methionine aminopeptidase inhibitors; compounds targeting/decreasing a protein or lipid kinase activity; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; anti-androgens; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitors of Ras oncogenic isoforms; telomerase inhibitors; proteasome inhibitors; compounds used in the treatment of hematologic malignancies; compounds which target, decrease or inhibit the activity of Flt-3; Hsp90 inhibitors; temozolomide (TEMODAL®); Hsp90 inhibitors such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17-DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temozolomide (TEMODAL®); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array PioPharma, AZD6244 from AstraZeneca, PD181461 or PD0325901 from Pfizer, leucovorin, EDG binders, antileukemia compounds, ribonucleotide reductase inhibitors, S-adenosylmethionine decarboxylase inhibitors, antiproliferative antibodies or other chemotherapeutic compounds. Where desired, the compounds or pharmaceutical composition of the present invention can be used in combination with commonly prescribed anti-cancer drugs such as Herceptin®, Avastin®, Erbitux®, Rituxan®, Taxol®, Arimidex®, Taxotere®, and Velcade®. Further information on compounds which may be used in conjunction with the compounds of the invention is provided below.
  • Proteasome inhibitors include compounds which target, decrease or inhibit the activity of the proteasome. Compounds which target, decrease or inhibit the activity of the proteasome include e.g., Bortezomid (Velcade™) and MLN 341. Matrix metalloproteinase inhibitors (“MMP” inhibitors) include, but are not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g., hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251, BAY 12-9566, TAA211, MMI270B or AAJ996. Compounds used in the treatment of hematologic malignancies include, but are not limited to, FMS-like tyrosine kinase inhibitors e.g., compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, 1-b-D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors e.g., compounds which target, decrease or inhibit anaplastic lymphoma kinase. Compounds which target, decrease or inhibit the activity of FMS-like tyrosine kinase receptors (Flt-3R) are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, e.g., PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.
  • Hsp90 inhibitors include compounds such as 17-AAG (17-allylaminogeldanamycin, NSC330507), 17-DMAG (17-dimethylaminoethylamino-17-demethoxy-geldanamycin, NSC707545), IPI-504, CNF1010, CNF2024, CNF1010 from Conforma Therapeutics; temozo-lomide (TEMODAL®); kinesin spindle protein inhibitors, such as SB715992 or SB743921 from GlaxoSmithKline, or pentamidine/chlorpromazine from CombinatoRx; MEK inhibitors such as ARRY142886 from Array PioPharma, AZD6244 from AstraZeneca, PD181461 from Pfizer, leucovorin, EDG binders, antileukemia compounds, ribonucleotide reductase inhibitors, S-adenosylmethionine decarboxylase inhibitors, antiproliferative antibodies or other chemotherapeutic compounds.
  • Histone deacetylase inhibitors (or “HDAC inhibitors”) include compounds which inhibit a histone deacetylase and which possess antiproliferative activity. This includes compounds disclosed in WO 02/22577, especially N-hydroxy-3-[4-[[(2-hydroxyethyl) [2-(1H-indol-3-yl) ethyl]-amino]methyl]phenyl]-2E-2-prop enamide, N-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)-ethyl]amino]methyl]phenyl]-2E-2-propenamide and pharmaceutically acceptable salts thereof. It further especially includes Suberoylanilide hydroxamic acid (SAHA).
  • Bisphosphonates for use in combination with the compounds of the invention include, but are not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid.
  • Compounds of the invention may also be used in conjunction with compounds targeting or decreasing a protein or lipid kinase activity, a protein or lipid phosphatase activity, or further anti-angiogenic compounds. Such compounds include, but are not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, e.g., compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, e.g., a N-phenyl-2-pyrimidine-amine derivative, e.g., imatinib, SU101, SU6668 and GFB-1 11; compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor-receptors (FGFR); compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF-IR), such as compounds which target, decrease or inhibit the activity of IGF-IR, especially compounds which inhibit the kinase activity of IGF-I receptor, such as those compounds disclosed in WO 02/092599 or such as OSI906, or antibodies that target the extracellular domain of IGF-I receptor such as CP-751871, R1507, AVE1642, IMC-A12, AMG479, MK-0646, SCH717454 or its growth factors; compounds targeting, decreasing or inhibiting the activity of the Trk receptor tyrosine kinase family, or ephrin B4 inhibitors; compounds targeting, decreasing or inhibiting the activity of the AxI receptor tyrosine kinase family; compounds targeting, decreasing or inhibiting the activity of the Ret receptor tyrosine kinase; compounds targeting, decreasing or inhibiting the activity of the Kit/SCFR receptor tyrosine kinase, e.g., imatinib; compounds targeting, decreasing or inhibiting the activity of the C-kit receptor tyrosine kinases—(part of the PDGFR family), such as compounds which target, decrease or inhibit the activity of the c-Kit receptor tyrosine kinase family, especially compounds which inhibit the c-Kit receptor, e.g., imatinib; compounds targeting, decreasing or inhibiting the activity of members of the c-Abl family, their gene-fusion products (e.g., BCR-AbI kinase) and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, e.g., a N-phenyl-2-pyrimidine-amine derivative, e g, imatinib or nilotinib (AMN107); PD180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825); compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK, FAK, PDK1, PKB/Akt, and Ras/MAPK family members, and/or members of the cyclin-dependent kinase family (CDK) and are especially those staurosporine derivatives disclosed in U.S. Pat. No. 5,093,330, e.g., midostaurin; examples of further compounds include e.g., UCN-01, safingol, BAY 43-9006, Bryostatin 1, Perifosine; llmofosine; RO 318220 and RO 320432; GO 6976; Isis 3521; LY333531/LY379196; isochinoline compounds such as those disclosed in WO 00/09495; FTIs; PD184352 or QAN697 (a P13K inhibitor) or AT7519 (CDK inhibitor); compounds targeting, decreasing or inhibiting the activity of protein-tyrosine kinase inhibitors, such as compounds which target, decrease or inhibit the activity of protein-tyrosine kinase inhibitors include imatinib mesylate (GLEEVEC) or tyrphostin. A tyrphostin is preferably a low molecular weight (Mr<1500) compound, or a pharmaceutically acceptable salt thereof, especially a compound selected from the benzylidenemalonitrile class or the S-arylbenzenemalonirile or bisubstrate quinoline class of compounds, more especially any compound selected from the group consisting of Tyrphostin A23/RG-50810; AG 99; Tyrphostin AG 213; Tyrphostin AG 1748; Tyrphostin AG 490; Tyrphostin B44; Tyrphostin B44 (+) enantiomer; Tyrphostin AG 555; AG 494; Tyrphostin AG 556, AG957 and adaphostin (4-{[(2,5-dihydroxyphenyl)methyl]amino}-benzoic acid adamantyl ester; NSC 680410, adaphostin).
  • Compounds of the invention may also be used in combination with compounds targeting, decreasing or inhibiting the activity of the epidermal growth factor family of receptor tyrosine kinases (EGFR, ErbB2, ErbB3, ErbB4 as homo- or heterodimers) and their mutants, such as compounds which target, decrease or inhibit the activity of the epidermal growth factor receptor family are especially compounds, proteins or antibodies which inhibit members of the EGF receptor tyrosine kinase family, e.g., EGF receptor, ErbB2, ErbB3 and ErbB4 or bind to EGF or EGF related ligands, and are in particular those compounds, proteins or monoclonal antibodies generically and specifically disclosed in WO 97/02266, e.g., the compound of ex. 39, or in EP 0 564 409, WO 99/03854, EP 0520722, EP 0 566 226, EP 0 787 722, EP 0 837 063, U.S. Pat. No. 5,747,498, WO 98/10767, WO 97/30034, WO 97/49688, WO 97/38983 and, especially, WO 96/30347 (e.g., compound known as CP 358774), WO 96/33980 (e.g., compound ZD 1839) and WO 95/03283 (e.g., compound ZM105180); e.g., trastuzumab (Herceptin™), cetuximab (Erbitux™), Iressa, Tarceva, OSI-774, Cl-1033, EKB-569, GW-2016, E1.1, E2.4, E2.5, E6.2, E6.4, E2.1 1, E6.3 or E7.6.3, and 7H-pyrrolo-[2,3-d]pyrimidine derivatives which are disclosed in WO 03/013541; and compounds targeting, decreasing or inhibiting the activity of the c-Met receptor, such as compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF. Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g., unrelated to protein or lipid kinase inhibition e.g., thalidomide (THALOMID) and TNP-470.
  • Non-receptor kinase angiogenesis inhibitors may also be useful in conjunction with the compounds of the present invention. Angiogenesis in general is linked to erbB21EGFR signaling since inhibitors of erbB2 and EGFR have been shown to inhibit angiogenesis, primarily VEGF expression. Accordingly, non-receptor tyrosine kinase inhibitors may be used in combination with the compounds of the present invention. For example, anti-VEGF antibodies, which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alphav beta3) that will inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the disclosed compounds. (See Bruns C J et al (2000), Cancer Res., 60: 2926-2935; Schreiber A B, Winkler M E, and Derynck R. (1986), Science, 232: 1250-1253; Yen L et al. (2000), Oncogene 19: 3460-3469).
  • Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase include e.g., inhibitors of phosphatase 1, phosphatase 2A, or CDC25, e.g., okadaic acid or a derivative thereof. Compounds which induce cell differentiation processes are e.g., retinoic acid, α- γ- or δ-tocopherol or α- γ- or δ-tocotrienol. Cyclooxygenase inhibitors include, but are not limited to, e.g., Cox-2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CELEBREX), rofecoxib (VIOXX), etoricoxib, valdecoxib or a 5-alkyl-2-arylaminophenylacetic acid, e.g., 5-methyl-2-(2′-chloro-6′-fluoroanilino)phenyl acetic acid, and lumiracoxib.
  • Heparanase inhibitors includes compounds which target, decrease or inhibit heparin sulfate degradation, including, but not limited to, PI-88. Biological response modifiers include lymphokines and interferons, e.g., interferon γ Inhibitors of Ras oncogenic isoforms include H-Ras, K-Ras, N-Ras, and other compounds which target, decrease or inhibit the oncogenic activity of Ras. Farnesyl transferase inhibitors include, but are not limited to, e.g., L-744832, DK8G557 and R115777 (Zarnestra).
  • Telomerase inhibitors include compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, e.g., telomestatin. Methionine aminopeptidase inhibitors are, for example, compounds which target, decrease or inhibit the activity of methionine aminopeptidase. Compounds which target, decrease or inhibit the activity of methionine aminopeptidase are e.g., bengamide or a derivative thereof.
  • Antiproliferative antibodies include, but are not limited to, trastuzumab (Herceptin™), Trastuzumab-DM1, erbitux, bevacizumab (Avastin™), rituximab (Rituxan®), PRO64553 (anti-CD40) and 2C4 Antibody. By antibodies is meant e.g., intact monoclonal antibodies, polyclonal antibodies, multispecific antibodies formed from at least 2 intact antibodies, and antibodies fragments so long as they exhibit the desired biological activity.
  • For the treatment of acute myeloid leukemia (AML), compounds of the invention can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML. In particular, compounds of the invention can be administered in combination with, e.g., farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP-16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
  • Antileukemic compound for use in combination with compounds of the invention include, for example, Ara-C, a pyrimidine analog, which is the 2′-alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate. Compounds which target, decrease or inhibit activity of histone deacetylase (HDAC) inhibitors such as sodium butyrate and suberoylanilide hydroxamic acid (SAHA) inhibit the activity of the enzymes known as histone deacetylases. Specific HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in U.S. Pat. No. 6,552,065, in particular, Λ/-hydroxy-3-[4-[[[2-(2-methyl-1H-indol-3-yl)-ethyl]-amino]methyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof and Λ/-hydroxy-3-[4-[(2-hydroxyethyl){2-(1/-/-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2-propenamide, or a pharmaceutically acceptable salt thereof, e.g., the lactate salt.
  • Somatostatin receptor antagonists include compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230 (pasireotide). Tumor cell damaging approaches include approaches such as ionizing radiation, e.g., ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4th Edition, Vol. 1, pp. 248-275 (1993). EDG binders includes immunosuppressants that modulate lymphocyte recirculation, such as FTY720.
  • Ribonucleotide reductase inhibitors include pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin. Ribonucleotide reductase inhibitors are e.g., hydroxyurea or 2-hydroxy-1/-/-isoindole-1,3-dione derivatives, such as PL-1, PL-2, PL-3, PL-4, PL-5, PL-6, PL-7 or PL-8 mentioned in Nandy et al., Acta Oncologica, Vol. 33, No. 8, pp. 953-961 (1994).
  • S-adenosylmethionine decarboxylase inhibitors include, but are not limited to the compounds disclosed in U.S. Pat. No. 5,461,076.
  • Also included are in particular those compounds, proteins or monoclonal antibodies of VEGF disclosed in WO 98/35958, e.g., 1-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof, e.g., the succinate, or in WO 00/09495, WO 00/27820, WO 00/59509, WO 98/11223, WO 00/27819 and EP 0 769 947; those as described by Prewett et al, Cancer Res, Vol. 59, pp. 5209-5218 (1999); Yuan et al., Proc Natl Acad Sci USA, Vol. 93, pp. 14765-14770 (1996); Zhu et al., Cancer Res, Vol. 58, pp. 3209-3214 (1998); and Mordenti et al., Toxicol Pathol, Vol. 27, No. 1, pp. 14-21 (1999); in WO 00/37502 and WO 94/10202; ANGIOSTATIN, described by O'Reilly et al., Cell, Vol. 79, pp. 315-328 (1994); ENDOSTATIN, described by O'Reilly et al., Cell, Vol. 88, pp. 277-285 (1997); anthranilic acid amides; ZD4190; ZD6474; SU5416; SU6668; bevacizumab; or anti-VEGF antibodies or anti-VEGF receptor antibodies, e.g., rhuMAb and RHUFab, VEGF aptamer e.g., Macugon; FLT-4 inhibitors, FLT-3 inhibitors, VEGFR-2 IgGI antibody, Angiozyme (RPI 4610) and Bevacizumab (Avastin™).
  • The compounds of the invention are also useful as co-therapeutic compounds for use in combination with other drug substances such as anti-inflammatory, bronchodilatory or antihistamine drug substances, particularly in the treatment of obstructive or inflammatory airways diseases such as those mentioned hereinbefore, for example as potentiators of therapeutic activity of such drugs or as a means of reducing required dosaging or potential side effects of such drugs. An inhibitor of the invention may be mixed with the other drug substance in a fixed pharmaceutical composition or it may be administered separately, before, simultaneously with or after the other drug substance. Accordingly the invention includes a combination of an inhibitor of the invention as described with an anti-inflammatory, bronchodilatory, antihistamine or anti-tussive drug substance, said compound of the invention and said drug substance being in the same or different pharmaceutical composition. Suitable anti-inflammatory drugs include steroids, in particular glucocorticosteroids such as budesonide, beclamethasone dipropionate, fluticasone propionate, ciclesonide or mometasone furoate, or steroids described in WO 02/88167, WO 02/12266, WO 02/100879, WO 02/00679 (especially those of Examples 3, 1 1, 14, 17, 19, 26, 34, 37, 39, 51, 60, 67, 72, 73, 90, 99 and 101), WO 03/035668, WO 03/048181, WO 03/062259, WO 03/064445, WO 03/072592, non-steroidal glucocorticoid receptor agonists such as those described in WO 00/00531, WO 02/10143, WO 03/082280, WO 03/082787, WO 03/104195, WO 04/005229; LTB4 antagonists such LY29311 1, CGS025019C, CP-195543, SC-53228, BIIL 284, ONO 4057, SB 209247 and those described in U.S. Pat. No. 5,451,700; LTD4 antagonists such as montelukast and zafirlukast; PDE4 inhibitors such cilomilast (Ariflo® GlaxoSmithKline), Roflumilast (Byk Gulden), V-1 1294A (Napp), BAY19-8004 (Bayer), SCH-351591 (Schering-Plough), Arofylline (Almirall Prodesfarma), PD189659/PD168787 (Parke-Davis), AWD-12-281 (Asta Medica), CDC-801 (Celgene), SeICID™ CC-10004 (Celgene), VM554/UM565 (Vernalis), T-440 (Tanabe), KW-4490 (Kyowa Hakko Kogyo), and those disclosed in WO 92/19594, WO 93/19749, WO 93/19750, WO 93/19751, WO 98/18796, WO 99/16766, WO 01/13953, WO 03/104204, WO 03/104205, WO 03/39544, WO 04/000814, WO 04/000839, WO 04/005258, WO 04/018450, WO 04/018451, WO 04/018457, WO 04/018465, WO 04/018431, WO 04/018449, WO 04/018450, WO 04/018451, WO 04/018457, WO 04/018465, WO 04/019944, WO 04/019945, WO 04/045607 and WO 04/037805; A2a agonists such as those disclosed in EP 409595A2, EP 1052264, EP 1241176, WO 94/17090, WO 96/02543, WO 96/02553, WO 98/28319, WO 99/24449, WO 99/24450, WO 99/24451, WO 99/38877, WO 99/41267, WO 99/67263, WO 99/67264, WO 99/67265, WO 99/67266, WO 00/23457, WO 00/77018, WO 00/78774, WO 01/23399, WO 01/27130, WO 01/27131, WO 01/60835, WO 01/94368, WO 02/00676, WO 02/22630, WO 02/96462, WO 03/086408, WO 04/039762, WO 04/039766, WO 04/045618 and WO 04/046083; A2b antagonists such as those described in WO 02/42298; and beta-2 adrenoceptor agonists such as albuterol (salbutamol), metaproterenol, terbutaline, salmeterol fenoterol, procaterol, and especially, formoterol and pharmaceutically acceptable salts thereof, and compounds (in free or salt or solvate form) of formula I of WO 0075114, which document is incorporated herein by reference, preferably compounds of the Examples thereof, as well as compounds (in free or salt or solvate form) of formula I of WO 04/16601, and also compounds of WO 04/033412. Suitable bronchodilatory drugs include anticholinergic or antimuscarinic compounds, in particular ipratropium bromide, oxitropium bromide, tiotropium salts and CHF 4226 (Chiesi), and glycopyrrolate, but also those described in WO 01/041 18, WO 02/51841, WO 02/53564, WO 03/00840, WO 03/87094, WO 04/05285, WO 02/00652, WO 03/53966, EP 424021, U.S. Pat. No. 5,171,744, U.S. Pat. No. 3,714,357, WO 03/33495 and WO 04/018422.
  • Suitable antihistamine drug substances include cetirizine hydrochloride, acetaminophen, clemastine fumarate, promethazine, loratidine, desloratidine, diphenhydramine and fexofenadine hydrochloride, activastine, astemizole, azelastine, ebastine, epinastine, mizolastine and tefenadine as well as those disclosed in WO 03/099807, WO 04/026841 and JP 2004107299.
  • Other useful combinations of compounds of the invention with anti-inflammatory drugs are those with antagonists of chemokine receptors, e.g., CCR-1, CCR-2, CCR-3, CCR-4, CCR-5, CCR-6, CCR-7, CCR-8, CCR-9 and CCR10, CXCR1, CXCR2, CXCR3, CXCR4, CXCR5, particularly CCR-5 antagonists such as Schering-Plough antagonists SC-351 125, SCH-55700 and SCH-D, Takeda antagonists such as TAK-770, and CCR-5 antagonists described in U.S. Pat. No. 6,166,037 (particularly claims 18 and 19), WO 00/66558 (particularly claim 8), WO 00/66559 (particularly claim 9), WO 04/018425 and WO 04/026873.
  • Anti-microtubule or anti-mitotic agents include phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle. Examples of anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids. Diterpenoids, which are derived from natural sources, are phase specific anti-cancer agents that operate at the G2/M phases of the cell cycle. It is believed that the diterpenoids stabilize the β-tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel. Paclitaxel, 5β,20-epoxy-1,2α,4,7β,10β,13α-hexa-hydroxytax-11-en-9-one 4,10-diacetate 2-benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL®. It is a member of the taxane family of terpenes. One mechanism for its activity relates to paclitaxel's capacity to bind tubulin, thereby inhibiting cancer cell growth. Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States and for the treatment of breast cancer. It is a potential candidate for treatment of neoplasms in the skin and head and neck carcinomas. The compound also shows potential for the treatment of polycystic kidney disease, lung cancer and malaria. Treatment of subjects with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R. J. et. al, Cancer Chemotherapy Pocket Guide, 1998) related to the duration of dosing above a threshold concentration (50 nM) (Kearns, C. M. et. al., Seminars in Oncology, 3(6) p. 16-23, 1995). Docetaxel, (2R,3S)-N-carboxy-3-phenylisoserine, N-tert-butyl ester, 13-ester with 5β-20-epoxy-1,2α,4,7β,10β,13α-hexahydroxytax-1-1-en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE®. Docetaxel is indicated for the treatment of breast cancer. Docetaxel is a semisynthetic derivative of paclitaxel q.v., prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree. The dose limiting toxicity of docetaxel is neutropenia.
  • Other compounds that can regulate apoptosis (e.g., BCL-2 inhibitors) can be used in conjunction.
  • Vinca alkaloids include phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine. Vinblastine, vincaleukoblastine sulfate, is commercially available as VELBAN® as an injectable solution. Although it has possible indication as a second line therapy of various solid tumors, it is primarily indicated in the treatment of testicular cancer and various lymphomas including Hodgkin's Disease, and lymphocytic and histiocytic lymphomas. Myelosuppression is the dose limiting side effect of vinblastine. Vincristine, vincaleukoblastine, 22-oxo-, sulfate, is commercially available as ONCOVIN® as an injectable solution. Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas. Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent myelosupression and gastrointestinal mucositis effects occur. Vinorelbine, 3′,4′-didehydro-4′-deoxy-C′-norvincaleukoblastine [R-(R*,R*)-2,3-dihydroxybutanedioate (1:2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE®), is a semisynthetic vinca alkaloid. Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non-small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine.
  • Platinum coordination complexes include non-phase specific anti-cancer agents, which interact with DNA. The platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor. Examples of platinum coordination complexes include, but are not limited to, cisplatin and carboplatin. Cisplatin, cis-diamminedichloroplatinum, is commercially available as PLATINOL® as an injectable solution. Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer. The primary dose limiting side effects of cisplatin are nephrotoxicity, which may be controlled by hydration and diuresis, and ototoxicity. Carboplatin, platinum, diammine [1,1-cyclobutane-dicarboxylate(2-)-O,O′], is commercially available as PARAPLATIN®) as an injectable solution. Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma. Bone marrow suppression is the dose limiting toxicity of carboplatin.
  • Alkylating agents include non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death. Examples of alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine. Cyclophosphamide, 2-[bis(2-chloroethyl)amino]tetrahydro-2H-1,3,2-oxazaphosphorine 2-oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN®. Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias. Alopecia, nausea, vomiting and leukopenia are the most common dose limiting side effects of cyclophosphamide. Melphalan, 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN®. Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan. Chlorambucil, 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease. Bone marrow suppression is the most common dose limiting side effect of chlorambucil. Busulfan, 1,4-butanediol dimethanesulfonate, is commercially available as MYLERAN® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia. Bone marrow suppression is the most common dose limiting side effects of busulfan. Carmustine, 1,3-[bis(2-chloroethyl)-1-nitrosourea, is commercially available as single vials of lyophilized material as BiCNU®. Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non-Hodgkin's lymphomas. Delayed myelosuppression is the most common dose limiting side effects of carmustine. Dacarbazine, 5-(3,3-dimethyl-1-triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome®. Dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin's Disease. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dacarbazine.
  • Antibiotic anti-neoplastics include non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death. Examples of antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins. Dactinomycin, also know as Actinomycin D, is commercially available in injectable form as COSMEGEN®. Dactinomycin is indicated for the treatment of Wilm's tumor and rhabdomyosarcoma. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dactinomycin. Daunorubicin, (8S-cis+8-acetyl-10-[(3-amino-2,3,6-trideoxy-α-L-lyxo-hexopyranosyl)oxy]-7,8,9,10-tetrahydro-6,8,11-trihydroxy-1-methoxy-5,12 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME® or as an injectable as CERUBIDINE®. Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma. Myelosuppression is the most common dose limiting side effect of daunorubicin. Doxorubicin, (8S,10S)-10-[(3-amino-2,3,6-trideoxy-α-L-lyxo-hexopyranosyl)oxy]-8-glycoloyl, 7,8,9,10-tetrahydro-6,8,11-trihydroxy-1-methoxy-5,12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX® or ADRIAMYCIN RDF®. Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblastic leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas. Myelosuppression is the most common dose limiting side effect of doxorubicin. Bleomycin, a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of Streptomyces verticillus, is commercially available as BLENOXANE®. Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas Pulmonary and cutaneous toxicities are the most common dose limiting side effects of bleomycin.
  • Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins. Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows. Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide. Etoposide, 4′-demethyl-epipodophyllotoxin 9[4,6-0-(R)-ethylidene-β-D-glucopyranoside], is commercially available as an injectable solution or capsules as VePESID® and is commonly known as VP-16. Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers. Myelosuppression is the most common side effect of etoposide. The incidence of leucopenia tends to be more severe than thrombocytopenia. Teniposide, 4′-demethyl-epipodophyllotoxin 9[4,6-0-(R)-thenylidene-β-D-glucopyranoside], is commercially available as an injectable solution as VUMON® and is commonly known as VM-26. Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children. Myelosuppression is the most common dose limiting side effect of teniposide. Teniposide can induce both leucopenia and thrombocytopenia. Other topoisomerase II inhibitors include epirubicin, idarubicin, nemorubicin, mitoxantrone, and losoxantrone.
  • Antimetabolite neoplastic agents include phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows. Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mercaptopurine, thioguanine, and gemcitabine. 5-fluorouracil, 5-fluoro-2,4-(1H,3H) pyrimidinedione, is commercially available as fluorouracil. Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death. 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas. Myelosuppression and mucositis are dose limiting side effects of 5-fluorouracil. Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate. Cytarabine, 4-amino-1-β-D-arabinofuranosyl-2(1H)-pyrimidinone, is commercially available as CYTOSAR-U® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5-azacytidine and 2′,2′-difluorodeoxycytidine (gemcitabine). Cytarabine induces leucopenia, thrombocytopenia, and mucositis. Mercaptopurine, 1,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURINETHOL®. Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression and gastrointestinal mucositis are expected side effects of mercaptopurine at high doses. A useful mercaptopurine analog is azathioprine. Thioguanine, 2-amino-1,7-dihydro-6H-purine-6-thione, is commercially available as TABLOID®. Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression, including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of thioguanine administration. However, gastrointestinal side effects occur and can be dose limiting. Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine. Gemcitabine, 2′-deoxy-2′,2′-difluorocytidine monohydrochloride (β-isomer), is commercially available as GEMZAR®. Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the G1/S boundary. Gemcitabine is indicated in combination with cisplatin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer. Myelosuppression, including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of gemcitabine administration. Methotrexate, N-[4-[[(2,4-diamino-6-pteridinyl)methyl]methylamino]benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate. Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder. Myelosuppression (leucopenia, thrombocytopenia, and anemia) and mucositis are expected side effect of methotrexate administration.
  • Topoisomerase I inhibitors include camptothecins such as camptothecin and camptothecin derivatives. Camptothecin cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan and topotecan. Irinotecan HCl, (4S)-4,11-diethyl-4-hydroxy-9-[(4-piperidinopiperidino) carbonyloxy]-1H-pyrano[3′,4′,6,7]indolizino[1,2-b]quinoline-3,14(4H,12H)-dione hydrochloride, is commercially available as the injectable solution CAMPTOSAR®. Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN-38, to the topoisomerase I-DNA complex. It is believed that cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I:DNA:irinotecan or SN-38 ternary complex with replication enzymes. Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum. The dose limiting side effects of irinotecan HCl are myelosuppression, including neutropenia, and GI effects, including diarrhea. Topotecan HCl, (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1H-pyrano[3′,4′,6,7]-indolizino[1,2-b]quinoline-3,14-(4H,12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN®. Topotecan is a derivative of camptothecin which binds to the topoisomerase I-DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule. Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer. The dose limiting side effect of topotecan HCl is myelosuppression, primarily neutropenia.
  • Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer. Examples of hormones and hormonal analogues useful in cancer treatment include, but are not limited to, adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children; aminoglutethimide and other aromatase inhibitors such as aminoglutethimide, roglethimide, pyridoglutethimide, trilostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole, letrazole, formestane, atamestane and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestrins such as megestrol acetate useful in the treatment of hormone dependent breast cancer and endometrial carcinoma; estrogens, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide, cyproterone acetate and 5α-reductases such as finasteride and dutasteride, useful in the treatment of prostatic carcinoma and benign prostatic hypertrophy; anti-estrogens such as fulvestrant, tamoxifen, toremifene, raloxifene, droloxifene, iodoxyfene, as well as selective estrogen receptor modulators (SERMS) such those described in U.S. Pat. Nos. 5,681,835, 5,877,219, and 6,207,716, useful in the treatment of hormone dependent breast carcinoma and other susceptible cancers; and gonadotropin-releasing hormone (GnRH) and analogues thereof which stimulate the release of leutinizing hormone (LH) and/or follicle stimulating hormone (FSH) for the treatment prostatic carcinoma, for instance, LHRH agonists and antagonists such as abarelix, goserelin, goserelin acetate and luprolide. SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, PI3-K p85 subunit, Src family kinases, adaptor molecules (Shc, Crk, Nck, Grb2) and Ras-GAP. SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T. E. (1995), Journal of Pharmacological and Toxicological Methods. 34(3) 125-32 Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta). IkB kinase family (IKKa, IKKb), PKB family kinases, akt kinase family members, and TGF beta receptor kinases. Such Serine/Threonine kinases and inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal of Biochemistry. 126 (5) 799-803; Brodt, P, Samani, A., and Navab, R. (2000), Biochemical Pharmacology, 60. 1101-1107; Massague, J., Weis-Garcia, F. (1996) Cancer Surveys. 27:41-64; Philip, P. A., and Harris, A. L. (1995), Cancer Treatment and Research. 78: 3-27, Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10), 2000, 223-226; U.S. Pat. No. 6,268,391; and Martinez-Iacaci, L., et al, Int. J. Cancer (2000), 88(1), 44-52.
  • Also of interest for use with the compounds of the invention are Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues. Such signal inhibitors are described in Powis, G., and Kozikowski A., (1994) New Molecular Targets for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC press 1994, London.
  • Another group of inhibitors are signal transduction pathway inhibitors such as inhibitors of Ras Oncogene. Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy. Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras, thereby acting as antiproliferation agents. Ras oncogene inhibition is discussed in Scharovsky, 0. G., Rozados, V. R., Gervasoni, S. I. Matar, P. (2000), Journal of Biomedical Science. 7(4) 292-8; Ashby, M. N. (1998), Current Opinion in Lipidology. 9 (2) 99-102; and BioChim Biophys. Acta, (19899) 1423(3):19-30.
  • This invention further relates to a method for using the compounds or pharmaceutical composition in combination with other tumor treatment approaches, including surgery, ionizing radiation, photodynamic therapy, or implants, e.g., with corticosteroids, hormones, or used as radiosensitizers.
  • One such approach may be, for example, radiation therapy in inhibiting abnormal cell growth or treating the hyperproliferative disorder in the mammal. Techniques for administering radiation therapy are known in the art, and these techniques can be used in the combination therapy described herein. The administration of the compound of the invention in this combination therapy can be determined as described herein.
  • Radiation therapy can be administered through one of several methods, or a combination of methods, including without limitation external-beam therapy, internal radiation therapy, implant radiation, stereotactic radiosurgery, systemic radiation therapy, radiotherapy and permanent or temporary interstitial brachytherapy. The term “brachytherapy,” as used herein, refers to radiation therapy delivered by a spatially confined radioactive material inserted into the body at or near a tumor or other proliferative tissue disease site. The term is intended without limitation to include exposure to radioactive isotopes (e.g., At-211, I-131, I-125, Y-90, Re-186, Re-188, Sm-153, Bi-212, P-32, and radioactive isotopes of Lu). Suitable radiation sources for use as a cell conditioner of the present invention include both solids and liquids. By way of non-limiting example, the radiation source can be a radionuclide, such as I-125, I-131, Yb-169, Ir-192 as a solid source, 1-125 as a solid source, or other radionuclides that emit photons, beta particles, gamma radiation, or other therapeutic rays. The radioactive material can also be a fluid made from any solution of radionuclide(s), e.g., a solution of I-125 or I-131, or a radioactive fluid can be produced using a slurry of a suitable fluid containing small particles of solid radionuclides, such as Au-198, Y-90. Moreover, the radionuclide(s) can be embodied in a gel or radioactive micro spheres.
  • Without being limited by any theory, the compounds of the present invention can render abnormal cells more sensitive to treatment with radiation for purposes of killing and/or inhibiting the growth of such cells. Accordingly, this invention further relates to a method for sensitizing abnormal cells in a mammal to treatment with radiation which comprises administering to the mammal an amount of an inhibitor of the present invention or pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, which amount is effective is sensitizing abnormal cells to treatment with radiation. The amount of the compound, salt, or solvate in this method can be determined according to the means for ascertaining effective amounts of such compounds described herein.
  • Photodynamic therapy includes therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers. Examples of photodynamic therapy include treatment with compounds, such as e.g., VISUDYNE and porfimer sodium. Angiostatic steroids include compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-α-epihydrocotisol, cortexolone, 17α-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • Implants containing corticosteroids include compounds, such as e.g., fluocinolone and dexamethasone. Other chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
  • The compounds or pharmaceutical compositions of the present invention can be used in combination with an amount of one or more substances selected from anti-angiogenesis agents, signal transduction inhibitors, and antiproliferative agents.
  • Anti-angiogenesis agents, such as MMP-2 (matrix-metalloproteinase 2) inhibitors, MMP-9 (matrix-metalloproteinase 9) inhibitors, and COX-11 (cyclooxygenase 11) inhibitors, can be used in conjunction with an inhibitor of the present invention and pharmaceutical compositions described herein. Examples of useful COX-II inhibitors include CELEBREX™ (alecoxib), valdecoxib, and rofecoxib. Examples of useful matrix metalloproteinase inhibitors are described in WO 96/33172 (published Oct. 24, 1996), WO 96/27583 (published Mar. 7, 1996), European Patent Application No. 97304971.1 (filed Jul. 8, 1997), European Patent Application No. 99308617.2 (filed Oct. 29, 1999), WO 98/07697 (published Feb. 26, 1998), WO 98/03516 (published Jan. 29, 1998), WO 98/34918 (published Aug. 13, 1998), WO 98/34915 (published Aug. 13, 1998), WO 98/33768 (published Aug. 6, 1998), WO 98/30566 (published Jul. 16, 1998), European Patent Publication 606,046 (published Jul. 13, 1994), European Patent Publication 931, 788 (published Jul. 28, 1999), WO 90/05719 (published May 31,1990), WO 99/52910 (published Oct. 21, 1999), WO 99/52889 (published Oct. 21, 1999), WO 99/29667 (published Jun. 17, 1999), PCT International Application No. PCT/IB98/01113 (filed Jul. 21, 1998), European Patent Application No. 99302232.1 (filed Mar. 25, 1999), Great Britain Patent Application No. 9912961.1 (filed Jun. 3, 1999), U.S. Provisional Application No. 60/148,464 (filed Aug. 12, 1999), U.S. Pat. No. 5,863,949 (issued Jan. 26, 1999), U.S. Pat. No. 5,861,510 (issued Jan. 19, 1999), and European Patent Publication 780,386 (published Jun. 25, 1997), all of which are incorporated herein in their entireties by reference. In some embodiments, MMP-2 and MMP-9 inhibitors have little or no activity inhibiting MMP-1, or selectively inhibit MMP-2 and/or AMP-9 relative to the other matrix-metalloproteinases (i. e., MAP-1, MMP-3, MMP-4, MMP-5, MMP-6, MMP-7, MMP-8, MMP-10, MMP-11, MMP-12, and MMP-13). Some specific examples of MMP inhibitors useful in the present invention are AG-3340, RO 32-3555, and RS 13-0830.
  • The invention also relates to a method of and to a pharmaceutical composition of treating a cardiovascular disease in a mammal which comprises an amount of an inhibitor of the present invention, or a pharmaceutically acceptable salt, ester, prodrug, solvate, hydrate or derivative thereof, or an isotopically-labeled derivative thereof, and an amount of one or more therapeutic agents use for the treatment of cardiovascular diseases.
  • Exemplary agents for use in cardiovascular disease applications are anti-thrombotic agents, e.g., prostacyclin and salicylates, thrombolytic agents, e.g., streptokinase, urokinase, tissue plasminogen activator (TPA) and anisoylated plasminogen-streptokinase activator complex (APSAC), anti-platelets agents, e.g., acetyl-salicylic acid (ASA) and clopidrogel, vasodilating agents, e.g., nitrates, calcium channel blocking drugs, anti-proliferative agents, e.g., colchicine and alkylating agents, intercalating agents, growth modulating factors such as interleukins, transformation growth factor-beta and congeners of platelet derived growth factor, monoclonal antibodies directed against growth factors, anti-inflammatory agents, both steroidal and non-steroidal, and other agents that can modulate vessel tone, function, arteriosclerosis, and the healing response to vessel or organ injury post intervention. Antibiotics can also be included in combinations or coatings comprised by the invention. Moreover, a coating can be used to effect therapeutic delivery focally within the vessel wall. By incorporation of the active agent in a swellable polymer, the active agent will be released upon swelling of the polymer.
  • Medicaments which may be administered in conjunction with the compounds described herein include any suitable drugs usefully delivered by inhalation for example, analgesics, e.g., codeine, dihydromorphine, ergotamine, fentanyl or morphine; anginal preparations, e.g., diltiazem; antiallergics, e.g., cromoglycate, ketotifen or nedocromil; anti-infectives, e.g., cephalosporins, penicillins, streptomycin, sulphonamides, tetracyclines or pentamidine; antihistamines, e.g., methapyrilene; anti-inflammatories, e.g., beclomethasone, flunisolide, budesonide, tipredane, triamcinolone acetonide or fluticasone; antitussives, e.g., noscapine; bronchodilators, e.g., ephedrine, adrenaline, fenoterol, formoterol, isoprenaline, metaproterenol, phenylephrine, phenylpropanolamine, pirbuterol, reproterol, rimiterol, salbutamol, salmeterol, terbutalin, isoetharine, tulobuterol, orciprenaline or (−)-4-amino-3,5-dichloro-α-[[[6-[2-(2-pyridinyl)ethoxy]hexyl]-amino]methyl]benzenemethanol; diuretics, e.g., amiloride; anticholinergics e.g., ipratropium, atropine or oxitropium; hormones, e.g., cortisone, hydrocortisone or prednisolone; xanthines e.g., aminophylline, choline theophyllinate, lysine theophyllinate or theophylline; and therapeutic proteins and peptides, e.g., insulin or glucagon. It will be clear to a person skilled in the art that, where appropriate, the medicaments may be used in the form of salts (e.g., as alkali metal or amine salts or as acid addition salts) or as esters (e.g., lower alkyl esters) or as solvates (e.g., hydrates) to optimize the activity and/or stability of the medicament.
  • Other exemplary therapeutic agents useful for a combination therapy include but are not limited to agents as described above, radiation therapy, hormone antagonists, hormones and their releasing factors, thyroid and antithyroid drugs, estrogens and progestins, androgens, adrenocorticotropic hormone; adrenocortical steroids and their synthetic analogs; inhibitors of the synthesis and actions of adrenocortical hormones, insulin, oral hypoglycemic agents, and the pharmacology of the endocrine pancreas, agents affecting calcification and bone turnover: calcium, phosphate, parathyroid hormone, vitamin D, calcitonin, vitamins such as water-soluble vitamins, vitamin B complex, ascorbic acid, fat-soluble vitamins, vitamins A, K, and E, growth factors, cytokines, chemokines, muscarinic receptor agonists and antagonists; anticholinesterase agents; agents acting at the neuromuscular junction and/or autonomic ganglia; catecholamines, sympathomimetic drugs, and adrenergic receptor agonists or antagonists; and 5-hydroxytryptamine (5-HT, serotonin) receptor agonists and antagonists.
  • Therapeutic agents can also include agents for pain and inflammation such as histamine and histamine antagonists, bradykinin and bradykinin antagonists, 5-hydroxytryptamine (serotonin), lipid substances that are generated by biotransformation of the products of the selective hydrolysis of membrane phospholipids, eicosanoids, prostaglandins, thromboxanes, leukotrienes, aspirin, nonsteroidal anti-inflammatory agents, analgesic-antipyretic agents, agents that inhibit the synthesis of prostaglandins and thromboxanes, selective inhibitors of the inducible cyclooxygenase, selective inhibitors of the inducible cyclooxygenase-2, autacoids, paracrine hormones, somatostatin, gastrin, cytokines that mediate interactions involved in humoral and cellular immune responses, lipid-derived autacoids, eicosanoids, β-adrenergic agonists, ipratropium, glucocorticoids, methylxanthines, sodium channel blockers, opioid receptor agonists, calcium channel blockers, membrane stabilizers and leukotriene inhibitors.
  • Additional therapeutic agents contemplated herein include diuretics, vasopressin, agents affecting the renal conservation of water, rennin, angiotensin, agents useful in the treatment of myocardial ischemia, anti-hypertensive agents, angiotensin converting enzyme inhibitors, β-adrenergic receptor antagonists, agents for the treatment of hypercholesterolemia, and agents for the treatment of dyslipidemia.
  • Other therapeutic agents contemplated include drugs used for control of gastric acidity, agents for the treatment of peptic ulcers, agents for the treatment of gastroesophageal reflux disease, prokinetic agents, antiemetics, agents used in irritable bowel syndrome, agents used for diarrhea, agents used for constipation, agents used for inflammatory bowel disease, agents used for biliary disease, agents used for pancreatic disease. Therapeutic agents used to treat protozoan infections, drugs used to treat Malaria, Amebiasis, Giardiasis, Trichomoniasis, Trypanosomiasis, and/or Leishmaniasis, and/or drugs used in the chemotherapy of helminthiasis. Other therapeutic agents include antimicrobial agents, sulfonamides, trimethoprim-sulfamethoxazole quinolones, and agents for urinary tract infections, penicillins, cephalosporins, and other, 13-Lactam antibiotics, an agent comprising an aminoglycoside, protein synthesis inhibitors, drugs used in the chemotherapy of tuberculosis, mycobacterium avium complex disease, and leprosy, antifungal agents, antiviral agents including nonretroviral agents and antiretroviral agents.
  • Examples of therapeutic antibodies that can be combined with a subject compound include but are not limited to anti-receptor tyrosine kinase antibodies (cetuximab, panitumumab, trastuzumab), anti CD20 antibodies (rituximab, tositumomab), and other antibodies such as alemtuzumab, bevacizumab, and gemtuzumab.
  • Moreover, therapeutic agents used for immunomodulation, such as immunomodulators, immunosuppressive agents, tolerogens, and immunostimulants are contemplated by the methods herein. In addition, therapeutic agents acting on the blood and the blood-forming organs, hematopoietic agents, growth factors, minerals, and vitamins, anticoagulant, thrombolytic, and antiplatelet drugs.
  • Further therapeutic agents that can be combined with a subject compound may be found in Goodman and Gilman's “The Pharmacological Basis of Therapeutics” Tenth Edition edited by Hardman, Limbird and Gilman or the Physician's Desk Reference, both of which are incorporated herein by reference in their entirety.
  • The examples and preparations provided below further illustrate and exemplify the compounds of the present invention and methods of preparing such compounds. It is to be understood that the scope of the present invention is not limited in any way by the scope of the following examples and preparations. In the following examples molecules with a single chiral center, unless otherwise noted, exist as a racemic mixture. Those molecules with two or more chiral centers, unless otherwise noted, exist as a racemic mixture of diastereomers. Single enantiomers/diastereomers may be obtained by methods known to those skilled in the art.
  • EXAMPLES Example 1 Expression and Inhibition Assays of p110α/p85α, p110β/p85α, p110δ/p85α, and p110γ
  • Commercial kits or systems for assaying PI3-K activities are available. The commercially available kits or systems can be used to screen for inhibitors and/or agonists of PI3Ks including but not limited to PI3-Kinase α, β, δ, and γ. An exemplary system is PI 3-Kinase (human) HTRF™ Assay from Upstate. The assay can be carried out according to the procedures suggested by the manufacturer. Briefly, the assay is a time resolved FRET assay that indirectly measures PIP3 product formed by the activity of a PI3-K. The kinase reaction is performed in a microtitre plate (e.g., a 384 well microtitre plate). The total reaction volume is approximately 20 ul per well. In the first step, each well receives 2 ul of test compound in 20% dimethylsulphoxide resulting in a 2% DMSO final concentration. Next, approximately 14.5 ul of a kinase/PIP2 mixture (diluted in 1× reaction buffer) is added per well for a final concentration of 0.25-0.3 ug/ml kinase and 10 μM PIP2. The plate is sealed and incubated for 15 minutes at room temperature. To start the reaction, 3.5 ul of ATP (diluted in 1× reaction buffer) is added per well for a final concentration of 10 μM ATP. The plate is sealed and incubated for 1 hour at room temperature. The reaction is stopped by adding 5 ul of Stop Solution per well and then 5 ul of Detection Mix is added per well. The plate is sealed, incubated for 1 hour at room temperature, and then read on an appropriate plate reader. Data is analyzed and IC50 is are generated using GraphPad Prism 5. For PI3K α, β, δ, and γ, the nM concentration of inhibitor to reach IC50 is provided. Inhibition of PI3K a at lower concentrations than those for 13, 6, and γ provides evidence of specificity within this group of kinases. Similar assays, and others known in the art, can be used to measure the percent inhibition of other kinases, including but not limited to PI3K class II kinases, phosphoinositide 4 kinases (PI4K), and phosphoinositide 5 kinases (PI5K).
  • Example 2 Expression and Inhibition Assays of Abl
  • The cross-activity or lack thereof of one or more compounds of the present invention against Abl kinase can be measured according to any procedures known in the art or methods disclosed below. For example, the compounds described herein can be assayed in triplicate against recombinant full-length Abl or Abl (T315I) (Upstate) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 200 μM ATP (2.5 μCi of γ-32P-ATP), and 0.5 mg/mL BSA. The optimized Abl peptide substrate EAIYAAPFAKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 3 Expression and Inhibition Assays of Hck
  • The cross-activity or lack thereof of one or more compounds of the present invention against Hck kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be assayed in triplicate against recombinant full-length Hck in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 200 μM ATP (2.5 μCi of γ-32P-ATP), and 0.5 mg/mL BSA. The optimized Src family kinase peptide substrate EIYGEFKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 4 Expression and Inhibition Assays of Insulin Receptor (IR)
  • The cross-activity or lack thereof of one or more compounds of the present invention against IR receptor kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be assayed in triplicate against recombinant insulin receptor kinase domain (Upstate) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 10 mM MnCl2, 200 μM ATP (2.5 μCi of γ-32P-ATP), and 0.5 mg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with 1M NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 5 Expression and Inhibition Assays of Src
  • The cross-activity or lack thereof of one or more compounds of the present invention against Src kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be assayed in triplicate against recombinant full-length Src or Src (T338I) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 200 μM ATP (2.5 μCi of γ-32P-ATP), and 0.5 mg/mL BSA. The optimized Src family kinase peptide substrate EIYGEFKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 6 Expression and Inhibition Assays of DNA-PK (DNAK)
  • The cross-activity or lack thereof of one or more compounds of the present invention against DNAK kinase can be measured according to any procedures known in the art. DNA-PK can be purchased from Promega and assayed using the DNA-PK Assay System (Promega) according to the manufacturer's instructions.
  • Example 7 Expression and Inhibition Assays of mTOR
  • The cross-activity or lack thereof of one or more compounds of the present invention against mTor can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant mTOR (Invitrogen) in an assay containing 50 mM HEPES, pH 7.5, 1 mM EGTA, 10 mM MgCl2, 2.5 mM, 0.01% Tween, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. Rat recombinant PHAS-1/4EBP1 (Calbiochem; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with 1M NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Other kits or systems for assaying mTOR activity are commercially available. For instance, one can use Invitrogen's LanthaScreen™ Kinase assay to test the inhibitors of mTOR disclosed herein. This assay is a time resolved FRET platform that measures the phosphorylation of GFP labeled 4EBP1 by mTOR kinase. The kinase reaction is performed in a white 384 well microtitre plate. The total reaction volume is 20 ul per well and the reaction buffer composition is 50 mM HEPES pH7.5, 0.01 % Polysorbate 20, 1 mM EGTA, 10 mM MnCl2, and 2 mM DTT. In the first step, each well receives 2 ul of test compound in 20% dimethylsulphoxide resulting in a 2% DMSO final concentration. Next, 8 ul of mTOR diluted in reaction buffer is added per well for a 60 ng/ml final concentration. To start the reaction, 10 ul of an ATP/GFP-4EBP1 mixture (diluted in reaction buffer) is added per well for a final concentration of 10 μM ATP and 0.5 μM GFP-4EBP1. The plate is sealed and incubated for 1 hour at room temperature. The reaction is stopped by adding 10 ul per well of a Tb-anti-pT46 4EBP1 antibody/EDTA mixture (diluted in TR-FRET buffer) for a final concentration of 1.3 nM antibody and 6.7 mM EDTA. The plate is sealed, incubated for 1 hour at room temperature, and then read on a plate reader set up for LanthaScreen™ TR-FRET. Data is analyzed and IC50 is are generated using GraphPad Prism 5.
  • Example 8 Expression and Inhibition Assays of Vascular Endothelial Growth Receptor
  • The cross-activity or lack thereof of one or more compounds of the present invention against VEGF receptor can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant KDR receptor kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 0.1% BME, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with 1M NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 9 Expression and Inhibition Assays of Ephrin Receptor B4 (EphB4)
  • The cross-activity or lack thereof of one or more compounds of the present invention against EphB4 can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant Ephrin receptor B4 kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 0.1% BME, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with 1M NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 10 Expression and Inhibition Assays of Epidermal Growth Factor Receptor (EGFR)
  • The cross-activity or lack thereof of one or more compounds of the present invention against EGFR kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant EGF receptor kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 0.1% BME, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with 1M NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 11 Expression and Inhibition Assays of KIT Assay
  • The cross-activity or lack thereof of one or more compounds of the present invention against KIT kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant KIT kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 1 mM DTT, 10 mM MnCl2, 10 ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with 1M NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 12 Expression and Inhibition Assays of RET
  • The cross-activity or lack thereof of one or more compounds of the present invention against RET kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant RET kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 2.5 mM DTT, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. The optimized Abl peptide substrate EAIYAAPFAKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 13 Expression and Inhibition Assays of Platelet Derived Growth Factor Receptor (PDGFR)
  • The cross-activity or lack thereof of one or more compounds of the present invention against PDGFR kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant PDG receptor kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 2.5 mM DTT, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. The optimized Abl peptide substrate EAIYAAPFAKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 14 Expression and Inhibition Assays of FMS-Related Tyrosine Kinase 3 (FLT-3)
  • The cross-activity or lack thereof of one or more compounds of the present invention against FLT-3 kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant FLT-3 kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 2.5 mM DTT, 10 μM ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. The optimized Abl peptide substrate EAIYAAPFAKKK is used as phosphoacceptor (200 μM). Reactions are terminated by spotting onto phosphocellulose sheets, which are washed with 0.5% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 15 Expression and Inhibition Assays of TEK Receptor Tyrosine Kinase (TIE2)
  • The cross-activity or lack thereof of one or more compounds of the present invention against TIE2 kinase can be measured according to any procedures known in the art or methods disclosed below. The compounds described herein can be tested against recombinant TIE2 kinase domain (Invitrogen) in an assay containing 25 mM HEPES, pH 7.4, 10 mM MgCl2, 2 mM DTT, 10 mM MnCl2, 10 ATP (2.5 μCi of μ-32P-ATP), and 3 mg/mL BSA. Poly E-Y (Sigma; 2 mg/mL) is used as a substrate. Reactions are terminated by spotting onto nitrocellulose, which is washed with 1M NaCl/1% phosphoric acid (approximately 6 times, 5-10 minutes each). Sheets are dried and the transferred radioactivity quantitated by phosphorimaging.
  • Example 16 B Cell Activation and Proliferation Assay
  • The ability of one or more subject compounds to inhibit B cell activation and proliferation is determined according to standard procedures known in the art. For example, an in vitro cellular proliferation assay is established that measures the metabolic activity of live cells. The assay is performed in a 96 well microtiter plate using Alamar Blue reduction. Balb/c splenic B cells are purified over a Ficoll-Paque™ PLUS gradient followed by magnetic cell separation using a MACS B cell Isolation Kit (Miletenyi). Cells are plated in 90 ul at 50,000 cells/well in B Cell Media (RPMI+10% FBS+Penn/Strep+50 μM bME+5 mM HEPES). A compound disclosed herein is diluted in B Cell Media and added in a 10 ul volume. Plates are incubated for 30 min at 37 C and 5% CO2 (0.2% DMSO final concentration). A 50 ul B cell stimulation cocktail is then added containing either 10 ug/ml LPS or 5 ug/ml F(ab′)2 Donkey anti-mouse IgM plus 2 ng/ml recombinant mouse IL4 in B Cell Media. Plates are incubated for 72 hours at 37° C. and 5% CO2. A volume of 15 mL of Alamar Blue reagent is added to each well and plates are incubated for 5 hours at 37 C and 5% CO2. Alamar Blue fluoresce is read at 560Ex/590Em, and IC50 or EC50 values are calculated using GraphPad Prism 5.
  • Example 17 Tumor Cell Line Proliferation Assay
  • The ability of one or more subject compounds to inhibit tumor cell line proliferation is determined according to standard procedures known in the art. For instance, an in vitro cellular proliferation assay can be performed to measure the metabolic activity of live cells. The assay is performed in a 96 well microtiter plate using Alamar Blue reduction. Human tumor cell lines are obtained from ATCC (e.g., MCF7, U-87 MG, MDA-MB-468, PC-3), grown to confluency in T75 flasks, trypsinized with 0.25% trypsin, washed one time with Tumor Cell Media (DMEM+10% FBS), and plated in 90 ul at 5,000 cells/well in Tumor Cell Media. A compound disclosed herein is diluted in Tumor Cell Media and added in a 10 ul volume. Plates are incubated for 72 hours at 37 C and 5% CO2. A volume of 10 uL of Alamar Blue reagent is added to each well and plates are incubated for 3 hours at 37 C and 5% CO2. Alamar Blue fluoresce is read at 560Ex/590Em, and IC50 values are calculated using GraphPad Prism 5. The results are expected to show that some of the compounds of the present invention are potent inhibitors of tumor cell line proliferation under the conditions tested.
  • Example 18 Antitumor Activity In Vivo
  • The compounds described herein can be evaluated in a panel of human and murine tumor models.
  • Paclitaxel-Refractory Tumor Models
  • 1. Clinically-derived Ovarian Carcinoma Model.
  • This tumor model is established from a tumor biopsy of an ovarian cancer patient. Tumor biopsy is taken from the patient.
  • The compounds described herein are administered to nude mice bearing staged tumors using an every 2 days×5 schedule.
  • 2. A2780Tax Human Ovarian Carcinoma Xenograft (Mutated Tubulin).
  • A2780Tax is a paclitaxel-resistant human ovarian carcinoma model. It is derived from the sensitive parent A2780 line by co-incubation of cells with paclitaxel and verapamil, an MDR-reversal agent. Its resistance mechanism has been shown to be non-MDR related and is attributed to a mutation in the gene encoding the beta-tubulin protein.
  • The compounds described herein can be administered to mice bearing staged tumors on an every 2 days×5 schedule.
  • 3. HCT116/VM46 Human Colon Carcinoma Xenograft (Multi-Drug Resistant).
  • HCT116/VM46 is an MDR-resistant colon carcinoma developed from the sensitive HCT116 parent line. In vivo, grown in nude mice, HCT116NM46 has consistently demonstrated high resistance to paclitaxel.
  • The compounds described herein can be administered to mice bearing staged tumors on an every 2 days×5 schedule.
  • 5. M5076 Murine Sarcoma Model
  • M5076 is a mouse fibrosarcoma that is inherently refractory to paclitaxel in vivo.
  • The compounds described herein can be administered to mice bearing staged tumors on an every 2 days×5 schedule.
  • One or more compounds of the invention can be used in combination other therapeutic agents in vivo in the multidrug resistant human colon carcinoma xenografts HCT/VM46 or any other model known in the art including those described herein.
  • The results are expected to show that one or more compounds of the present invention are potent inhibitors of tumor growth in vivo under the conditions tested.
  • Example 19 Microsome Stability Assay
  • The stability of one or more subject compounds is determined according to standard procedures known in the art. For example, stability of one or more subject compounds is established by an in vitro assay. In particular, an in vitro microsome stability assay is established that measures stability of one or more subject compounds when reacting with mouse, rat or human microsomes from liver. The microsome reaction with compounds is performed in 1.5 mL Eppendorf tube. Each tube contains 0.1 μL of 10.0 mg/ml NADPH; 75 μL of 20.0 mg/ml mouse, rat or human liver microsome; 0.4 μL of 0.2 M phosphate buffer, and 425 μL of ddH2O. Negative control (without NADPH) tube contains 75 μL of 20.0 mg/ml mouse, rat or human liver microsome; 0.4 μL of 0.2 M phosphate buffer, and 525 μL of ddH2O. The reaction is started by adding 1.0 μL of 10.0 mM tested compound. The reaction tubes are incubated at 37° C. 100 μL sample is collected into new Eppendorf tube containing 300 μL cold Methanol at 0, 5, 10, 15, 30 and 60 minutes of reaction. Samples are centrifuged at 15,000 rpm to remove protein. Supernatant of centrifuged sample is transferred to new tube. Concentration of stable compound after reaction with microsome in the supernatant is measured by Liquid Chromatography/Mass Spectrometry (LC-MS).
  • Example 20 Plasma Stability Assay
  • The stability of one or more subject compounds in plasma is determined according to standard procedures known in the art. See, e.g., Rapid Commun. Mass Spectrom., 10: 1019-1026. The following procedure is an HPLC-MS/MS assay using human plasma; other species including monkey, dog, rat, and mouse are also available. Frozen, heparinized human plasma is thawed in a cold water bath and spun for 10 minutes at 2000 rpm at 4° C. prior to use. A subject compound is added from a 400 μM stock solution to an aliquot of pre-warmed plasma to give a final assay volume of 400 μL (or 800 μL for half-life determination), containing 5 μM test compound and 0.5% DMSO. Reactions are incubated, with shaking, for 0 minutes and 60 minutes at 37° C., or for 0, 15, 30, 45 and 60 minutes at 37 C for half life determination. Reactions are stopped by transferring 50 μL of the incubation mixture to 200 μL of ice-cold acetonitrile and mixed by shaking for 5 minutes. The samples are centrifuged at 6000×g for 15 minutes at 4° C. and 120 μL of supernatant removed into clean tubes. The samples are then evaporated to dryness and submitted for analysis by HPLC-MS/MS.
  • Where desired, one or more control or reference compounds (5 μM) are tested simultaneously with the test compounds: one compound, propoxycaine, with low plasma stability and another compound, propantheline, with intermediate plasma stability.
  • Samples are reconstituted in acetonitrile/methanol/water (1/1/2, v/v/v) and analyzed via (RP)HPLC-MS/MS using selected reaction monitoring (SRM). The HPLC conditions consist of a binary LC pump with autosampler, a mixed-mode, C12, 2×20 mm column, and a gradient program. Peak areas corresponding to the analytes are recorded by HPLC-MS/MS. The ratio of the parent compound remaining after 60 minutes relative to the amount remaining at time zero, expressed as percent, is reported as plasma stability. In case of half-life determination, the half-life is estimated from the slope of the initial linear range of the logarithmic curve of compound remaining (%) vs. time, assuming first order kinetics.
  • Example 21 Chemical Stability
  • The chemical stability of one or more subject compounds is determined according to standard procedures known in the art. The following details an exemplary procedure for ascertaining chemical stability of a subject compound. The default buffer used for the chemical stability assay is phosphate-buffered saline (PBS) at pH 7.4; other suitable buffers can be used. A subject compound is added from a 100 μM stock solution to an aliquot of PBS (in duplicate) to give a final assay volume of 400 μL, containing 5 μM test compound and 1% DMSO (for half-life determination a total sample volume of 700 μL is prepared). Reactions are incubated, with shaking, for 0 minutes and 24 hours at 37° C.; for half-life determination samples are incubated for 0, 2, 4, 6, and 24 hours. Reactions are stopped by adding immediately 100 μL of the incubation mixture to 100 μL of acetonitrile and vortexing for 5 minutes. The samples are then stored at −20° C. until analysis by HPLC-MS/MS. Where desired, a control compound or a reference compound such as chlorambucil (5 μM) is tested simultaneously with a subject compound of interest, as this compound is largely hydrolyzed over the course of 24 hours. Samples are analyzed via (RP)HPLC-MS/MS using selected reaction monitoring (SRM). The HPLC conditions consist of a binary LC pump with autosampler, a mixed-mode, C12, 2×20 mm column, and a gradient program. Peak areas corresponding to the analytes are recorded by HPLC-MS/MS. The ratio of the parent compound remaining after 24 hours relative to the amount remaining at time zero, expressed as percent, is reported as chemical stability. In case of half-life determination, the half-life is estimated from the slope of the initial linear range of the logarithmic curve of compound remaining (%) vs. time, assuming first order kinetics.
  • Example 22 Akt Kinase Assay
  • Cells comprising components of the Akt/mTOR pathway, including but not limited to L6 myoblasts, B-ALL cells, B-cells, T-cells, leukemia cells, bone marrow cells, p190 transduced cells, Philadelphia chromosome positive cells (Ph+), and mouse embryonic fibroblasts, are typically grown in cell growth media such as DMEM supplemented with fetal bovine serum and/or antibiotics, and grown to confluency.
  • In order to compare the effect of one or more compounds disclosed herein on Akt activation, said cells are serum starved overnight and incubated with one or more compounds disclosed herein or about 0.1% DMSO for approximately 1 minute to about 1 hour prior to stimulation with insulin (e.g., 100 nM) for about 1 minutes to about 1 hour. Cells are lysed by scraping into ice cold lysis buffer containing detergents such as sodium dodecyl sulfate and protease inhibitors (e.g., PMSF). After contacting cells with lysis buffer, the solution is briefly sonicated, cleared by centrifugation, resolved by SDS-PAGE, transferred to nitrocellulose or PVDF and immunoblotted using antibodies to phospho-Akt S473, phospho-Akt T308, Akt, and β-actin (Cell Signaling Technologies).
  • Example 23 Kinase Signaling in Blood
  • PI3K/Akt/mTor signaling is measured in blood cells using the phosflow method (Methods Enzymol. 2007; 434:131-54). The advantage of this method is that it is by nature a single cell assay so that cellular heterogeneity can be detected rather than population averages. This allows concurrent distinction of signaling states in different populations defined by other markers. Phosflow is also highly quantitative. To test the effects of one or more compounds disclosed herein, unfractionated splenocytes, or peripheral blood mononuclear cells are stimulated with anti-CD3 to initiate T-cell receptor signaling. The cells are then fixed and stained for surface markers and intracellular phosphoproteins. It is expected that inhibitors disclosed herein inhibit anti-CD3 mediated phosphorylation of Akt-S473 and S6, whereas rapamycin inhibits S6 phosphorylation and enhances Akt phosphorylation under the conditions tested.
  • Similarly, aliquots of whole blood are incubated for 15 minutes with vehicle (e.g., 0.1% DMSO) or kinase inhibitors at various concentrations, before addition of stimuli to crosslink the T cell receptor (TCR) (anti-CD3 with secondary antibody) or the B cell receptor (BCR) using anti-kappa light chain antibody (Fab′2 fragments). After approximately 5 and 15 minutes, samples are fixed (e.g., with cold 4% paraformaldehyde) and used for phosflow. Surface staining is used to distinguish T and B cells using antibodies directed to cell surface markers that are known to the art. The level of phosphrylation of kinase substrates such as Akt and S6 are then measured by incubating the fixed cells with labeled antibodies specific to the phosphorylated isoforms of these proteins. The population of cells is then analyzed by flow cytometry.
  • The results are expected to show that one or more of the compounds of the present invention are potent and selective inhibitors of one or more members of one or more of PI3K, mTOR, and Akt signaling in blood cells under the conditions tested.
  • Example 24 Colony Formation Assay
  • Murine bone marrow cells freshly transformed with a p190 BCR-Abl retrovirus (herein referred to as p190 transduced cells) are plated in the presence of various drug combinations in M3630 methylcellulose media for about 7 days with recombinant human IL-7 in about 30% serum, and the number of colonies formed is counted by visual examination under a microscope. Alternatively, human peripheral blood mononuclear cells are obtained from Philadelphia chromosome positive (Ph+) and negative (Ph−) patients upon initial diagnosis or relapse. Live cells are isolated and enriched for CD19+CD34+ B cell progenitors. After overnight liquid culture, cells are plated in methocult GF+H4435, Stem Cell Technologies) supplemented with cytokines (IL-3, IL-6, IL-7, G-CSF, GM-CSF, CF, Flt3 ligand, and erythropoietin) and various concentrations of known chemotherapeutic agents in combination with either compounds of the present disclosure. Colonies are counted by microscopy 12-14 days later. This method can be used to test for evidence of additive or synergistic activity.
  • The results are expected to show that one or more the compounds of the present invention are potent and selective inhibitors of p190 transduced cell colony formation under the conditions tested.
  • Example 25 In Vivo Effect of Kinase Inhibitors on Leukemic Cells
  • Female recipient mice are lethally irradiated from a γ source in two doses about 4 hr apart, with approximately 5 Gy each. About 1 hr after the second radiation dose, mice are injected i.v. with about 1×106 leukemic cells (e.g., Ph+ human or murine cells, or p190 transduced bone marrow cells). These cells are administered together with a radioprotective dose of about 5×106 normal bone marrow cells from 3-5 week old donor mice. Recipients are given antibiotics in the water and monitored daily. Mice who become sick after about 14 days are euthanized and lymphoid organs are harvested for analysis. Kinase inhibitor treatment begins about ten days after leukemic cell injection and continues daily until the mice become sick or a maximum of approximately 35 days post-transplant Inhibitors are given by oral lavage.
  • Peripheral blood cells are collected approximately on day 10 (pre-treatment) and upon euthanization (post treatment), contacted with labeled anti-hCD4 antibodies and counted by flow cytometry. This method can be used to demonstrate that the synergistic effect of one or more compounds disclosed herein in combination with known chemotherapeutic agents significantly reduce leukemic blood cell counts as compared to treatment with known chemotherapeutic agents (e.g., Gleevec) alone under the conditions tested.
  • Example 26 Treatment of Lupus Disease Model Mice
  • Mice lacking the inhibitory receptor FcγRIIb that opposes PI3K signaling in B cells develop lupus with high penetrance. FcγRIIb knockout mice (R2KO, Jackson Labs) are considered a valid model of the human disease as some lupus patients show decreased expression or function of FcγRIIb (S. Bolland and J. V. Ravtech 2000. Immunity 12:277-285).
  • The R2KO mice develop lupus-like disease with anti-nuclear antibodies, glomerulonephritis and proteinuria within about 4-6 months of age. For these experiments, the rapamycin analogue RAD001 (available from LC Laboratories) is used as a benchmark compound, and administered orally. This compound has been shown to ameliorate lupus symptoms in the B6.Sle1z.Sle3z model (T. Wu et al. J. Clin Invest. 117:2186-2196).
  • Lupus disease model mice such as R2KO, BXSB or MLR/lpr are treated at about 2 months old, approximately for about two months. Mice are given doses of: vehicle, RAD001 at about 10 mg/kg, or compounds disclosed herein at approximately 1 mg/kg to about 500 mg/kg. Blood and urine samples are obtained at approximately throughout the testing period, and tested for antinuclear antibodies (in dilutions of serum) or protein concentration (in urine). Serum is also tested for anti-ssDNA and anti-dsDNA antibodies by ELISA. Animals are euthanized at day 60 and tissues harvested for measuring spleen weight and kidney disease. Glomerulonephritis is assessed in kidney sections stained with H&E. Other animals are studied for about two months after cessation of treatment, using the same endpoints.
  • This model established in the art can be employed to test that the kinase inhibitors disclosed herein can suppress or delay the onset of lupus symptoms in lupus disease model mice.
  • Example 27 Murine Bone Marrow Transplant Assay
  • Female recipient mice are lethally irradiated from a γ ray source. About 1 hr after the radiation dose, mice are injected with about 1×106 leukemic cells from early passage p190 transduced cultures (e.g., as described in Cancer Genet Cytogenet. 2005 August; 161(1):51-6). These cells are administered together with a radioprotective dose of approximately 5×106 normal bone marrow cells from 3-5 wk old donor mice. Recipients are given antibiotics in the water and monitored daily. Mice who become sick after about 14 days are euthanized and lymphoid organs harvested for flow cytometry and/or magnetic enrichment. Treatment begins on approximately day 10 and continues daily until mice become sick, or after a maximum of about 35 days post-transplant. Drugs are given by oral gavage (p.o.). In a pilot experiment a dose of chemotherapeutic that is not curative but delays leukemia onset by about one week or less is identified; controls are vehicle-treated or treated with chemotherapeutic agent, previously shown to delay but not cure leukemogenesis in this model (e.g., imatinib at about 70 mg/kg twice daily). For the first phase p190 cells that express eGFP are used, and postmortem analysis is limited to enumeration of the percentage of leukemic cells in bone marrow, spleen and lymph node (LN) by flow cytometry. In the second phase, p190 cells that express a tailless form of human CD4 are used and the postmortem analysis includes magnetic sorting of hCD4+ cells from spleen followed by immunoblot analysis of key signaling endpoints: p Akt-T308 and S473; pS6 and p4EBP-1. As controls for immunoblot detection, sorted cells are incubated in the presence or absence of kinase inhibitors of the present disclosure inhibitors before lysis. Optionally, “phosflow” is used to detect p Akt-S473 and pS6-S235/236 in hCD4-gated cells without prior sorting. These signaling studies are particularly useful if, for example, drug-treated mice have not developed clinical leukemia at the 35 day time point. Kaplan-Meier plots of survival are generated and statistical analysis done according to methods known in the art. Results from p190 cells are analyzed separated as well as cumulatively.
  • Samples of peripheral blood (100-200 μl) are obtained weekly from all mice, starting on day 10 immediately prior to commencing treatment. Plasma is used for measuring drug concentrations, and cells are analyzed for leukemia markers (eGFP or hCD4) and signaling biomarkers as described herein.
  • This general assay known in the art may be used to test that effective therapeutic doses of the compounds disclosed herein can be used for inhibiting the proliferation of leukemic cells.
  • Example 28 Rat Developing Type II Collagen Induced Arthritis Assay
  • In order to study the effects of the compounds of the present invention on the autoimmune disease arthritis, a collagen induced developing arthritis model is used. Female Lewis rats are given collagen injections at day 0. Bovine type II collagen is prepared as a 4 mg/ml solution in 0.01N acetic acid. Equal volumes of collagen and Freund's incomplete adjuvant are emulsified by hand mixing until a bead of the emulsified material holds its form in water. Each rodent receives a 300 μl injection of the mixture at each injection time spread over three subcutaneous sites on the back.
  • Oral compound administration begins on day 0 and continues through day 16 with vehicle (5% NMP, 85 % PEG 400, 10% Solutol) or compounds of the present invention in vehicle or control (e.g., methotrexate) at 12 hour intervals daily. Rats are weighed on days 0, 3, 6, 9-17 and caliper measurements of ankles are taken on days 9-17. Final body weights are taken, and then the animals are euthanized on day 17. After euthanization, blood is drawn and hind paws and knees are removed. Blood is further processed for pharmacokinetics experiments as well as an anti-type II collagen antibody ELISA assay. Hind paws are weighed and then, with the knees, preserved in 10% formalin. The paws and knees are subsequently processed for microscopy. Livers, spleen and thymus are weighed. Sciatic nerves are prepared for histopathology.
  • Knee and ankle joints are fixed for 1-2 days and decalcified for 4-5 days. Ankle joints are cut in half longitudinally, and knees are cut in half along the frontal plane. Joints are processed, embedded, sectioned and stained with toluidine blue. Scoring of the joints is done according to the following criteria:
  • Knee and Ankle Inflammation 0=Normal
  • 1=Minimal infiltration of inflammatory cells in synovium/periarticular tissue
    2=Mild infiltration
    3=Moderate infiltration with moderate edema
    4=Marked infiltration with marked edema
    5=Severe infiltration with severe edema
  • Ankle Pannus 0=Normal
  • 1=Minimal infiltration of pannus in cartilage and subchondral bone
    2=Mild infiltration (<¼ of tibia or tarsals at marginal zones)
    3=Moderate infiltration (¼ to ⅓ of tibia or small tarsals affected at marginal zones)
    4=Marked infiltration (½-¾ of tibia or tarsals affected at marginal zones)
    5=Severe infiltration (>¾ of tibia or tarsals affected at marginal zones, severe distortion of overall architecture)
  • Knee Pannus 0=Normal
  • 1=Minimal infiltration of pannus in cartilage and subchondral bone
    2=Mild infiltration (extends over up tot/4 of surface or subchondral area of tibia or femur)
    3=Moderate infiltration (extends over >¼ but <½ of surface or subchondral area of tibia or femur)
    4=Marked infiltration (extends over ½ to ¾ of tibial or femoral surface)
    5=Severe infiltration (covers >¾ of surface)
  • Cartilage Damage (Ankle, Emphasis on Small Tarsals) 0=Normal
  • 1=Minimal=minimal to mild loss of toluidine blue staining with no obvious chondrocyte loss or collagen disruption
    2=Mild=mild loss of toluidine blue staining with focal mild (superficial) chondrocyte loss and/or collagen disruption
    3=Moderate=moderate loss of toluidine blue staining with multifocal moderate (depth to middle zone) chondrocyte loss and/or collagen disruption, smaller tarsals affected to ½-¾ depth
    4=Marked=marked loss of toluidine blue staining with multifocal marked (depth to deep zone) chondrocyte loss and/or collagen disruption, 1 or more small tarsals have full thickness loss of cartilage
    5=Severe=severe diffuse loss of toluidine blue staining with multifocal severe (depth to tide mark) chondrocyte loss and/or collagen disruption
  • Cartilage Damage (Knee, Emphasis on Femoral Condyles) 0=Normal
  • 1=Minimal=minimal to mild loss of toluidine blue staining with no obvious chondrocyte loss or collagen disruption
    2=Mild=mild loss of toluidine blue staining with focal mild (superficial) chondrocyte loss and/or collagen disruption
    3=Moderate=moderate loss of toluidine blue staining with multifocal to diffuse moderate (depth to middle zone) chondrocyte loss and/or collagen disruption
    4=Marked=marked loss of toluidine blue staining with multifocal to diffuse marked (depth to deep zone) chondrocyte loss and/or collagen disruption or single femoral surface with total or near total loss
    5=Severe=severe diffuse loss of toluidine blue staining with multifocal severe (depth to tide mark) chondrocyte loss and/or collagen disruption on both femurs and/or tibias Bone Resorption (Ankle)
  • 0=Normal
  • 1=Minimal=small areas of resorption, not readily apparent on low magnification, rare osteoclasts
    2=Mild=more numerous areas of resorption, not readily apparent on low magnification, osteoclasts more numerous, <¼ of tibia or tarsals at marginal zones resorbed
    3=Moderate=obvious resorption of medullary trabecular and cortical bone without full thickness defects in cortex, loss of some medullary trabeculae, lesion apparent on low magnification, osteoclasts more numerous, ¼ to ⅓ of tibia or tarsals affected at marginal zones
    4=Marked=Full thickness defects in cortical bone, often with distortion of profile of remaining cortical surface, marked loss of medullary bone, numerous osteoclasts, ½-¾ of tibia or tarsals affected at marginal zones
    5=Severe=Full thickness defects in cortical bone, often with distortion of profile of remaining cortical surface, marked loss of medullary bone, numerous osteoclasts, >¾ of tibia or tarsals affected at marginal zones, severe distortion of overall architecture
  • Bone Resorption (Knee) 0=Normal
  • 1=Minimal=small areas of resorption, not readily apparent on low magnification, rare osteoclasts
    2=Mild=more numerous areas of resorption, definite loss of subchondral bone involving ¼ of tibial or femoral surface (medial or lateral)
    3=Moderate=obvious resorption of subchondral bone involving >¼ but <½ of tibial or femoral surface (medial or lateral)
    4=Marked=obvious resorption of subchondral bone involving >½ but <¾ of tibial or femoral surface (medial or lateral)
    5=Severe=distortion of entire joint due to destruction involving >¾ of tibial or femoral surface (medial or lateral)
    Statistical analysis of body/paw weights, paw AUC parameters and histopathologic parameters were evaluated using a Student's t-test or other appropriate (ANOVA with post-test) with significance set at the 5% significance level. Percent inhibition of paw weight and AUC was calculated using the following formula:

  • % Inhibition=A−B/A×100
  • A=Mean Disease Control—Mean Normal B=Mean Treated—Mean Normal
  • The results are expected to show, relative to vehicle only control or to methotrexate control, that the compounds of the present invention exhibit a significant reduction in arthritis induced ankle diameter increase over time, and reduction of ankle histopathology in at least one or more of the categories of inflammation, pannus, cartilage damage, and bone resporption as described above. The results are expected to show that one or more compounds of the present invention may be useful for the treatment and reduction of arthritis disease symptoms.
  • The results further are expected to show a reduction at 10, 20, and 60 mg/kg dosage levels of serum anti-type II collagen levels for selected test compounds, suggesting that one or more compounds of the present invention may not only be useful for the treatment and reduction of arthritis disease symptoms, but may also be useful for the inhibition of the autoimmune reaction itself.
  • Example 29 Rat Established Type II Collagen Induced Arthritis Assay
  • In order to examine the dose responsive efficacy of the compounds of the present invention in inhibiting the inflammation, cartilage destruction and bone resorption of 10 day established type II collagen induced arthritis in rats, compounds are administered orally daily or twice daily for 6 days.
  • Female Lewis rats are anesthetized and given collagen injections prepared and administered as described previously on day 0. On day 6, animals are anesthetized and given a second collagen injection. Caliper measurements of normal (pre-disease) right and left ankle joints are performed on day 9. On days 10-11, arthritis typically occurs and rats are randomized into treatment groups. Randomization is performed after ankle joint swelling is obviously established and there is evidence of bilateral disease.
  • After an animal is selected for enrollment in the study, treatment is initiated by the oral route. Animals are given vehicle, control (Enbrel) or compound doses, twice daily or once daily (BID or QD respectively). Administration is performed on days 1-6 using a volume of 2.5 ml/kg (BID) or 5 ml/kg (QD) for oral solutions. Rats are weighed on days 1-7 following establishment of arthritis and caliper measurements of ankles taken every day. Final body weights are taken on day 7 and animals are euthanized.
  • The results are expected to show reduction in mean ankle diameter increase over time for selected test compounds under the conditions tested.
  • Example 30 Adjuvant Induced Arthritis Assay
  • Intrathecal Catheterization of Rats
  • Isoflurane-anesthetized Lewis rats (200-250 g) are implanted with an intrathecal (IT) catheter. After a 6 d recovery period, all animals except those that appeared to have sensory or motor abnormalities (generally fewer than 5% of the total number) are used for experiments. For IT administration, 10 μl of drug or saline followed by 10 μl of isotonic saline is injected through the catheter.
  • Adjuvant Arthritis and Drug Treatment
  • Lewis rats are immunized at the base of the tail with 0.1 ml of complete Freund's adjuvant (CFA) on day 0 several days after catheter implantation (n=6/group). Drug (e.g., one or more compounds of the present invention or vehicle) treatment is generally started on day 8 and is continued daily until day 20. Clinical signs of arthritis generally begin on day 10, and paw swelling is determined every second day by water displacement plethysmometry.
  • The results are expected to show that one or more compounds of the present invention may be useful for the treatment of one or more of the diseases or conditions described herein.
  • Example 31 Rodent Pharmacokinetic Assay
  • In order to study the pharmacokinetics of the compounds of the present invention a set of 4-10 week old mice are grouped according to the following table:
  • Compound Administration
    Group# Mice/group (mg/kg) Route Regimen
    1 3 1 Po One
    2 3 3 dose
    3 3 10
    4 3 30
    5 3 60
  • Compounds of the present invention are dissolved in an appropriate vehicle (e.g., 5% 1-methyl-2-pyrrolidinone, 85 % polyethylene glycol 400, 10% Solutor) and administered orally at 12 hour intervals daily. All animals are euthanized in CO 2 2 hours after the final compound is administered. Blood is collected immediately and kept on ice for plasma isolation. Plasma is isolated by centrifuging at 5000 rpm for 10 minutes. Harvested plasma is frozen for pharmacokinetic detection.
  • The results are expected to demonstrate the pharmacokinetic parameters such as absorption, distribution, metabolism, excretion, and toxicity for the compounds of the present invention.
  • Example 32 Basotest Assay
  • The barotest assay is performed using Orpegen Pharma Basotest reagent kit. Heparinized whole blood is pre-incubated with test compound or solvent at 37 C for 20 min. Blood is then incubated with assay kit stimulation buffer (to prime cells for response) followed by allergen (dust mite extract or grass extract) for 20 min. The degranulation process is stopped by incubating the blood samples on ice. The cells are then labeled with anti-IgE-PE to detect basophilic granulocytes, and anti-gp53-FITC to detect gp53 (a glycoprotein expressed on activated basophils). After staining red blood cells are lysed by addition of Lysing Solution. Cells are washed, and analyzed by flow cytometry. Test compounds, when evaluated in this assay inhibit allergen induced activation of basophilic granulocytes at sub micromolar range. The results are expected to demonstrate that under the conditions tested one or more compounds of the present invention are capable of inhabiting allergen induced activation of basophils.
  • Example 33 Use of the Compounds of the Present Invention for Inhibition of Tumor Growth
  • Cell Lines
  • Cell lines of interest (A549, U87, ZR-75-1 and 786-O) are obtained from American Type Culture Collection (ATCC, Manassas, Va.). Cells are proliferated and preserved cryogenically at early passage (e.g., passage 3). One aliquot is used for further proliferation to get enough cells for one TGI study (at about passage 9).
  • Animals
  • Female athymic nude mice are supplied by Harlan. Mice are received at 4 to 6 weeks of age. All mice are acclimated for about one day to two weeks prior to handling. The mice are housed in microisolator cages and maintained under specific pathogen-free conditions. The mice are fed with irradiated mouse chow and freely available autoclaved water is provided.
  • Tumor Xenograft Model
  • Mice are inoculated subcutaneously in the right flank with 0.01 to 0.5 ml of tumor cells (approximately 1.0×105 to 1.0×108 cells/mouse). Five to 10 days following inoculation, tumors are measured using calipers and tumor weight is calculated, for example using the animal study management software, such as Study Director V.1.6.70 (Study Log). Mice with tumor sizes of about 120 mg are pair-matched into desired groups using Study Director (Day 1). Body weights are recorded when the mice are pair-matched. Tumor volume and bodyweight measurements are taken one to four times weekly and gross observations are made at least once daily. On Day 1, compounds of the present invention and reference compounds as well as vehicle control are administered by oral gavage or iv as indicated. At the last day of the experiment, mice are sacrificed and their tumors are collected 1-4 hours after the final dose. The tumors are excised and cut into two sections. One third of the tumor is fixed in formalin and embedded in paraffin blocks and the remaining two thirds of tumor is snap frozen and stored at −80° C.
  • Data and Statistical Analysis
  • Mean tumor growth inhibition (TGI) is calculated utilizing the following formula:
  • TGI = [ 1 - ( χ _ Treated ( Final ) - χ _ Treated ( Day 1 ) ) ( χ _ Control ( Final ) - χ _ Control ( Day 1 ) ) ] × 100 %
  • Tumors that regress from the Day 1 starting size are removed from the calculations. Individual tumor shrinkage (TS) is calculated using the formula below for tumors that show regression relative to Day 1 tumor weight. The mean tumor shrinkage of each group is calculated and reported.
  • TS = [ 1 - ( Tumor Weight ( Final ) ) ( Tumor Weight ( Day 1 ) ) ] × 100 %
  • The model can be employed to show whether the compounds of the present invention can inhibit tumor cell growth such as renal carcinomoa cell growth, breast cancer cell growth, lung cancer cell growth, or glioblastoma cell growth under the conditions tested.
  • Example 34 Inhibition of PI3K Pathway and Proliferation of Tumor Cells with PI3Kα Mutation
  • Cells comprising one or more mutations in PI3Kα, including but not limited to breast cancer cells (e.g., MDA-MB-361, T47D, SKOV-3), and cells comprising one or more mutations in PTEN including but not limited to prostate cancer cells (e.g., PC3), are typically grown in cell growth media such as DMEM supplemented with fetal bovine serum and/or antibiotics, and grown to confluency. Cells are then treated with various concentrations of test compound for about 2 hours and subsequently lysed in cell lysis buffer. Lysates are subjected to SDS-PAGE followed by Western blot analysis to detect downstream signaling markers, including but not limited to pAKT(S473), pAKT(T308), pS6, and p4E-BP1. Degree of proliferation (and proliferation inhibition) can also be measured for cells at various doses of compound of the present invention such as Compound A (compound 54 of Table 3) or Compound B (compound 1 of Table 1). β-Actin can be used as a housekeeping protein to ascertain proper loading. FIG. 7 shows a Western blot depicting differential inhibition of Akt phosphorylation at serine 473 over threonine 308 by Compound B (top panel) in a PC3 model. Also shown is the comparison of Akt phosphorylation inhibition for Pan-PI3K inhibitor versus Compound B in a SKOV-3 model. FIG. 12 shows a Western blot depicting inhibition of Akt phosphorylation at serine 473 by Compound A in MDA-MB-361cells relative to PC3 cells, indicating that compounds of the invention are preferentially capable of inhibiting tumor proliferation of cells with mutations in the PI3Kα pathway relative to cells with mutations in the PTEN pathway. Compound A inhibited pAKT with an IC50 of 1000 nM for MDA-MB-361 cells vs. an IC50 of ˜10,000 nM for PC3 cells. Similarly, Compound A inhibited cell proliferation with an IC50 of ˜1900 for MDA-MB-361 cells vs. an IC50 of >10,000 nM for PC3 cells.
  • A western blot depicting inhibition of the PI3K pathway by Compound A in cell lines with elevated PI3K α activity is shown in FIG. 11. MDA-MB-361(PIK3CA-E545K), MDA-MB-453 (PIK3CA-H1047R), and SK-Br3 (Her2) cells were treated with various concentrations of Compound A for 2 h and subsequently lysed. Lysates were subjected to SDS-PAGE followed by Western blot analysis as described above to detect downstream pathway markers. The left column shows data from MDA-MB-361 breast cancer cells harboring PIK3CA mutation. The middle column shows data from MDA-MB-453 breast cancer cells harboring PIK3CA mutation. The right column shows data from SKBr3breast cancer cells harboring HER2 mutation.
  • The effect of Compound A on proliferation of tumor cells harboring PI3Kα mutations is shown in FIG. 13. Cells were seeded at 5000-10,000 per well in a 96 well flat bottom plate 90 ul growth media. Pre-diluted compound (at 10 times the final concentration) or DMSO (control) was added. The compounds were prepared such that the dose range extended from a final concentration of 30 μM to 14 nM via 3-fold serial dilutions (8 point curve). The final DMSO concentration was 0.3%. Cells were incubated for 72 hours at 37° C. in a CO2 incubator. After 72 hours, proliferation was measured using CellTiter-Glo Luminescent reagent (Promega, PR-G7573). CellTiter-Glo reagent was added at 100 μL per well which was equal to the volume of culture media present in each well. Contents were mixed for 30 minutes on an orbital shaker to lyse cells Luminescence was read on a PerkinElmer EnVision plate reader. Curves and IC50 values were generated in GraphPad Prism using a sigmoidal dose response non linear regression fit. Inhibition % was calculated as: 1−(cells+inhibitor)−background signal)/(cells+DMSO)−background signal)×100.
  • Example 35 In Vitro Inhibition of Angiogenesis
  • Inhibition of angiogenesis in the presence of test compound is evaluated using a tube formation assay, such as by using a tube formation assay kit (e.g., commerically available from Invitrogen). Angiogenic capacity can be measured in vitro using an endothelial cell line, such as human umbilical vein endothelial cells (HUVEC). The assay is conducted according to the kit instructions, in the presence or absence of compound. Briefly, a gel matrix is applied to a cell culture surface, cells are added to the matrix-covered surface along with growth factors, with some samples also receiving an inhibitor compound, cells are incubated at 37° C. and 5% CO2 long enough for control samples (no compound added) to form tube structures (such as overnight), cells are stained using a cell-permeable dye (e.g., calcein), and cells are visualized to identify the degree of tube formation. Any decrease in tube formation relative to un-inhibited control cells is indicative of angiogenic inhibition. Based on doses tested and the corresponding degree of tube formation inhibition, IC50 values for tube formation are calculated. IC50 values for cell viability can be measured using any number of methods known in the art, such as staining methods that distinguish live from dead cells (e.g., Image-iT DEAD Green viability stain commercially available from Invitrogen.
  • Example 36 In Vivo Efficacy in Xenogenic Mouse Model of Breast Cancer
  • Nude mice harboring tumors (150-250 mm3) derived from implantation of human breast adenocarcinoma cells MDA-MB-361 (PI3Kα/HER2 carcinoma) were separated into untreated control (vehicle only) and treatment groups. Mice in the treatment group were further divided into mice receiving 60 mg/kg (60 mpk) of a PI3Kα inhibitor (Compound A), 0.3 mg/kg of an mTor inhibitor (Compound B), or a combination of Compound A and Compound B. Groups receiving 5 mg/kg of paclitaxel (intravenously), alone or in combination with Compound B, were also included. Mice in the treatment group received the defined dose daily by oral lavage for 30 days, during which time tumor weight was calculated as described above (e.g. every 2-5 days). Blood glucose was monitored periodically following administration of treatment. 2 hours after the final treatment, tumors were harvested and proteins are analyzed by Western blot as described above. The effect of the compounds on the localization/viability of marginal zone B cells in the spleen is also evaluated at the conclusion of treatment. The synergistic effect of combined treatment with Compound A and Compound B on tumor weight in a preclinical breast cancer model are shown in FIG. 2. These results are further supported by FIG. 3, which further shows Western Blot results for levels of various markers for kinase activity.
  • A similar experiment was conducted in which nude mice harboring tumors derived from implantation of human breast adenocarcinoma cells MDA-MB-361 (PI3Kα/HER2 carcinoma) were separated into untreated control (vehicle only) and treatment groups. Mice in the treatment group were then further divided into mice receiving 60 mg/kg (60 mpk) of a PI3Kα inhibitor (Compound A), an mTor inhibitor which is rapamycin (0.5 mg/kg intraperitoneal), or a combination of Compound A and rapamycin. Treatment of tumors (MDA-MB-361) with compounds was discontinued on Day 30. Tumors were measured every 2-5 days. FIG. 4 shows a synergistic effect of combined treatment with Compound A and rapamycin in this preclinical breast cancer model.
  • A similar experiment using 786-0 cells, a human kidney carcinoma cell line having a non-mutated PI3Kα, instead of MDA cells was used to further demonstrate the specificity of test compounds. Compound A was administered orally at 60 and 120 mg/kg and compound A was administered orally as a solution at 1 mg/kg (QD). For example, a PI3Kα inhibitor is compared to a kinase inhibitor with specificity for mTor. The inhibitor with affinity for mTor (Compound B) is effective at inhibiting kinase activity (measured by Western blot analysis of downstream markers) and tumor growth in this model, whereas a PI3Kα inhibitor (Compound A), while still showing some anti-tumor activity, has decreased activity by comparison to that seen in the MDA derived tumors (compare FIG. 2 and FIG. 6). FIG. 6C also provides biochemical potency data for the two compared inhibitors. PI3 Kinase α, β, γ and δ activity was assayed using the PI3 Kinase HTRF® assay kit (Millipore Corporation). Purified recombinant PI3Kα, PI3Kβ, PI3Kγ and PI3Kδ were obtained from Millipore Corporation. Purified recombinant PI3K was used to catalyze the phosphorylation of phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol 3,4,5-trisphosphate (PIP3) in the presence of 10 μM ATP. Emission ratios were converted into percent inhibitions and imported into GraphPad Prism software. The concentration of Compound A or B necessary to achieve inhibition of enzyme activity by 50% (IC50) was calculated using concentrations ranging from 20 μM to 0.1 nM (12-point curve). IC50 values were determined using a nonlinear regression model available in GraphPad Prism 5.
  • FIG. 9 shows a graph depicting reduction in tumor weight for breast cancer model using 70 mg/kg Pan-PI3K inhibitor and 60 mg/kg compound A (left panel) and a reduced presence of MZB cells in mouse spleen for 70 mg/kg Pan-PI3K inhibitor compared to 60 mg/kg Compound A (right panel). Spleens were harvested 2 hrs after the last dose, processed and stained with appropriate antibodies (marginal zone B cell: B220+; CD23−; CD21hi) and analyzed for staining on GUAVA EasyCyte FACS machine using Guava Cytosoft 5.2 software. This data shows that, in contrast to the Pan-PI3K inhibitor, a compound which is a selective PI3Kα inhibitor such as Compound A does not affect localization/viability of marginal zone B cells.
  • Example 37 TNP-Ficoll T-cell Independent B-cell Activation Assay
  • To test the effects of the compounds of the present invention in suppressing T cell independent antibody production, the TNP-Ficoll B-cell activation assay is used as described herein. Compounds of the present invention are dissolved in an appropriate vehicle (e.g. 5% 1-methyl-2-pyrrolidinone, 85 % polyethylene glycol 400, 10% Solutor). Compounds are administered orally approximately 1 hr before TNP-Ficoll treatment to 4-10 week old mice. To study the effects of the compounds on B-cell activation, one set of mice are grouped according to the following table:
  • Antigen Compound Administration
    Mice/ Comp injection at day−1 from day−1 to day−7
    Group# group treated Group TNP-F Route (mg/kg) Route Regimen
    1 4 Vehicle Antigen 200 uL ip 0 Po BID for 7
    only (0.5 mg/ml) days
    2 8 Antigen 0
    only
    3 8 Reference reference 30
    compound
    #
    1
    4 8 Test Antigen + 1
    5 8 compound cmp 3
    6 8 10
    7 8 30
    8 8 60
  • Four animals in group 1, and eight animals in groups 2 to 7 are euthanized in CO 2 2 hours after the last compound administration on day 7. Blood is immediately collected by cadio-puncture and kept at 37° C. for 1 hr to clot followed by overnight incubation at 4° C. to allow the clot to contract. The following day, serum is collected by decanting and centrifugation at 3000 rpm for 10 min. The collected serum is then frozen at −80° C. for future analysis.
  • Serum samples are analyzed for anti-TNP antibody titers by ELISA as described herein. TNP-BSA is coated onto a Nunc Maxisorb microtiter plate with 100 μl/well at a concentration of 10 μg/ml in phosphate buffered saline (PBS). The Maxisorb plate is incubated for 1.5 hours at room temperature and the solution is removed. 200 μl/well of blocking buffer (e.g. 1% BSA in PBS) is added to each well and incubated 1 hr at room temperature. The plate is washed once with 200 μl/well of PBS 0.05% Tween-20 (wash buffer). A 1:2 dilution of serum from each mouse in blocking buffer is added to each well in the first column (1) of the microtiter plate. The serum in each well of column 1 is then diluted 3-fold in blocking buffer and added to column 2 The serum in each well of column 2 is diluted 3-fold in blocking buffer and added to column 3 The procedure is repeated across the twelve columns of the microtiter plate. The microtiter plate is incubated 1 hr at room temperature. Serum is removed from the plate and the plate is washed three times with wash buffer. 100 μl/well of goat anti-mouse IgG3-HRP diluted 1:250 in blocking buffer is added to each well and incubated 1 hr at room temperature. The anti-mouse IgG3-HRP is removed from the microtiter plate and the plate is washed six times with wash buffer. HRP substrate (200 μl ABTS solution+30% H2O2+10 ml citrate buffer) is added to each well at 100 μl/well, incubated 2-20 minutes in the dark and the amount of anti-TNP IgG3 is determined spectrophotometrically at 405 nm. Similarly, anti-TNP IgM and total anti-TNP Ab are determined using anti-mouse IgM-HRP and anti-mouse Ig-HRP respectively.
  • A graph showing that Pan-PI3K inhibitor, but not a PI3Kα inhibitor such as Compound A, blocks B cell function in vivo is shown in FIG. 8. Mice were immunized with TNP-Ficoll and treated with 1) vehicle 2) 70 mg/kg GDC0941 3) 30 mg/kg Compound A 4) 60 mg/kg Compound A or 5) 120 mg/kg Compound A for 7 days. Antibody production was measured using ELISA and plotted as a percentage of control group that were treated with vehicle.
  • Example 38 In Vitro Effect on mTOR Pathways
  • An in vitro experiment using 786-0 cells treated with various concentrations of Compound B or Rapamycin was performed as described in Example 34. Cells were subsequently lysed and lysates were subjected to SDS-PAGE followed by Western blot analysis to detect the downstream pathway marker, pNDRG1. Results, shown in FIG. 5, indicate the differential effects on mTORC1 and mTORC2 for rapamycin relative to Compound B.
  • While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.
  • While preferred embodiments of the present invention have been shown and described herein, it will be obvious to those skilled in the art that such embodiments are provided by way of example only. Numerous variations, changes, and substitutions will now occur to those skilled in the art without departing from the invention. It should be understood that various alternatives to the embodiments of the invention described herein may be employed in practicing the invention. It is intended that the following claims define the scope of the invention and that methods and structures within the scope of these claims and their equivalents be covered thereby.

Claims (11)

1. A method for treating cancer comprising administering to a subject in need thereof a therapeutically effective amount of the mTOR inhibitor:
Figure US20160235758A1-20160818-C01259
or a pharmaceutically acceptable salt thereof; and a therapeutically effective amount of the PI3-kinase α inhibitor:
Figure US20160235758A1-20160818-C01260
or a pharmaceutically acceptable salt thereof: wherein said cancer is renal cell carcinoma or endometrial cancer.
2. (canceled)
3. The method of claim 1, wherein said cancer is endometrial cancer.
4. The method of claim 1, wherein said cancer is renal cell carcinoma.
5-18. (canceled)
19. The method of claim 1, wherein the PI3-kinase α inhibitor and/or the mTOR inhibitor are administered parenterally, orally, intraperitoneally, intravenously, intraarterially, transdermally, intramuscularly, liposomally, via local delivery by catheter or stent, subcutaneously, intraadiposally, or intrathecally.
20. The method of claim 1, wherein the PI3-kinase α inhibitor and/or the mTOR inhibitor are co-administered to the subject in the same formulation.
21. The method of claim 1, wherein the PI3-kinase α inhibitor and/or the mTOR inhibitor are co-administered to the subject in different formulations.
22-38. (canceled)
39. The method of claim 1, wherein the PI3-kinase α inhibitor and/or the mTOR inhibitor are administered orally.
40. The method of claim 1, wherein the PI3-kinase α inhibitor and/or the mTOR inhibitor are administered simultaneously or sequentially.
US15/041,172 2011-02-23 2016-02-11 Combination of mtor inhibitors and pi3-kinase inhibitors, and uses thereof Abandoned US20160235758A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/041,172 US20160235758A1 (en) 2011-02-23 2016-02-11 Combination of mtor inhibitors and pi3-kinase inhibitors, and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161446016P 2011-02-23 2011-02-23
US13/403,856 US9295673B2 (en) 2011-02-23 2012-02-23 Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US15/041,172 US20160235758A1 (en) 2011-02-23 2016-02-11 Combination of mtor inhibitors and pi3-kinase inhibitors, and uses thereof

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/403,856 Continuation US9295673B2 (en) 2011-02-23 2012-02-23 Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof

Publications (1)

Publication Number Publication Date
US20160235758A1 true US20160235758A1 (en) 2016-08-18

Family

ID=47072669

Family Applications (3)

Application Number Title Priority Date Filing Date
US13/403,856 Expired - Fee Related US9295673B2 (en) 2011-02-23 2012-02-23 Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US14/253,782 Abandoned US20150087627A1 (en) 2011-02-23 2014-04-15 Combination of kinase inhibitors and uses thereof
US15/041,172 Abandoned US20160235758A1 (en) 2011-02-23 2016-02-11 Combination of mtor inhibitors and pi3-kinase inhibitors, and uses thereof

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US13/403,856 Expired - Fee Related US9295673B2 (en) 2011-02-23 2012-02-23 Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US14/253,782 Abandoned US20150087627A1 (en) 2011-02-23 2014-04-15 Combination of kinase inhibitors and uses thereof

Country Status (8)

Country Link
US (3) US9295673B2 (en)
EP (1) EP2678018A4 (en)
JP (1) JP6130305B2 (en)
CN (2) CN106619647A (en)
AR (1) AR085397A1 (en)
CA (1) CA2828483A1 (en)
TW (1) TWI592411B (en)
WO (1) WO2012148540A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160089371A1 (en) * 2013-03-15 2016-03-31 Intellikine, Llc Combination of Kinase Inhibitors and Uses Thereof
WO2018237370A1 (en) * 2017-06-23 2018-12-27 Accro Bioscience Inc. Heteroaryl compounds as inhibitors of necrosis, composition and method using the same
CN109111464A (en) * 2017-06-23 2019-01-01 爱科诺生物医药股份有限公司 A kind of heterocyclic compound with meronecrosis inhibitory activity
US11352328B2 (en) 2016-07-12 2022-06-07 Arisan Therapeutics Inc. Heterocyclic compounds for the treatment of arenavirus

Families Citing this family (59)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1831225A2 (en) 2004-11-19 2007-09-12 The Regents of the University of California Anti-inflammatory pyrazolopyrimidines
ES2363725T3 (en) * 2006-08-16 2011-08-12 Novartis Ag METHOD FOR PREPARING SOLID DISPERSIONS OF MIDOSTAURINE.
WO2009046448A1 (en) 2007-10-04 2009-04-09 Intellikine, Inc. Chemical entities and therapeutic uses thereof
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
CN102124009B (en) 2008-07-08 2014-07-23 因特利凯公司 Kinase inhibitors and methods of use
US20110224223A1 (en) 2008-07-08 2011-09-15 The Regents Of The University Of California, A California Corporation MTOR Modulators and Uses Thereof
US8476282B2 (en) 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
WO2011047384A2 (en) 2009-10-16 2011-04-21 The Regents Of The University Of California Methods of inhibiting ire1
GB201007286D0 (en) 2010-04-30 2010-06-16 Astex Therapeutics Ltd New compounds
WO2011149937A1 (en) * 2010-05-24 2011-12-01 Intellikine, Inc. Heterocyclic compounds and uses thereof
GB201020179D0 (en) 2010-11-29 2011-01-12 Astex Therapeutics Ltd New compounds
WO2012116237A2 (en) 2011-02-23 2012-08-30 Intellikine, Llc Heterocyclic compounds and uses thereof
MX370814B (en) 2011-09-02 2020-01-08 Univ California Substituted pyrazolo[3,4-d]pyrimidines and uses thereof.
GB201118675D0 (en) 2011-10-28 2011-12-14 Astex Therapeutics Ltd New compounds
GB201118656D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
GB201118652D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
GB201118654D0 (en) 2011-10-28 2011-12-07 Astex Therapeutics Ltd New compounds
US20140323721A1 (en) * 2011-11-23 2014-10-30 Intellikine, Llc Heterocyclic compounds and uses thereof
GB201209609D0 (en) 2012-05-30 2012-07-11 Astex Therapeutics Ltd New compounds
GB201209613D0 (en) 2012-05-30 2012-07-11 Astex Therapeutics Ltd New compounds
EP2900673A4 (en) 2012-09-26 2016-10-19 Univ California Modulation of ire1
EP2968340A4 (en) * 2013-03-15 2016-08-10 Intellikine Llc Combination of kinase inhibitors and uses thereof
GB201307577D0 (en) 2013-04-26 2013-06-12 Astex Therapeutics Ltd New compounds
CN104177346A (en) * 2013-05-21 2014-12-03 苏州科捷生物医药有限公司 Quinazoline compound and use thereof
WO2015095329A1 (en) * 2013-12-19 2015-06-25 Medimmune, Llc Compositions and methods for treating sarcoma
JO3512B1 (en) 2014-03-26 2020-07-05 Astex Therapeutics Ltd Quinoxaline derivatives useful as fgfr kinase modulators
LT3122358T (en) 2014-03-26 2021-04-12 Astex Therapeutics Ltd. Combinations of fgfr- and cmet-inhibitors for the treatment of cancer
US10736900B2 (en) 2014-03-26 2020-08-11 Astex Therapeutics Ltd Combinations of an FGFR inhibitor and an IGF1R inhibitor
CN103864800A (en) * 2014-04-03 2014-06-18 定陶县友帮化工有限公司 Synthesis method for 6-chloroimidazo[1,2-b] pyridazine
WO2015160868A1 (en) 2014-04-16 2015-10-22 Signal Pharmaceuticals, Llc Methods for treating cancer using tor kinase inhibitor combination therapy
AR101106A1 (en) 2014-07-02 2016-11-23 Pharmacyclics Llc BRUTON THYROSINE QUINASE INHIBITORS
CN105330699B (en) * 2014-08-13 2018-12-04 山东汇睿迪生物技术有限公司 A kind of phosphorous pyrido [2,3-d] pyrimidin-7-ones class compound or its pharmaceutically acceptable salt, pharmaceutical composition and its application
CN107073066B (en) 2014-09-11 2021-09-17 加利福尼亚大学董事会 mTORC1 inhibitors
WO2016057931A1 (en) 2014-10-10 2016-04-14 The Research Foundation For The State University Of New York Trifluoromethoxylation of arenes via intramolecular trifluoromethoxy group migration
WO2016064958A1 (en) 2014-10-22 2016-04-28 Bristol-Myers Squibb Company Heteroaryl substituted pyrrolotriazine amine compounds as pi3k inhibitors
ES2807785T3 (en) 2014-10-22 2021-02-24 Bristol Myers Squibb Co Bicyclic heteroarylamine compounds as pi3k inhibitors
JOP20200201A1 (en) 2015-02-10 2017-06-16 Astex Therapeutics Ltd Pharmaceutical compositions comprising n-(3,5-dimethoxyphenyl)-n'-(1-methylethyl)-n-[3-(1-methyl-1h-pyrazol-4-yl)quinoxalin-6-yl]ethane-1,2-diamine
CN107530291A (en) 2015-03-03 2018-01-02 奎尔波特股份有限公司 Double heavy duty liposomal pharmaceutical preparations
EP3265059A4 (en) 2015-03-03 2018-08-29 Cureport Inc. Combination liposomal pharmaceutical formulations
US10478494B2 (en) 2015-04-03 2019-11-19 Astex Therapeutics Ltd FGFR/PD-1 combination therapy for the treatment of cancer
WO2016197190A1 (en) * 2015-06-09 2016-12-15 The Walter And Eliza Hall Institute Of Medical Research Structure of plasmepsin v in complex with an inhibitor and uses thereof
CN108026136A (en) 2015-08-06 2018-05-11 奇默里克斯公司 Pyrrolopyrimidine nucleosides as useful antivirotic and the like
LT3353177T (en) 2015-09-23 2020-08-25 Janssen Pharmaceutica Nv Tricyclic heterocycles for the treatment of cancer
CA2996989C (en) 2015-09-23 2023-10-03 Janssen Pharmaceutica Nv Bi-heteroaryl substituted 1,4-benzodiazepines and uses thereof for the treatment of cancer
CN105396036A (en) * 2015-12-07 2016-03-16 赵培序 Traditional Chinese medicine composition for treating cervical spondylosis and lubar intervertebral disc protrusion
WO2017172596A1 (en) 2016-03-28 2017-10-05 Incyte Corporation Pyrrolotriazine compounds as tam inhibitors
EP3442535B1 (en) 2016-04-15 2022-06-01 Cancer Research Technology Limited Heterocyclic compounds as ret kinase inhibitors
KR102390578B1 (en) * 2016-04-15 2022-04-26 캔써 리서치 테크놀로지 리미티드 Heterocyclic compounds as RET kinase inhibitors
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
GB201705971D0 (en) 2017-04-13 2017-05-31 Cancer Res Tech Ltd Inhibitor compounds
EP3684771A1 (en) 2017-09-21 2020-07-29 Chimerix, Inc. MORPHIC FORMS OF 4-AMINO-7-(3,4-DIHYDROXY-5-(HYDROXYMETHYL)TETRAHYDROFURAN-2-YL)-2-METHYL-7H-PYRROLO[2,3-d]PYRIMIDINE-5-CARBOXAMIDE AND USES THEREOF
FR3075794A1 (en) * 2017-12-21 2019-06-28 Galderma Research & Development NEW MTOR INHIBITOR COMPOUNDS
FR3075795A1 (en) 2017-12-21 2019-06-28 Galderma Research & Development NEW MTOR INHIBITOR COMPOUNDS
SG11202010560QA (en) 2018-05-01 2020-11-27 Revolution Medicines Inc C26-linked rapamycin analogs as mtor inhibitors
JP7358387B2 (en) 2018-05-01 2023-10-10 レヴォリューション・メディスンズ,インコーポレイテッド C40-, C28- and C-32-linked rapamycin analogs as MTOR inhibitors
CR20210050A (en) * 2018-06-27 2021-06-10 Reborna Biosciences Inc Prophylactic or therapeutic agent for spinal muscular atrophy
US11426472B2 (en) 2018-10-17 2022-08-30 Purdue Research Foundation Fibroblast activation protein (FAP) targeted imaging and therapy in fibrosis
WO2022020114A2 (en) 2020-07-10 2022-01-27 Ting Therapeutics Llc Methods for the prevention and treatment of hearing loss
CN116514661B (en) * 2023-06-30 2023-09-12 研峰科技(北京)有限公司 Continuous flow synthesis method of 4-bromo-2-nitrophenol

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010049481A1 (en) * 2008-10-31 2010-05-06 Novartis Ag Combination of a phosphatidylinositol-3-kinase (pi3k) inhibitor and a mtor inhibitor
WO2011022439A1 (en) * 2009-08-17 2011-02-24 Intellikine, Inc. Heterocyclic compounds and uses thereof

Family Cites Families (364)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB812366A (en) 1955-08-18 1959-04-22 Wellcome Found Improvements in and relating to derivatives of pyrimidine and the preparation thereof
GB937725A (en) 1960-05-11 1963-09-25 Ciba Ltd Pyrazolo[3:4-d]pyrimidines
DE2004713A1 (en) 1970-02-03 1971-08-12 Boehnnger Mannheim GmbH, 6800 Mann heim Waldhof Nitrofuryl triazolo pyrimidine and process for their preparation
IT1153216B (en) 1981-10-16 1987-01-14 Schering Ag PROCEDURE FOR THE PREPARATION OF CYANETHEROCYCLIC COMPOUNDS
DE3406533A1 (en) 1984-02-23 1985-08-29 Boehringer Mannheim Gmbh, 6800 Mannheim USE OF ADENOSINE DERIVATIVES AS ANTIALLERGICA AND MEDICINAL PRODUCTS CONTAINING THEM
US5310731A (en) 1984-06-28 1994-05-10 Whitby Research, Inc. N-6 substituted-5'-(N-substitutedcarboxamido)adenosines as cardiac vasodilators and antihypertensive agents
JPS61109797U (en) 1984-12-25 1986-07-11
WO1990003370A1 (en) 1988-09-28 1990-04-05 Microprobe Corporation DERIVATIVES OF PYRAZOLO[3,4-d]PYRIMIDINE
US5428125A (en) 1989-07-17 1995-06-27 The Dow Chemical Company Mesogenic polycyanates and thermosets thereof
US5442039A (en) 1989-07-17 1995-08-15 The Dow Chemical Company Mesogenic polycyanates and thermosets thereof
US5763597A (en) 1989-09-15 1998-06-09 Metabasis Therapeutics, Inc. Orally active adenosine kinase inhibitors
US5795977A (en) 1989-09-15 1998-08-18 Metabasis Therapeutics, Inc. Water soluble adenosine kinase inhibitors
US5721356A (en) 1989-09-15 1998-02-24 Gensia, Inc. Orally active adenosine kinase inhibitors
US5674998A (en) 1989-09-15 1997-10-07 Gensia Inc. C-4' modified adenosine kinase inhibitors
US5646128A (en) 1989-09-15 1997-07-08 Gensia, Inc. Methods for treating adenosine kinase related conditions
US5763596A (en) 1989-09-15 1998-06-09 Metabasis Therapeutics, Inc. C-4' modified adenosine kinase inhibitors
GB9009542D0 (en) 1990-04-27 1990-06-20 Beecham Group Plc Novel compounds
GB9113137D0 (en) 1990-07-13 1991-08-07 Ici Plc Thioxo heterocycles
US5563257A (en) 1990-08-20 1996-10-08 Boehringer Mannheim Gmbh Phospholipid derivatives of nucleosides
DE4026265A1 (en) 1990-08-20 1992-02-27 Boehringer Mannheim Gmbh NEW PHOSPHOLIPID DERIVATIVES OF NUCLEOSIDES, THEIR PRODUCTION AND THEIR USE AS ANTIVIRAL MEDICINAL PRODUCTS
US5652366A (en) 1990-09-25 1997-07-29 Rhone-Poulenc Rorer Pharmaceuticals Inc. DI (1R)-(-)camphosulfonic acid) salt, preparation thereof and use thereof
JP2505085B2 (en) 1990-09-25 1996-06-05 ローヌ−プーラン ローラー インターナショナル (ホウルディングス) インコーポレイテッド Compounds with antihypertensive and antiischemic properties
US5561134A (en) 1990-09-25 1996-10-01 Rhone-Poulenc Rorer Pharmaceuticals Inc. Compounds having antihypertensive, cardioprotective, anti-ischemic and antilipolytic properties
GB9103839D0 (en) 1991-02-23 1991-04-10 Smithkline Beecham Plc Pharmaceuticals
US5710158A (en) 1991-05-10 1998-01-20 Rhone-Poulenc Rorer Pharmaceuticals Inc. Aryl and heteroaryl quinazoline compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
US5480883A (en) 1991-05-10 1996-01-02 Rhone-Poulenc Rorer Pharmaceuticals Inc. Bis mono- and bicyclic aryl and heteroaryl compounds which inhibit EGF and/or PDGF receptor tyrosine kinase
MX9300141A (en) 1992-01-13 1994-07-29 Smithkline Beecham Corp NOVEL IMIDAZOLE COMPOUNDS, PROCEDURE FOR THE PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING IT.
US5916891A (en) 1992-01-13 1999-06-29 Smithkline Beecham Corporation Pyrimidinyl imidazoles
DE4204032A1 (en) 1992-02-12 1993-08-19 Boehringer Mannheim Gmbh NEW LIPONUCLEOTIDES, THEIR PRODUCTION AND THEIR USE AS ANTIVIRAL MEDICINAL PRODUCTS
DE4204031A1 (en) 1992-02-12 1993-08-19 Boehringer Mannheim Gmbh NEW LIPID PHOSPHONIC ACID-NUCLEOSIDE CONJUGATES AND THEIR USE AS ANTIVIRAL MEDICINAL PRODUCTS
WO1993018035A1 (en) 1992-03-04 1993-09-16 Abbott Laboratories Angiotensin ii receptor antagonists
TW225528B (en) 1992-04-03 1994-06-21 Ciba Geigy Ag
GB9208135D0 (en) 1992-04-13 1992-05-27 Ludwig Inst Cancer Res Polypeptides having kinase activity,their preparation and use
JPH07506252A (en) 1992-04-24 1995-07-13 エス・アール・アイ・インターナシヨナル In vivo homologous sequence targeting in eukaryotic cells
CA2117476C (en) 1992-06-19 2000-02-22 R. Andrew Wood Infrared camera with thermoelectric temperature stabilization
US6057305A (en) 1992-08-05 2000-05-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic Antiretroviral enantiomeric nucleotide analogs
TW444018B (en) 1992-12-17 2001-07-01 Pfizer Pyrazolopyrimidines
CA2154681A1 (en) 1993-02-03 1994-08-18 Mark David Erion Adenosine kinase inhibitors comprising lyxofuranosyl derivatives
IL108523A0 (en) 1993-02-03 1994-05-30 Gensia Inc Pharmaceutical compositions containing adenosine kinase inhibitors for preventing or treating conditions involving inflammatory responses and pain
WO1995012588A1 (en) 1993-11-05 1995-05-11 Biochem Pharma Inc. Antineoplastic heteronaphthoquinones
US5679683A (en) 1994-01-25 1997-10-21 Warner-Lambert Company Tricyclic compounds capable of inhibiting tyrosine kinases of the epidermal growth factor receptor family
IL112249A (en) 1994-01-25 2001-11-25 Warner Lambert Co Pharmaceutical compositions containing di and tricyclic pyrimidine derivatives for inhibiting tyrosine kinases of the epidermal growth factor receptor family and some new such compounds
US6632789B1 (en) 1994-04-29 2003-10-14 The United States Of America As Represented By The Secretary Of The Navy Methods for modulating T cell responses by manipulating intracellular signal transduction
DE4418690A1 (en) 1994-05-28 1996-01-11 Boehringer Mannheim Gmbh Nucleoside 5'-mono-phosphate ester(s) of glyceryl di:ether derivs.
US6323201B1 (en) 1994-12-29 2001-11-27 The Regents Of The University Of California Compounds for inhibition of ceramide-mediated signal transduction
DE69609602T2 (en) 1995-04-03 2001-04-12 Novartis Ag PYRAZOLE DERIVATIVES AND METHOD FOR THE PRODUCTION THEREOF
US6312894B1 (en) 1995-04-03 2001-11-06 Epoch Pharmaceuticals, Inc. Hybridization and mismatch discrimination using oligonucleotides conjugated to minor groove binders
US5977061A (en) 1995-04-21 1999-11-02 Institute Of Organic Chemistry And Biochemistry Of The Academy Of Sciences Of The Czech Republic N6 - substituted nucleotide analagues and their use
US5593997A (en) 1995-05-23 1997-01-14 Pfizer Inc. 4-aminopyrazolo(3-,4-D)pyrimidine and 4-aminopyrazolo-(3,4-D)pyridine tyrosine kinase inhibitors
US6403599B1 (en) 1995-11-08 2002-06-11 Pfizer Inc Corticotropin releasing factor antagonists
IL122246A (en) 1995-06-07 2004-06-01 Searle & Co Spironolactone and angiotensin ii antagonist combination for treatment of congestive heart failure
AU5982296A (en) 1995-06-07 1996-12-30 G.D. Searle & Co. Method to treat cardiofibrosis with a combination of an angi otensin ii antagonist and spironolactone
US5665721A (en) 1995-06-07 1997-09-09 Abbott Laboratories Heterocyclic substituted cyclopentane compounds
ES2167571T3 (en) 1995-06-07 2002-05-16 Searle & Co ANTIGONIST COMBINATION THERAPY OF ALDOSTERONE EPOXY-STEROID AND ANGIOTENSIN II ANTAGONIST FOR THE TREATMENT OF CONGESTIVE CARDIAC FAILURE.
US5763885A (en) 1995-12-19 1998-06-09 Loral Infrared & Imaging Systems, Inc. Method and apparatus for thermal gradient stabilization of microbolometer focal plane arrays
US5747235A (en) 1996-01-26 1998-05-05 Eastman Kodak Company Silver halide light sensitive emulsion layer having enhanced photographic sensitivity
DE19603576A1 (en) 1996-02-01 1997-08-07 Bayer Ag Acylated 4-amino and 4-hydrazinopyrimidines
CH690773A5 (en) 1996-02-01 2001-01-15 Novartis Ag Pyrrolo (2,3-d) pyrimides and their use.
US5914488A (en) 1996-03-05 1999-06-22 Mitsubishi Denki Kabushiki Kaisha Infrared detector
GB9611460D0 (en) 1996-06-01 1996-08-07 Ludwig Inst Cancer Res Novel lipid kinase
IL127559A0 (en) 1996-06-20 1999-10-28 Univ Texas Compounds and methods for providing pharmacologically active preparations and uses thereof
ES2239779T3 (en) 1996-10-02 2005-10-01 Novartis Ag PIRIMIDINE DERIVATIVES AND PROCEDURES FOR THE PREPARATION OF THE SAME.
EP0837063A1 (en) 1996-10-17 1998-04-22 Pfizer Inc. 4-Aminoquinazoline derivatives
US5922753A (en) 1996-10-23 1999-07-13 Zymogenetics, Inc. Methods for treating bone deficit conditions with benzothiazole
US6342514B1 (en) 1996-10-23 2002-01-29 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5965573A (en) 1996-10-23 1999-10-12 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5994358A (en) 1996-10-23 1999-11-30 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US6251901B1 (en) 1996-10-23 2001-06-26 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5919808A (en) 1996-10-23 1999-07-06 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5990169A (en) 1996-10-23 1999-11-23 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5948776A (en) 1996-10-23 1999-09-07 Zymogenetic, Inc. Compositions and methods for treating bone deficit conditions
US6153631A (en) 1996-10-23 2000-11-28 Zymogenetics, Inc. Compositions and methods for treating bone deficit conditions
US5858753A (en) 1996-11-25 1999-01-12 Icos Corporation Lipid kinase
CA2274250A1 (en) 1996-12-06 1998-06-11 Vertex Pharmaceuticals Incorporated Inhibitors of interleukin-1.beta. converting enzyme
US6093737A (en) 1996-12-30 2000-07-25 Merck & Co., Inc. Inhibitors of farnesyl-protein transferase
JPH10206995A (en) 1997-01-21 1998-08-07 Konica Corp Silver halide photographic sensitive material
ATE271130T1 (en) 1997-02-07 2004-07-15 Univ Princeton MODIFIED PROTEIN KINASES THAT CAN USE MODIFIED NUCLEOTIDE TRIPHOSPHATE SUBSTRATES
ATE242245T1 (en) 1997-03-19 2003-06-15 Basf Ag PYRROLO(2,3-D)PYRIMIDINES AND THEIR USE AS TYROSINE KINASE INHIBITORS
US7863444B2 (en) 1997-03-19 2011-01-04 Abbott Laboratories 4-aminopyrrolopyrimidines as kinase inhibitors
AU6780398A (en) 1997-03-28 1998-10-22 Du Pont Merck Pharmaceutical Company, The Heterocyclic integrin inhibitor prodrugs
AU7449598A (en) 1997-05-23 1998-12-11 Nippon Shinyaku Co. Ltd. Medicinal composition for prevention or treatment of hepatopathy
GB9711650D0 (en) 1997-06-05 1997-07-30 Pfizer Ltd Compounds useful in therapy
US6207679B1 (en) 1997-06-19 2001-03-27 Sepracor, Inc. Antimicrobial agents uses and compositions related thereto
US6340759B1 (en) 1997-10-02 2002-01-22 Eisai Co., Ltd. Fused pyridine derivatives
US6649631B1 (en) 1997-10-23 2003-11-18 The Board Of Regents Of The University Of Texas System Compositions and methods for treating bone deficit conditions
CA2309350C (en) 1997-11-12 2007-04-03 Mitsubishi Chemical Corporation Purine derivatives and medicaments comprising the same as active ingredient
US6191170B1 (en) 1998-01-13 2001-02-20 Tularik Inc. Benzenesulfonamides and benzamides as therapeutic agents
US7715989B2 (en) 1998-04-03 2010-05-11 Elitech Holding B.V. Systems and methods for predicting oligonucleotide melting temperature (TmS)
US6127121A (en) 1998-04-03 2000-10-03 Epoch Pharmaceuticals, Inc. Oligonucleotides containing pyrazolo[3,4-D]pyrimidines for hybridization and mismatch discrimination
US6251689B1 (en) 1998-05-14 2001-06-26 Telik, Inc. Methods for the solid phase synthesis of combinatorial libraries of benzimidazoles benzoxazoles benzothiazoles and derivatives thereof
BR9911612A (en) 1998-06-02 2001-02-06 Osi Pharm Inc Pyrrole [2,3d] pyrimidine compositions and their uses
JP2000072773A (en) 1998-08-28 2000-03-07 Zeria Pharmaceut Co Ltd Purine derivative
JP2002527359A (en) 1998-09-18 2002-08-27 ビーエーエスエフ アクチェンゲゼルシャフト 4-aminopyrrolipyrimidine as a kinase inhibitor
US6713474B2 (en) 1998-09-18 2004-03-30 Abbott Gmbh & Co. Kg Pyrrolopyrimidines as therapeutic agents
ID28945A (en) 1998-11-16 2001-07-19 Basf Ag TURUNAN-TURUNAN 3- (BENZALOL-7-IL) -IH-PIRIMIDIN-2,4-DION
US6319660B1 (en) 1998-12-28 2001-11-20 Eastman Kodak Company Color photographic element containing speed improving compound
WO2000042042A2 (en) 1999-01-11 2000-07-20 Princeton University High affinity inhibitors for target validation and uses thereof
CZ27399A3 (en) 1999-01-26 2000-08-16 Ústav Experimentální Botaniky Av Čr Substituted nitrogen heterocyclic derivatives process of their preparation, the derivatives employed as medicaments, pharmaceutical composition and a compound pharmaceutical preparation in which these derivatives are comprised as well as use of these derivatives for preparing medicaments
EP1040831A3 (en) 1999-04-02 2003-05-02 Pfizer Products Inc. Use of corticotropin releasing factor (CRF) antagonists to prevent sudden death
SE515856C2 (en) 1999-05-19 2001-10-22 Ericsson Telefon Ab L M Carrier for electronic components
US7049312B1 (en) 1999-06-03 2006-05-23 Abbott Gmbh & Co. Kg Benzothiazinone and benzoxazinone compounds
US6387894B1 (en) 1999-06-11 2002-05-14 Pfizer Inc. Use of CRF antagonists and renin-angiotensin system inhibitors
TWI262914B (en) 1999-07-02 2006-10-01 Agouron Pharma Compounds and pharmaceutical compositions for inhibiting protein kinases
PE20010306A1 (en) 1999-07-02 2001-03-29 Agouron Pharma INDAZOLE COMPOUNDS AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM USEFUL FOR THE INHIBITION OF PROTEIN KINASE
GB9919588D0 (en) 1999-08-18 1999-10-20 Hoechst Schering Agrevo Gmbh Fungicidal compounds
JP4831906B2 (en) 1999-08-27 2011-12-07 ケモセントリックス, インコーポレイテッド Heterocyclic compounds and methods for modulating CXCR3 function
AU780846B2 (en) 1999-09-16 2005-04-21 Curis, Inc. Mediators of hedgehog signaling pathways, compositions and uses related thereto
CA2385747A1 (en) 1999-09-17 2001-03-22 Gavin C. Hirst Pyrazolopyrimidines as therapeutic agents
US6921763B2 (en) 1999-09-17 2005-07-26 Abbott Laboratories Pyrazolopyrimidines as therapeutic agents
US6506769B2 (en) 1999-10-06 2003-01-14 Boehringer Ingelheim Pharmaceuticals, Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
CA2384378C (en) 1999-10-06 2011-05-24 Boehringer Ingelheim Pharmaceuticals, Inc. Heterocyclic compounds useful as inhibitors of tyrosine kinases
US6472153B1 (en) 1999-10-26 2002-10-29 Epoch Biosciences, Inc. Hybridization-triggered fluorescent detection of nucleic acids
US6660845B1 (en) 1999-11-23 2003-12-09 Epoch Biosciences, Inc. Non-aggregating, non-quenching oligomers comprising nucleotide analogues; methods of synthesis and use thereof
GB0002032D0 (en) 2000-01-28 2000-03-22 Zeneca Ltd Chemical compounds
US7217722B2 (en) 2000-02-01 2007-05-15 Kirin Beer Kabushiki Kaisha Nitrogen-containing compounds having kinase inhibitory activity and drugs containing the same
US7115653B2 (en) 2000-03-30 2006-10-03 Curis, Inc. Small organic molecule regulators of cell proliferation
US6613798B1 (en) 2000-03-30 2003-09-02 Curis, Inc. Small organic molecule regulators of cell proliferation
US6667300B2 (en) 2000-04-25 2003-12-23 Icos Corporation Inhibitors of human phosphatidylinositol 3-kinase delta
GEP20084317B (en) 2000-04-25 2008-02-25 Icos Corp Inhibitors of human phosphatidyl-inositol 3-kinase delta
AU2001265137B2 (en) 2000-05-30 2007-06-14 Indiana University Research And Technology Corporation Compositions and methods for identifying agents which modulate PTEN function and PI-3 kinase pathways
WO2002000650A2 (en) 2000-06-27 2002-01-03 Genelabs Technologies, Inc. Novel compounds possessing antibacterial, antifungal or antitumor activity
US6534691B2 (en) 2000-07-18 2003-03-18 E. I. Du Pont De Nemours And Company Manufacturing process for α-olefins
PE20020354A1 (en) 2000-09-01 2002-06-12 Novartis Ag HYDROXAMATE COMPOUNDS AS HISTONE-DESACETILASE (HDA) INHIBITORS
CA2425259A1 (en) 2000-10-11 2002-04-18 Tularik, Inc. Modulation of ccr4 function
ATE431355T1 (en) 2000-10-11 2009-05-15 Applera Corp FLUORESCENT NUCLEOBASECONJUGATES WITH ANIONIC LINKERS
JP2002131859A (en) 2000-10-19 2002-05-09 Konica Corp Infrared sensitive silver halide photographic sensitive material for photographing and infrared sensitive silver halide emulsion
US6890747B2 (en) 2000-10-23 2005-05-10 Warner-Lambert Company Phosphoinositide 3-kinases
HUP0500880A2 (en) 2000-12-11 2006-05-29 Tularik Inc Cxcr3 antagonists and pharmaceutical compositions thereof
US6632809B2 (en) 2000-12-15 2003-10-14 Vertex Pharmaceuticals Incorporated Gyrase inhibitors and uses thereof
US7157487B2 (en) 2000-12-28 2007-01-02 Daiichi Pharmaceutical Co., Ltd. Vla-4 inhibitors
AU2002243791B2 (en) 2001-01-22 2006-06-29 Isis Pharmaceuticals, Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
US7105499B2 (en) 2001-01-22 2006-09-12 Merck & Co., Inc. Nucleoside derivatives as inhibitors of RNA-dependent RNA viral polymerase
MXPA03008560A (en) 2001-03-22 2004-06-30 Abbot Gmbh & Co Kg Single-stage pfc + ballast control circuit/general purpose power converter.
US7250569B2 (en) 2001-04-26 2007-07-31 New York University School Of Medicine Method for dissolving nanostructural materials
CA2444531A1 (en) 2001-05-08 2002-11-14 Kudos Pharmaceuticals Limited Isoquinolinone derivatives as parp inhibitors
US7144903B2 (en) 2001-05-23 2006-12-05 Amgen Inc. CCR4 antagonists
CA2450628A1 (en) 2001-06-13 2002-12-19 Genesoft Pharmaceuticals, Inc. Benzothiophene compounds having antiinfective activity
US6777425B2 (en) 2001-06-13 2004-08-17 Genesoft Pharmaceuticals, Inc. Isoquinoline compounds having antiinfective activity
US20030236198A1 (en) 2001-06-13 2003-12-25 Genesoft, Inc. Antipathogenic benzamide compounds
GB0115109D0 (en) 2001-06-21 2001-08-15 Aventis Pharma Ltd Chemical compounds
WO2003000187A2 (en) 2001-06-21 2003-01-03 Ariad Pharmaceuticals, Inc. Novel pyrazolo-and pyrrolo-pyrimidines and uses thereof
ES2274986T3 (en) 2001-08-01 2007-06-01 MERCK &amp; CO., INC. BENZIMIDAZO DERIVATIVES 4,5-F / ISOQUINOLINONA.
AU2002327422A1 (en) 2001-08-03 2003-03-18 Abbott Laboratories Method of identifying inhibitors of lck
WO2003016275A1 (en) 2001-08-10 2003-02-27 Shionogi & Co., Ltd. Antiviral agent
JP2003073357A (en) 2001-09-03 2003-03-12 Mitsubishi Pharma Corp Rho KINASE INHIBITOR COMPRISING AMIDE COMPOUND
CA2458926A1 (en) 2001-09-13 2003-03-13 Genesoft Pharmaceuticals, Inc. Methods of treating infection by drug resistant bacteria
US7101884B2 (en) 2001-09-14 2006-09-05 Merck & Co., Inc. Tyrosine kinase inhibitors
AUPR769501A0 (en) 2001-09-14 2001-10-11 Biomolecular Research Institute Limited Cytokine receptor 1
TWI330183B (en) 2001-10-22 2010-09-11 Eisai R&D Man Co Ltd
US7241890B2 (en) 2001-10-30 2007-07-10 Conforma Therapeutics Corporation Purine analogs having HSP90-inhibiting activity
US7319858B2 (en) 2001-11-16 2008-01-15 Cingular Wireless Ii, Llc System and method for querying message information
WO2003048081A2 (en) 2001-12-04 2003-06-12 Bristol-Myers Squibb Company Glycinamides as factor xa inhibitors
WO2003059884A1 (en) 2001-12-21 2003-07-24 X-Ceptor Therapeutics, Inc. Modulators of lxr
JP4085237B2 (en) 2001-12-21 2008-05-14 日本電気株式会社 Mobile phone usage contract system and communication method
BR0207583A (en) 2001-12-26 2004-04-27 Genelabs Tech Inc Polyamide derivatives having antibacterial, antifungal or antitumor activity
US7414036B2 (en) 2002-01-25 2008-08-19 Muscagen Limited Compounds useful as A3 adenosine receptor agonists
US20030225098A1 (en) 2002-03-21 2003-12-04 Hirst Gavin C. Kinase inhibitors
AU2003225933A1 (en) 2002-03-22 2003-10-13 Cellular Genomics, Inc. AN IMPROVED FORMULATION OF CERTAIN PYRAZOLO(3,4-d) PYRIMIDINES AS KINASE MODULATORS
JP4663986B2 (en) 2002-03-26 2011-04-06 ツェントファーム・ゲーエムベーハー Fredericamycin-derivative
US7166293B2 (en) 2002-03-29 2007-01-23 Carlsbad Technology, Inc. Angiogenesis inhibitors
DE10217046A1 (en) 2002-04-17 2003-11-06 Bioleads Gmbh Fredericamycin derivatives
BR0309556A (en) 2002-04-26 2005-02-09 Pfizer Prod Inc Pyrimidine-2,4,6-trione metalloproteinase inhibitors
US20050203110A1 (en) 2002-05-23 2005-09-15 Coleman Paul J. Mitotic kinesin inhibitors
WO2003106426A1 (en) 2002-06-14 2003-12-24 Cytokinetics, Inc. Compounds, compositions, and methods
US7265111B2 (en) 2002-06-27 2007-09-04 Sanofi-Aventis Deutschland Gmbh Adenosine analogues and their use as pharmaceutical agents
PL215132B1 (en) 2002-06-27 2013-10-31 Novo Nordisk As Aryl carbonyl derivatives as therapeutic agents
CN1678311A (en) 2002-06-27 2005-10-05 诺沃挪第克公司 Aryl carbonyl derivatives as therapeutic agents
WO2004004644A2 (en) 2002-07-05 2004-01-15 Beth Israel Deaconess Medical Center Combination of mtor inhibitor and a tyrosine kinase inhibitor for the treatment of neoplasms
DE10230917A1 (en) 2002-07-09 2004-02-05 Bioleads Gmbh Fredericamycin derivatives
WO2004006906A2 (en) 2002-07-15 2004-01-22 Combinatorx, Incorporated Methods for the treatment of neoplasms
WO2004014377A1 (en) 2002-08-13 2004-02-19 Warner-Lambert Company Llc 4-hydroxyquinoline derivatives as matrix metalloproteinase inhibitors
US7598377B2 (en) 2002-08-16 2009-10-06 Astrazeneca Ab Inhibition of phosphoinositide 3-kinase β
AU2003262747A1 (en) 2002-08-21 2004-03-11 Cytokinetics, Inc. Compounds, compositions, and methods
US20030139427A1 (en) 2002-08-23 2003-07-24 Osi Pharmaceuticals Inc. Bicyclic pyrimidinyl derivatives and methods of use thereof
AU2003266668A1 (en) 2002-09-30 2004-04-23 Banyu Pharmaceutical Co., Ltd. 2-aminobenzimidazole derivative
JP2004161716A (en) 2002-11-15 2004-06-10 Takeda Chem Ind Ltd Jnk inhibitor
UA80171C2 (en) 2002-12-19 2007-08-27 Pfizer Prod Inc Pyrrolopyrimidine derivatives
DE60335911D1 (en) 2002-12-20 2011-03-10 Daiichi Sankyo Co Ltd ISOCHINOLINE DERIVATIVES AND THEIR USE AS MEDICAMENTS
US7071355B2 (en) 2002-12-23 2006-07-04 4 Sc Ag Compounds as anti-inflammatory, immunomodulatory and anti-proliferatory agents
US7247736B2 (en) 2002-12-23 2007-07-24 4Sc Ag Method of identifying inhibitors of DHODH
US7365094B2 (en) 2002-12-23 2008-04-29 4Sc Ag Compounds as anti-inflammatory, immunomodulatory and anti-proliferatory agents
JP2006521373A (en) 2003-03-06 2006-09-21 デーエスエム アイピー アセッツ ベー. ヴェー. Method for preparing α-aminocarbonyl compound
WO2004087053A2 (en) 2003-03-25 2004-10-14 Syrrx, Inc. Dipeptidyl peptidase inhibitors
GB0306907D0 (en) 2003-03-26 2003-04-30 Angiogene Pharm Ltd Boireductively-activated prodrugs
WO2004103270A2 (en) 2003-04-02 2004-12-02 Suntory Pharmaceutical Research Laboratories Llc Compounds and methods for treatment of thrombosis
WO2004111014A1 (en) 2003-06-06 2004-12-23 Vertex Pharmaceuticals Incorporated Pyrimidine derivatives as modulators of atp-binding cassette transporters
US7429596B2 (en) 2003-06-20 2008-09-30 The Regents Of The University Of California 1H-pyrrolo [2,3-D] pyrimidine derivatives and methods of use thereof
CA2572324A1 (en) 2003-07-02 2005-01-13 Warner-Lambert Company Llc Combination of an allosteric inhibitor of matrix metalloproteinase-13 and a ligand to an alpha-2-delta receptor
EP1648455A4 (en) 2003-07-23 2009-03-04 Exelixis Inc Anaplastic lymphoma kinase modulators and methods of use
JP2007500725A (en) 2003-07-29 2007-01-18 アイアールエム・リミテッド・ライアビリティ・カンパニー Compounds and compositions as protein kinase inhibitors
GB0317951D0 (en) 2003-07-31 2003-09-03 Trigen Ltd Compounds
WO2005014534A1 (en) 2003-08-08 2005-02-17 Transtech Pharma, Inc. Aryl and heteroaryl compounds, compositions, and methods of use
US7208601B2 (en) 2003-08-08 2007-04-24 Mjalli Adnan M M Aryl and heteroaryl compounds, compositions, and methods of use
US20050043239A1 (en) 2003-08-14 2005-02-24 Jason Douangpanya Methods of inhibiting immune responses stimulated by an endogenous factor
WO2005016349A1 (en) 2003-08-14 2005-02-24 Icos Corporation Methods of inhibiting leukocyte accumulation
AU2004264419B2 (en) 2003-08-15 2009-01-15 Irm Llc 6-substituted anilino purines as RTK inhibitors
US7390820B2 (en) 2003-08-25 2008-06-24 Amgen Inc. Substituted quinolinone derivatives and methods of use
WO2005044181A2 (en) 2003-09-09 2005-05-19 Temple University-Of The Commonwealth System Of Higher Education Protection of tissues and cells from cytotoxic effects of ionizing radiation by abl inhibitors
NZ546611A (en) 2003-09-18 2010-02-26 Conforma Therapeutics Corp Novel heterocyclic compounds as HSP90-inhibitors
GB0322409D0 (en) 2003-09-25 2003-10-29 Astrazeneca Ab Quinazoline derivatives
MXPA06003996A (en) 2003-10-08 2006-07-05 Irm Llc Compounds and compositions as protein kinase inhibitors.
TWI344364B (en) 2003-11-10 2011-07-01 Synta Pharmaceuticals Corp Fused heterocyclic compounds
BRPI0416656A (en) 2003-11-17 2007-01-16 Pfizer Prod Inc pyrrolopyrimidine compounds useful in cancer treatment
AR046845A1 (en) 2003-11-21 2005-12-28 Novartis Ag DERIVATIVES OF 1H-IMIDAZO [4,5-C] QUINOLINE FOR THE TREATMENT OF PROTEIN-KINASE DEPENDENT DISEASES
JP2007513154A (en) 2003-12-08 2007-05-24 サイトキネティクス・インコーポレーテッド Compounds, compositions and methods
MXPA06007095A (en) 2003-12-22 2006-09-04 Gilead Sciences Inc Kinase inhibitor phosphonate conjugates.
BRPI0418112A (en) 2003-12-23 2007-04-17 Novartis Ag heterocyclic bicyclic p-38 kinase inhibitors
US20050239809A1 (en) 2004-01-08 2005-10-27 Watts Stephanie W Methods for treating and preventing hypertension and hypertension-related disorders
JP2007520559A (en) 2004-02-03 2007-07-26 アボット・ラボラトリーズ Aminobenzoxazoles as therapeutic agents
BRPI0507629A (en) 2004-02-13 2007-07-03 Banyu Pharma Co Ltd compound, receptor antagonist and h3 histamine receptor inverse agonist, prophylactic or therapeutic agent for a metabolic disease, a circulatory disease or a nervous system disease, or a pharmaceutically acceptable salt thereof
US20050187418A1 (en) 2004-02-19 2005-08-25 Small Brooke L. Olefin oligomerization
JP2007523202A (en) 2004-02-24 2007-08-16 ビオアクソン・テラプティーク・インコーポレーテッド 4-substituted piperidine derivatives
BRPI0508220A (en) 2004-02-27 2007-07-17 Hoffmann La Roche fused heteroaryl pyrazole derivatives
WO2005085249A1 (en) 2004-02-27 2005-09-15 F. Hoffmann-La Roche Ag Fused derivatives of pyrazole
EP2497836B1 (en) 2004-03-02 2015-01-14 The Johns Hopkins University Mutations of the PIK3CA gene in human cancers
EP1740591B1 (en) 2004-04-02 2009-06-17 OSI Pharmaceuticals, Inc. 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitors
US20070293489A1 (en) 2004-04-02 2007-12-20 Jerry Adams Substituted Pyrazolopyrimidines
BRPI0510305A (en) 2004-04-30 2007-10-02 Takeda Pharmaceutical compound or a salt thereof, prodrug or a salt thereof, pharmaceutical agent, method of producing the compound or a salt thereof, matrix metalloproteinase inhibitor or a salt thereof or a prodrug thereof, method of inhibiting a metalloproteinase matrix, and use of a compound or salt thereof or a prodrug thereof
DE102004022897A1 (en) 2004-05-10 2005-12-08 Bayer Cropscience Ag Azinyl-imidazoazines
SI2612862T1 (en) 2004-05-13 2017-04-26 Icos Corporation Quinazolinones as inhibitors of human phosphatidylinositol 3-kinase delta
WO2005112935A1 (en) 2004-05-13 2005-12-01 Vanderbilt University Phosphoinositide 3-kinase delta selective inhibitors for inhibiting angiogenesis
EP1755609A1 (en) 2004-05-25 2007-02-28 Icos Corporation Methods for treating and/or preventing aberrant proliferation of hematopoietic cells
EP1750715A1 (en) 2004-06-04 2007-02-14 Icos Corporation Methods for treating mast cell disorders
GB0420722D0 (en) 2004-09-17 2004-10-20 Addex Pharmaceuticals Sa Novel allosteric modulators
WO2006038865A1 (en) 2004-10-01 2006-04-13 Betagenon Ab Nucleotide derivatives for the treatment of type 2 diabetes and other disorders
EP2343303A1 (en) 2004-10-07 2011-07-13 Boehringer Ingelheim International GmbH PI3-kinase inhibitors
US7622451B2 (en) 2004-11-03 2009-11-24 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
US8212011B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues
US7608594B2 (en) 2004-11-03 2009-10-27 University Of Kansas Novobiocin analogues as anticancer agents
US8212012B2 (en) 2004-11-03 2012-07-03 University Of Kansas Novobiocin analogues having modified sugar moieties
GB0425035D0 (en) 2004-11-12 2004-12-15 Novartis Ag Organic compounds
EP1831225A2 (en) 2004-11-19 2007-09-12 The Regents of the University of California Anti-inflammatory pyrazolopyrimidines
WO2006065480A2 (en) 2004-11-23 2006-06-22 Ptc Therapeutics, Inc. Tetrahydrocarbazoles as active agents for inhibiting vegf production by translational control
TW200637843A (en) 2005-01-10 2006-11-01 Bristol Myers Squibb Co Phenylglycinamide derivatives useful as anticoagulants
US20060156485A1 (en) 2005-01-14 2006-07-20 The Procter & Gamble Company Keratin dyeing compounds, keratin dyeing compositions containing them, and use thereof
PE20061119A1 (en) 2005-01-19 2006-11-27 Aventis Pharma Sa PYRAZOLO PYRIDINES SUBSTITUTED AS INHIBITORS OF FAK, KDR AND Tie KINASES
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
CA2598409A1 (en) 2005-02-17 2006-08-24 Icos Corporation Phosphoinositide 3-kinase inhibitors for inhibiting leukocyte accumulation
US7579348B2 (en) 2005-02-25 2009-08-25 Pgxhealth, Llc Derivatives of 8-substituted xanthines
US20090124654A1 (en) 2005-03-01 2009-05-14 Mjalli Adnan M M Aryl and Heteroaryl Compounds, Compositions, Methods of Use
CA2603250A1 (en) 2005-04-06 2006-10-12 Irm Llc Diarylamine-containing compounds and compositions, and their use as modulators of steroid hormone nuclear receptors
KR100781704B1 (en) 2005-04-20 2007-12-03 에스케이케미칼주식회사 Novel pyridine derivatives, process for preparing thereof and pharmaceutical compositions containing them
EP1871754A1 (en) 2005-04-22 2008-01-02 Wyeth a Corporation of the State of Delaware Dihydrobenzofuran derivatives and uses thereof
WO2006114064A2 (en) 2005-04-25 2006-11-02 Institute Of Organic Chemistry And Biochemistry Ofthe Academy Of Sciences Of The Czech Republic Use of compounds to enhance processivity of telomerase
WO2007002667A2 (en) 2005-06-22 2007-01-04 Chemocentryx, Inc. Azaindazole compounds and methods of use
US7799795B2 (en) 2005-06-27 2010-09-21 Amgen Inc. Aryl nitrile compounds and compositions and their uses in treating inflammatory and related disorders
US20100168102A9 (en) 2005-07-11 2010-07-01 Devgen Nv Amide Derivatives as Kinase Inhibitors
US20100190788A1 (en) 2005-07-11 2010-07-29 Olivier Defert Amide derivatives as kinase inhitors
GB0516723D0 (en) 2005-08-15 2005-09-21 Novartis Ag Organic compounds
JP2009506009A (en) 2005-08-25 2009-02-12 エフ.ホフマン−ラ ロシュ アーゲー p38MAP kinase inhibitor and method of using the same
JP2009506006A (en) 2005-08-25 2009-02-12 エフ.ホフマン−ラ ロシュ アーゲー p38 MAP kinase inhibitor and method of using the same
WO2007025090A2 (en) 2005-08-25 2007-03-01 Kalypsys, Inc. Heterobicyclic and - tricyclic inhibitors of mapk/erk kinase
WO2007025534A1 (en) 2005-09-01 2007-03-08 Bioagency Ag Fredericamycin derivatives
CN101309904A (en) 2005-09-29 2008-11-19 惠氏公司 1- (1h- indol- 1-yl) -3- (methylamino) -1- phenylpropan-2-ol derivatives and related compounds as modulators of the monoamine reuptake for the treatment of vasomotor symptoms (vms)
KR20080078668A (en) 2005-11-17 2008-08-27 오에스아이 파마슈티컬스, 인코포레이티드 Fused bicyclic mtor inhibitors
GB0523654D0 (en) 2005-11-21 2005-12-28 Novartis Ag Organic compounds
US20090088452A1 (en) 2005-11-22 2009-04-02 Coleman Paul J Indole Orexin Receptor Antagonists
AR057960A1 (en) 2005-12-02 2007-12-26 Osi Pharm Inc BICYCLE PROTEIN QUINASE INHIBITORS
US20080299113A1 (en) 2005-12-19 2008-12-04 Arnold Lee D Combined treatment with and composition of 6,6-bicyclic ring substituted heterobicyclic protein kinase inhibitor and anti-cancer agents
US7772231B2 (en) 2005-12-29 2010-08-10 Abbott Laboratories Substituted pyrazolo[3,4-d]pyrimidines as protein kinase inhibitors
EP1981890A2 (en) 2006-01-25 2008-10-22 OSI Pharmaceuticals, Inc. UNSATURATED mTOR INHIBITORS
US20080058521A1 (en) 2006-01-26 2008-03-06 Wyeth Processes for the preparation of compounds
PE20070978A1 (en) 2006-02-14 2007-11-15 Novartis Ag HETEROCICLIC COMPOUNDS AS INHIBITORS OF PHOSPHATIDYLINOSITOL 3-KINASES (PI3Ks)
BRPI0708615A2 (en) 2006-03-07 2011-06-07 Array Biopharma Inc heterobicyclic pyrazole compounds and methods of use
WO2007106503A2 (en) 2006-03-13 2007-09-20 Osi Pharmaceuticals, Inc. Combined treatment with an egfr kinase inhibitor and an agent that sensitizes tumor cells to the effects of egfr kinase inhibitors
EP2650305A1 (en) 2006-03-24 2013-10-16 Biogen Idec Hemophilia Inc. PC5 as a factor IX propeptide processing enzyme
EP2007373A4 (en) 2006-03-29 2012-12-19 Foldrx Pharmaceuticals Inc Inhibition of alpha-synuclein toxicity
KR20150038395A (en) 2006-04-04 2015-04-08 더 리젠트스 오브 더 유니이버시티 오브 캘리포니아 Kinase antagonists
WO2007121453A2 (en) 2006-04-17 2007-10-25 The Regents Of The University Of California 2-hydroxy-1-oxo 1,2 dihydro isoquinoline chelating agents
GB0607950D0 (en) 2006-04-21 2006-05-31 Novartis Ag Organic compounds
PL2013211T3 (en) 2006-04-21 2012-08-31 Novartis Ag Purine derivatives for use as adenosin a2a receptor agonists
CA2649379C (en) 2006-04-21 2016-08-23 Shell Internationale Research Maatschappij B.V. Heating systems and methods using high strength alloys
GB0607948D0 (en) 2006-04-21 2006-05-31 Novartis Ag Organic compounds
JP2009536620A (en) 2006-04-25 2009-10-15 アステックス、セラピューティックス、リミテッド Pharmaceutical combination
WO2007125315A2 (en) 2006-04-25 2007-11-08 Astex Therapeutics Limited Pharmaceutical compounds
DE102006020327A1 (en) 2006-04-27 2007-12-27 Bayer Healthcare Ag Heterocyclic substituted, fused pyrazole derivatives and their use
WO2007135398A1 (en) 2006-05-22 2007-11-29 Astrazeneca Ab Indole derivatives
GB0610242D0 (en) 2006-05-23 2006-07-05 Novartis Ag Organic compounds
US20080003254A1 (en) 2006-05-23 2008-01-03 Abbott Laboratories Systems and methods for delivering a rapamycin analog that do not inhibit human coronary artery endothelial cell migration
GB0610317D0 (en) 2006-05-24 2006-07-05 Medical Res Council Antiparasitic compounds and compositions
DE602007006835D1 (en) 2006-07-20 2010-07-08 Amgen Inc SUBSTITUTED PYRIDONE COMPOUNDS AND APPLICATION METHODS
ES2379830T3 (en) 2006-07-28 2012-05-04 Novartis Ag 2,4-substituted quinazolines as lipid kinase inhibitors
CA2661307C (en) 2006-08-22 2016-07-19 Technion Research And Development Foundation Ltd. Heterocyclic derivatives binding to the peripheral-type benzodiazepine receptor (pbr)
WO2008024977A2 (en) 2006-08-24 2008-02-28 Serenex, Inc. Isoquinoline, quinazoline and phthalazine derivatives
WO2008025755A1 (en) 2006-09-01 2008-03-06 Basf Se Use of n-containing heterocycles in dermocosmetics
CA2974246C (en) 2006-09-01 2020-02-25 Senhwa Biosciences, Inc. Tricyclic heteroaryl compounds and their use as protein modulators
EP1903044A1 (en) 2006-09-14 2008-03-26 Novartis AG Adenosine Derivatives as A2A Receptor Agonists
EP1900729A1 (en) 2006-09-15 2008-03-19 Novartis AG Benzoxazoles and oxazolopyridines being useful as Janus kinases inhibitors
EP2081933B1 (en) 2006-09-29 2011-03-23 Novartis AG Pyrazolopyrimidines as pi3k lipid kinase inhibitors
US8492405B2 (en) 2006-10-18 2013-07-23 Takeda Pharmaceutical Company Limited Glucokinase-activating fused heterocyclic compounds and methods of treating diabetes and obesity
US7772180B2 (en) 2006-11-09 2010-08-10 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
CA2669399A1 (en) 2006-11-13 2008-05-29 Eli Lilly & Co. Thienopyrimidinones for treatment of inflammatory disorders and cancers
WO2008063625A2 (en) 2006-11-20 2008-05-29 Adolor Corporation Pyridine compounds and methods of their use
CA2672940A1 (en) 2006-12-20 2008-07-10 Schering Corporation Novel jnk inhibitors
ATE553105T1 (en) 2006-12-22 2012-04-15 Ind Res Ltd AZETIDINE ANALOGUES AIS NUCLEOSIDASE AND PHOSPHORYLASE INHIBITORS
WO2008083070A1 (en) 2006-12-29 2008-07-10 Neurogen Corporation Crf1 receptor ligands comprising fused bicyclic heteroaryl moieties
US20100056494A1 (en) 2007-01-26 2010-03-04 Irm Llc Purine compounds and compositions as kinase inhibitors for the treatment of plasmodium related diseases
WO2008112715A2 (en) 2007-03-12 2008-09-18 Vm Discovery Inc. Novel agents of calcium ion channel modulators
US20080234262A1 (en) 2007-03-21 2008-09-25 Wyeth Pyrazolopyrimidine analogs and their use as mtor kinase and pi3 kinase inhibitors
WO2008118454A2 (en) 2007-03-23 2008-10-02 Amgen Inc. Derivatives of quinoline or benzopyrazine and their uses for the treatment of (inter alia) inflammatory diseases, autoimmune diseases or various kinds of cancer
RS54706B1 (en) 2007-03-23 2016-08-31 Amgen Inc. Heterocyclic compounds and their uses
AU2008231385B2 (en) 2007-03-23 2012-02-02 Amgen Inc. Delta3- substituted quinoline or quinoxaline derivatives and their use as phosphatidylinositol 3-kinase ( PI3K) inhibitors
WO2008156513A2 (en) 2007-03-29 2008-12-24 University Of Connecticut Methods to protect skeletal muscle against injury
WO2008124026A1 (en) * 2007-04-05 2008-10-16 Wyeth Wortmannin-rapamycin conjugate and uses thereof
KR20100015546A (en) 2007-04-13 2010-02-12 사노피-아벤티스 A transition metal catalyzed synthesis of n-aminoindoles
WO2008125014A1 (en) 2007-04-13 2008-10-23 Institute Of Pharmacology And Toxicology Academy Of Military Medical Sciences P.L.A. Urea compounds, preparation methods and pharmaceutical uses thereof
JP2010163361A (en) 2007-04-27 2010-07-29 Dainippon Sumitomo Pharma Co Ltd Quinoline derivative
US7960353B2 (en) 2007-05-10 2011-06-14 University Of Kansas Novobiocin analogues as neuroprotective agents and in the treatment of autoimmune disorders
WO2008147852A1 (en) 2007-05-22 2008-12-04 Taigen Biotechnology Co., Ltd. Kinesin inhibitors
US9603848B2 (en) 2007-06-08 2017-03-28 Senomyx, Inc. Modulation of chemosensory receptors and ligands associated therewith
US7928111B2 (en) 2007-06-08 2011-04-19 Senomyx, Inc. Compounds including substituted thienopyrimidinone derivatives as ligands for modulating chemosensory receptors
WO2008152394A1 (en) 2007-06-12 2008-12-18 F.Hoffmann-La Roche Ag Pharmaceutical compounds
ATE538120T1 (en) 2007-06-26 2012-01-15 Sanofi Sa REGIOSelective, METAL-CATALYzed SYNTHESIS OF ANNATED BENZIMIDAZOLES AND AZABENZIMIDAZOLES
WO2009004621A1 (en) 2007-07-02 2009-01-08 Technion Research & Development Foundation Ltd. Compositions, articles and methods comprising tspo ligands for preventing or reducing tobacco-associated damage
JP2010532756A (en) 2007-07-06 2010-10-14 オーエスアイ・ファーマスーティカルズ・インコーポレーテッド Combination anti-cancer therapy comprising inhibitors of both mTORC1 and mTORC2
RU2345996C1 (en) 2007-07-17 2009-02-10 Андрей Александрович Иващенко Annelated azaheterocyclic amides, which include pyrimidine fragment, method of obtaining and their application
CA2694136C (en) * 2007-08-02 2013-09-17 Amgen Inc. Pi3 kinase modulators and methods of use
WO2009042294A2 (en) 2007-08-10 2009-04-02 Burnham Institute For Medical Research Tissue-nonspecific alkaline phosphatase (tnap) activators and uses thereof
CA2695583A1 (en) 2007-08-13 2009-02-19 Metabasis Therapeutics, Inc. Novel activators of glucokinase
EP2176259B1 (en) 2007-08-14 2011-03-30 Bayer Schering Pharma Aktiengesellschaft Fused imidazoles for cancer treatment
WO2009044707A1 (en) 2007-10-03 2009-04-09 Riken Nitrotriazole derivative and method for producing compound using the same
WO2009046448A1 (en) 2007-10-04 2009-04-09 Intellikine, Inc. Chemical entities and therapeutic uses thereof
KR20100089082A (en) 2007-10-15 2010-08-11 아스트라제네카 아베 Combination 059
JP5256693B2 (en) 2007-10-31 2013-08-07 信越化学工業株式会社 Production method of titanium oxide photocatalyst thin film
CA2699312A1 (en) 2007-11-02 2009-05-07 Taiga Biotechnologies, Inc. Compounds for treating abnormal cellular proliferation
EA201070572A1 (en) 2007-11-07 2010-12-30 Фолдркс Фармасьютикалз, Инк. MODULATION OF TRANSPORT OF PROTEINS
GEP20125635B (en) 2007-11-13 2012-09-10 Icos Corp Inhibitors of human phosphatidyl-inositol 3-kinase delta
US20090163481A1 (en) 2007-12-13 2009-06-25 Murphy Brian J Ppar-delta ligands and methods of their use
US8153813B2 (en) 2007-12-20 2012-04-10 Abbott Laboratories Benzothiazole and benzooxazole derivatives and methods of use
US7960397B2 (en) 2007-12-28 2011-06-14 Institute Of Experimental Botany, Academy Of Sciences Of The Czech Republic 6,9-disubstituted purine derivatives and their use as cosmetics and cosmetic compositions
US8193182B2 (en) 2008-01-04 2012-06-05 Intellikine, Inc. Substituted isoquinolin-1(2H)-ones, and methods of use thereof
NZ702041A (en) 2008-01-04 2016-03-31 Intellikine Llc Heterocyclic containing entities, compositions and methods
JP2011509305A (en) 2008-01-09 2011-03-24 ピージーエックスヘルス、リミテッド、ライアビリティー、カンパニー Intrathecal treatment of neuropathic pain with A2AR agonists
US20090192176A1 (en) 2008-01-30 2009-07-30 Wyeth 1H-PYRAZOLO[3,4-D]PYRIMIDINE, PURINE, 7H-PURIN-8(9H)-ONE, 3H-[1,2,3]TRIAZOLO[4,5-D]PYRIMIDINE, AND THIENO[3,2-D]PYRIMIDINE COMPOUNDS, THEIR USE AS mTOR KINASE AND PI3 KINASE INHIBITORS, AND THEIR SYNTHESES
JP5608099B2 (en) 2008-01-30 2014-10-15 ジェネンテック, インコーポレイテッド Pyrazolopyrimidine PI3K inhibitor compounds and methods of use
JP2011511802A (en) 2008-02-07 2011-04-14 ギリアード・パロ・アルト・インコーポレイテッド Compounds that increase ABCA-1 and methods of using such compounds
EP2244709A4 (en) 2008-02-07 2012-02-29 Synta Pharmaceuticals Corp Topical formulations for the treatment of psoriasis
TWI444384B (en) 2008-02-20 2014-07-11 Gilead Sciences Inc Nucleotide analogues and their use in the treatment of malignancies
US8993580B2 (en) 2008-03-14 2015-03-31 Intellikine Llc Benzothiazole kinase inhibitors and methods of use
EP2252293B1 (en) 2008-03-14 2018-06-27 Intellikine, LLC Kinase inhibitors and methods of use
WO2009117157A1 (en) 2008-03-20 2009-09-24 Amgen Inc. Aurora kinase modulators and method of use
US20090312406A1 (en) 2008-06-12 2009-12-17 Hsing-Pang Hsieh Coumarin compounds and their use for treating viral infection
EP2303890A4 (en) 2008-06-19 2012-04-11 Progenics Pharm Inc Phosphatidylinositol 3 kinase inhibitors
CN102124009B (en) * 2008-07-08 2014-07-23 因特利凯公司 Kinase inhibitors and methods of use
US20110224223A1 (en) 2008-07-08 2011-09-15 The Regents Of The University Of California, A California Corporation MTOR Modulators and Uses Thereof
JP2011528368A (en) 2008-07-16 2011-11-17 シェーリング コーポレイション Bicyclic heterocyclic derivatives and their use as GPCR modulators
US8450344B2 (en) 2008-07-25 2013-05-28 Aerie Pharmaceuticals, Inc. Beta- and gamma-amino-isoquinoline amide compounds and substituted benzamide compounds
AU2009282480B2 (en) 2008-08-11 2015-05-07 Children's Medical Center Corporation Halofuginone analogs for inhibition of tRNA synthetases and uses thereof
US8513295B2 (en) 2008-09-23 2013-08-20 Georgetown University Viral and fungal inhibitors
JP5731978B2 (en) 2008-09-26 2015-06-10 インテリカイン, エルエルシー Heterocyclic kinase inhibitor
TW201018685A (en) * 2008-09-30 2010-05-16 Exelixis Inc Pyridopyrimidinone inhibitors of PI3Kα and mTOR
AU2009305669A1 (en) 2008-10-16 2010-04-22 The Regents Of The University Of California Fused ring heteroaryl kinase inhibitors
US8476282B2 (en) * 2008-11-03 2013-07-02 Intellikine Llc Benzoxazole kinase inhibitors and methods of use
WO2010059593A1 (en) 2008-11-18 2010-05-27 Intellikine, Inc. Methods and compositions for treatment of ophthalmic conditions
WO2010088050A2 (en) 2009-01-28 2010-08-05 Cara Therapeutics, Inc. Bicyclic pyrazolo-heterocycles
WO2010099139A2 (en) 2009-02-25 2010-09-02 Osi Pharmaceuticals, Inc. Combination anti-cancer therapy
ES2797523T3 (en) 2009-03-23 2020-12-02 Lilly Co Eli Imaging agents to detect neurological disorders
WO2010118367A2 (en) 2009-04-10 2010-10-14 Progenics Pharmaceuticals, Inc. Antiviral pyrimidines
JP5789252B2 (en) 2009-05-07 2015-10-07 インテリカイン, エルエルシー Heterocyclic compounds and uses thereof
CN101602768B (en) 2009-07-17 2012-05-30 河南省农科院农副产品加工研究所 Method for purifying sesamin and sesamolin
PL2470546T3 (en) 2009-08-28 2013-12-31 Takeda Pharmaceuticals Co Hexahydrooxazinopteridine compounds for use as mtor inhibitors
TWI499592B (en) 2009-09-09 2015-09-11 Avila Therapeutics Inc Pi3 kinase inhibitors and uses thereof
WO2011047384A2 (en) 2009-10-16 2011-04-21 The Regents Of The University Of California Methods of inhibiting ire1
WO2011149937A1 (en) 2010-05-24 2011-12-01 Intellikine, Inc. Heterocyclic compounds and uses thereof

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010049481A1 (en) * 2008-10-31 2010-05-06 Novartis Ag Combination of a phosphatidylinositol-3-kinase (pi3k) inhibitor and a mtor inhibitor
WO2011022439A1 (en) * 2009-08-17 2011-02-24 Intellikine, Inc. Heterocyclic compounds and uses thereof

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160089371A1 (en) * 2013-03-15 2016-03-31 Intellikine, Llc Combination of Kinase Inhibitors and Uses Thereof
US11352328B2 (en) 2016-07-12 2022-06-07 Arisan Therapeutics Inc. Heterocyclic compounds for the treatment of arenavirus
WO2018237370A1 (en) * 2017-06-23 2018-12-27 Accro Bioscience Inc. Heteroaryl compounds as inhibitors of necrosis, composition and method using the same
CN109111464A (en) * 2017-06-23 2019-01-01 爱科诺生物医药股份有限公司 A kind of heterocyclic compound with meronecrosis inhibitory activity

Also Published As

Publication number Publication date
CN106619647A (en) 2017-05-10
TWI592411B (en) 2017-07-21
JP6130305B2 (en) 2017-05-17
AR085397A1 (en) 2013-09-25
US9295673B2 (en) 2016-03-29
EP2678018A1 (en) 2014-01-01
WO2012148540A8 (en) 2013-09-26
CN103491962B (en) 2016-10-12
CN103491962A (en) 2014-01-01
JP2014506600A (en) 2014-03-17
WO2012148540A1 (en) 2012-11-01
US20150087627A1 (en) 2015-03-26
EP2678018A4 (en) 2015-09-30
TW201302737A (en) 2013-01-16
CA2828483A1 (en) 2012-11-01
US20120294930A1 (en) 2012-11-22

Similar Documents

Publication Publication Date Title
US9295673B2 (en) Combination of mTOR inhibitors and P13-kinase inhibitors, and uses thereof
US11547697B2 (en) Heterocyclic compounds and uses thereof
US9682141B2 (en) Combination of kinase inhibitors and uses thereof
US10172858B2 (en) Combination pharmaceutical compositions and uses thereof
US9127000B2 (en) Heterocyclic compounds and uses thereof
US9359352B2 (en) Substituted benzimidazoles as PI3 kinase inhibitors
US20170209448A1 (en) Treatment regimens using multiple pharmaceutical agents
EP2968340A1 (en) Combination of kinase inhibitors and uses thereof
US20160089371A1 (en) Combination of Kinase Inhibitors and Uses Thereof
WO2012154608A1 (en) Reactive mtor and pi3 kinase inhibitors and uses thereof
US20140323721A1 (en) Heterocyclic compounds and uses thereof
WO2012154610A1 (en) Reactive pi3k kinase inhibitors and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: INTELLIKINE LLC, CALIFORNIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:REN, PINGDA;LIU, YI;JESSEN, KATAYOUN;AND OTHERS;SIGNING DATES FROM 20120822 TO 20120920;REEL/FRAME:038442/0743

STCB Information on status: application discontinuation

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