WO2015181624A2 - Nucleoside derivatives for the treatment of cancer - Google Patents

Nucleoside derivatives for the treatment of cancer Download PDF

Info

Publication number
WO2015181624A2
WO2015181624A2 PCT/IB2015/000957 IB2015000957W WO2015181624A2 WO 2015181624 A2 WO2015181624 A2 WO 2015181624A2 IB 2015000957 W IB2015000957 W IB 2015000957W WO 2015181624 A2 WO2015181624 A2 WO 2015181624A2
Authority
WO
WIPO (PCT)
Prior art keywords
amino
compound
alkyl
hydrogen
methoxy
Prior art date
Application number
PCT/IB2015/000957
Other languages
French (fr)
Other versions
WO2015181624A3 (en
Inventor
Cyril Dousson
David Dukhan
Christophe Claude Parsy
François-René ALEXANDRE
Rachid Rahali
Jean-Laurent Paparin
Original Assignee
Idenix Pharmaceuticals, Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Idenix Pharmaceuticals, Inc filed Critical Idenix Pharmaceuticals, Inc
Priority to JP2016569379A priority Critical patent/JP2017516779A/en
Priority to US15/313,646 priority patent/US20170101431A1/en
Priority to CN201580028114.XA priority patent/CN106459127A/en
Priority to RU2016149761A priority patent/RU2016149761A/en
Priority to CA2947939A priority patent/CA2947939A1/en
Priority to AU2015265607A priority patent/AU2015265607A1/en
Priority to EP15770630.0A priority patent/EP3149017B1/en
Priority to MX2016015568A priority patent/MX2016015568A/en
Priority to KR1020167036090A priority patent/KR20170005492A/en
Publication of WO2015181624A2 publication Critical patent/WO2015181624A2/en
Publication of WO2015181624A3 publication Critical patent/WO2015181624A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/20Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids
    • C07H19/213Purine radicals with the saccharide radical esterified by phosphoric or polyphosphoric acids containing cyclic phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/06Pyrimidine radicals
    • C07H19/067Pyrimidine radicals with ribosyl as the saccharide radical
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H19/00Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof
    • C07H19/02Compounds containing a hetero ring sharing one ring hetero atom with a saccharide radical; Nucleosides; Mononucleotides; Anhydro-derivatives thereof sharing nitrogen
    • C07H19/04Heterocyclic radicals containing only nitrogen atoms as ring hetero atom
    • C07H19/16Purine radicals
    • C07H19/167Purine radicals with ribosyl as the saccharide radical

Definitions

  • nucleoside derivatives Provided herein are nucleoside derivatives, pharmaceutical compositions comprising the compounds, processes of preparation thereof, and methods of use thereof for treating cancer.
  • Cancer is a disease characterized primarily by an uncontrolled divisions of abnormal cells derived from a given normal tissue and the invasion of adjacent tissues by these malignant cells. Blood or lymphatic transportation can spread cancer cells to other parts of the body leading to regional lymph nodes and to distant sites (metastasis). Cancer is a complex, multistep process that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia. There are more than 100 different types of cancer, which can be grouped into broader categories. The main categories include: carcinoma, sarcoma, leukemia, lymphoma and myeloma, and central nervous system cancers.
  • Hematologic or hematopoietic malignancies are cancers of the blood or bone marrow, including leukemia and lymphoma.
  • Leukemia is a type of cancer of the blood characterized by abnormal accumulation of immature white blood cells.
  • leukemia There are four types of leukemia: acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), and chronic myelogenous leukemia (CML).
  • ALL acute lymphocytic leukemia
  • AML acute myelogenous leukemia
  • CLL chronic lymphocytic leukemia
  • CML chronic myelogenous leukemia
  • Acute leukemia is a rapidly progressing disease that results in the accumulation of immature, functionless cells in the marrow and blood. The marrow often stops producing enough normal red cells, white cells and platelets.
  • chronic leukemia progresses more slowly and allows greater numbers of more mature, functional cells
  • Leukemia can affect people at any age. The cause of most cases of leukemia is not known. Extraordinary doses of radiation and certain cancer therapies are possible causes. About 90% of leukemia are diagnosed in adults. In 2000 approximately 256,000 children and adults around the world developed some form of leukemia, and 209,000 died from it. Cases of Chronic leukemia account for 4.5 percent more cases than acute leukemia. The most common types of leukemia in adults are acute myelogenous leukemia (AML), with estimated 14,590 new cases in 2013, and chronic lymphocytic leukemia (CLL), with about 15,680 new cases in 2013. Chronic myelogenous leukemia (CML) was estimated to affect about 5,920 persons in 2013 (data from the Leukemia and Lymphoma Society, Facts 2013, August 2013).
  • AML acute myelogenous leukemia
  • CLL chronic lymphocytic leukemia
  • phosphoramidate derivatives and cyclic phosphate derivatives of a variety of therapeutic agents are provided, as well as pharmaceutical compositions and methods of treatment of a variety of cancer including hematopoietic malignancies.
  • the therapeutic agent is, for example, an anti-cancer agent that includes, or has been derivatized to include, a reactive group, such as a hydroxyl, for attachment of the phosphoroamidate and cyclic phosphate moiety.
  • Such therapeutic agents include, but are not limited to nucleosides and nucleoside analogs including acyclic nucleosides.
  • a nucleoside analog refers to a structurally modified nucleoside.
  • the nucleosides or nucleoside analogs are derivitized at the 5'- and/or the 3 '-position by removal of a hydrogen from an hydroxyl group to incorporate a phosphoramidate and cyclic phosphate group. In one embodiment, the nucleosides or nucleoside analogs are derivitized at the 5'- and/or the 2'-position by removal of a hydrogen from an hydroxyl group to incorporate a phosphoramidate and cyclic phosphate group. In one embodiment, phosphorodiamidates of nucleosides and nucleoside analogs are provided.
  • the compound is a -S'-pivaloyl-2-thioethyl phosphoroamidate or S- hydroxypivaloyl-2-thioethyl phosphoroamidate.
  • ⁇ S'-pivaloyl-2- thioethyl disulfide phosphoroamidate or ⁇ -dimethyl-S-oxopropanoate 2-thioethyl benzyl phosphoroamidate or benzylphosphoramidate of nucleosides and nucleoside analogs are provided.
  • the derivative is l-methyl-2-nitro-lH- imidazol-5-methoxymethyl N- benzylphosphoramidate or 5-nitrofuran-2-methoxymethyl N-benzyl phosphoramidate.
  • the derivative is a 3', 5'-cyclic phosphate. In another embodiment the derivative is a 2', 5'-cyclic phosphate.
  • the anti-cancer agents include, but are not limited to, modified nucleosides.
  • the anti-cancer agent is clofarabine, cytarabine, isocladribine, cladribine, fludarabine or nelarabine.
  • the anti-cancer agent is clofarabine, cytarabine, isocladribine, cladribine or fludarabine.
  • the anti-cancer agent is clofarabine or cytarabine.
  • alkyl refers to a saturated straight or branched hydrocarbon.
  • the alkyl group is a primary, secondary, or tertiary hydrocarbon.
  • the alkyl group includes one to ten carbon atoms, i.e., to C 10 alkyl.
  • the alkyl group is methyl, CF 3 , CCI 3 , CFCI 2 , CF 2 C1, ethyl, CH 2 CF 3 , CF 2 CF 3 , propyl, isopropyl, butyl, isobutyl, sec-butyl, t- butyl, pentyl, isopentyl, neopentyl, hexyl, isohexyl, 3-methylpentyl, 2,2-dimethylbutyl, or 2,3-dimethylbutyl.
  • the term includes both substituted and unsubstituted alkyl groups, including halogenated alkyl groups, preferably unsubstituted or halogenated alkyl groups.
  • the alkyl group is a fluorinated alkyl group.
  • Non-limiting examples of moieties with which the alkyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic
  • lower alkyl refers to a saturated straight or branched hydrocarbon having one to six carbon atoms, i.e., Ci to C 6 alkyl.
  • the lower alkyl group is a primary, secondary, or tertiary hydrocarbon.
  • the term includes both substituted and unsubstituted moieties, preferably unsubstituted
  • cycloalkyl refers to a saturated cyclic hydrocarbon.
  • the cycloalkyl group may be a saturated, and/or bridged, and/or non-bridged, and/or a fused bicyclic group.
  • the cycloalkyl group includes three to ten carbon atoms, i.e., C 3 to C 10 cycloalkyl.
  • the cycloalkyl has from 3 to 15 (C 3 -15), from 3 to 10 (C 3 -10), or from 3 to 7 (C 3 -7) carbon atoms.
  • the cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cyclohexylmethyl, cycloheptyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1 ]heptyl, decalinyl, or adamantyl.
  • the term includes both substituted and unsubstituted cycloalkyl groups, including halogenated cycloalkyl groups.
  • Non-limiting examples of moieties with which the cycloalkyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Group
  • alkenyl refers to monovalent olefinically unsaturated hydrocarbon groups, in certain embodiment, having up to about 11 carbon atoms, from 2 to 8 carbon atoms, or from 2 to 6 carbon atoms, which can be straight-chained or branched and having at least 1 or from 1 to 2 sites of olefinic unsaturation.
  • alkenyl embraces radicals having a "CJV or "trans” configuration or a mixture thereof, or alternatively, a "Z” or “E” configuration or a mixture thereof, as appreciated by those of ordinary skill in the art.
  • C2-6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms.
  • the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C 2-6 ) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C 3 -20), 3 to 15 (C 3 -15), 3 to 10 (C 3 -10), or 3 to 6 (C 3 -6) carbon atoms.
  • alkenyl groups include, but are not limited to, ethenyl, propen-l-yl, propen-2-yl, allyl, butenyl, and 4-methylbutenyl.
  • the term includes both substituted and unsubstituted alkenylene groups, including halogenated cycloalkyl groups, preferably unsubstituted or halogenated cycloalkyl.
  • Non-limiting examples of moieties with which the alkenylene group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in
  • cycloalkenyl refers to an unsaturated cyclic hydrocarbon and includes both substituted and unsubstituted cycloalkenyl groups, preferably unsubstituted.
  • moieties with which the cycloalkenyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic
  • alkenylene refers to a linear or branched divalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one or two, carbon-carbon double bond(s).
  • alkenylene embraces radicals having a "c/V or "trans'” configuration or a mixture thereof, or alternatively, a "Z” or “E” configuration or a mixture thereof, as appreciated by those of ordinary skill in the art.
  • C 2- 6 alkenylene refers to a linear unsaturated divalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated divalent hydrocarbon radical of 3 to 6 carbon atoms.
  • the alkenylene is a linear divalent hydrocarbon radical of 2 to 20 (C 2-2 o), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched divalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C 3 -10), or 3 to 6 (C 3 -6) carbon atoms.
  • alkenylene groups include, but are not limited to, ethenylene, allylene, propenylene, butenylene, and 4-methylbutenylene. The term includes both substituted and unsubstituted groups, including halogenated groups, preferably unsubstituted.
  • Non-limiting examples of moieties with which the alkenylene group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxy carbonyl, aryloxy carbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al , Protective Groups in Organic
  • Alkynyl refers to acetylenically unsaturated hydrocarbon groups, in certain embodiments, having up to about 11 carbon atoms or from 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 or from 1 to 2 sites of alkynyl unsaturation.
  • alkynyl groups include acetylenic, ethynyl (- C ⁇ CH), propargyl (-CI3 ⁇ 4C ⁇ CH), and the like.
  • the term also includes both substituted and unsubstituted alkynyl groups, preferably unsubstituted.
  • Non-limiting examples of moieties with which the alkynyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al, Protective Groups in Organic
  • aryl refers to phenyl, biphenyl, or naphthyl. The term includes both substituted and unsubstituted moieties.
  • An aryl group can be substituted with any described moiety, including, but not limited to, one or more moieties selected from halogen (fluoro, chloro, bromo, or iodo), alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, cycloakenyl, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylary
  • Alkoxy refers to the group -OR' where R' is alkyl or cycloalkyl where alkyl and cycloalkyl are as defined herein. Alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
  • Aryloxy refers to the group -OR' where R' is aryl or heteroaryl where aryl and heteroaryl are as defined herein.
  • Alkoxycarbonyl refers to a radical -C(0)-alkoxy where alkoxy is as defined herein.
  • Amino refers to the radical -NH 2 .
  • Carboxyl or “carboxy” refers to the radical -C(0)OH.
  • Carbonyl refers to the radical - C(O)-.
  • alkylamino refers to an amino group that has one or two alkyl substituents (-NHR' or -NR'R', where R' is alkyl as defined herein), one or two aryl substituents (-NHR' or - NR'R', where R' is aryl as defined herein), or one alkyl substituent and one aryl substituent (-NR'R", where one of R' and R" is alkyl as defined herein and the other is aryl as defined herein), respectively.
  • the alkyl substituent is lower alkyl.
  • the alkyl or lower alkyl is unsubstituted.
  • Halogen or halo refers to fluoro, chloro, bromo, or iodo.
  • Thioalkoxy refers to the group -SR' where R' is alkyl or cycloalkyl each of which is as defined herein.
  • Thioaryloxy refers to the group -SR' where R' is aryl or heteroaryl each of which is as defined herein.
  • Alkyldisulfanyl refers to the group R'-S-S- where R' is alkyl or cycloalkyl each of which is as defined herein.
  • heterocyclyl refers to a monovalent monocyclic non- aromatic ring system and/or multicyclic ring system that contains at least one non- aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from O, S, or N; and the remaining ring atoms are carbon atoms.
  • the heterocyclyl or heterocyclic group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms.
  • Heterocyclyl groups are bonded to the rest of the molecule through the non-aromatic ring.
  • the heterocyclyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include a fused or bridged ring system, and in which the nitrogen or sulfur atoms may be optionally oxidized, the nitrogen atoms may be optionally quaternized, and some rings may be partially or fully saturated, or aromatic.
  • the heterocyclyl may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound.
  • heterocyclic radicals include, but are not limited to, azepinyl, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, ⁇ -carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydrofuryl, dihydroisoindolyl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1 ,4-dit
  • the term also includes both substituted and unsubstituted heterocyclyl groups.
  • moieties with which the heterocyclyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example
  • heteroaryl refers to a monovalent monocyclic aromatic group and/or multicyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, S, and N in the ring. Heteroaryl groups are bonded to the rest of the molecule through the aromatic ring.
  • Each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, and/or one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom.
  • the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms.
  • monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl.
  • bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl, thiadiazolopyrimi
  • tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl.
  • a heteroaryl group can be substituted with any described moiety, including, but not limited to, one or more moieties selected from halogen (fluoro, chloro, bromo, or iodo), oxo, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, cycloakenyl, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaryloxy, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or
  • alkylaryl refers to an aryl group with an alkyl substituent.
  • aralkyl or arylalkyl includes an alkyl group with an aryl substituent.
  • alkylheterocyclyl refers to a heterocyclyl group with an alkyl substituent.
  • alkylheterocyclyl includes an alkyl group with a heterocyclyl substituent.
  • alkylheteroaryl refers to a heteroaryl group with an alkyl substituent.
  • alkylheteroaryl includes an alkyl group with a heteroaryl substituent.
  • protecting group refers to a group that is added to an oxygen, nitrogen, or phosphorus atom to prevent its further reaction or for other purposes.
  • oxygen and nitrogen protecting groups are known to those skilled in the art of organic synthesis.
  • “Pharmaceutically acceptable salt” refers to any salt of a compound provided herein which retains its biological properties and which is not toxic or otherwise undesirable for pharmaceutical use. Such salts may be derived from a variety of organic and inorganic counter-ions well known in the art.
  • Such salts include, but are not limited to: (1) acid addition salts formed with organic or inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, sulfamic, acetic, trifluoroacetic, trichloroacetic, propionic, hexanoic, cyclopentylpropionic, glycolic, glutaric, pyruvic, lactic, malonic, succinic, sorbic, ascorbic, malic, maleic, fumaric, tartaric, citric, benzoic, 3-(4- hydroxybenzoyl)benzoic, picric, cinnamic, mandelic, phthalic, lauric, methanesulfonic, ethanesulfonic, 1,2-ethane-disulfonic, 2-hydroxyethanesulfonic, benzenesulfonic, 4- chlorobenzenesulfonic, 2-naphthalenesulfonic, 4-to
  • Pharmaceutically acceptable salts further include, by way of example only and without limitation, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like, and when the compound contains a basic functionality, salts of non-toxic organic or inorganic acids, such as hydrohalides, e.g.
  • purine or "pyrimidine” base refers to, but is not limited to, adenine, guanine, adenine, hypoxanthine, 7-deazaguanine, 7-deazaadenine, 2,6-diaminopurine, 6- chloropurine, N 6 -alkylpurines, N ⁇ acylpurines (wherein acyl is C(0)(alkyl, aryl, alkylaryl, or arylalkyl), N 6 -benzylpurine, 6-halopurine, N 6 -vinylpurine, N 6 -acetylenic purine, N 6 - hydroxyalkyl purine, N 6 -alkylaminopurine, N ⁇ thioalkyl purine, N 2 -alkylpurines, N 2 -alkyl- 6-thiopurines, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, including 6-azacytos
  • Suitable protecting groups are well known to those skilled in the art, and include trimethylsilyl, dimethylhexylsilyl, /-butyldimethylsilyl, and t- butyldiphenylsilyl, trityl, alkyl groups, acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
  • acyl or "O-linked ester” refers to a group of the formula O C(0)R', wherein R' is alkyl or cycloalkyl (including lower alkyl), carboxylate reside of amino acid, aryl including phenyl, alkaryl, arylalkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl.
  • R' can be substituted alkyl (including lower alkyl), aryl including phenyl optionally substituted with chloro, bromo, fluoro, iodo, Ci to C 4 alkyl or Ci to C 4 alkoxy, sulfonate esters such as alkyl or arylalkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxy-trityl, substituted benzyl, alkaryl, arylalkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl.
  • Aryl groups in the esters optimally comprise a phenyl group.
  • acyl groups include acetyl, trifluoroacetyl, methylacetyl, cyclpropylacetyl, propionyl, butyryl, hexanoyl, heptanoyl, octanoyl, neo-heptanoyl, phenylacetyl, 2-acetoxy-2-phenylacetyl, diphenylacetyl, a- methoxy-a-trifluoromethyl-phenylacetyl, bromoacetyl, 2-nitro-benzeneacetyl, 4-chloro- benzeneacetyl, 2-chloro-2,2-diphenylacetyl, 2-chloro-2 -phenylacetyl, trimethylacetyl, chlorodifluoroacetyl, perfluoroacetyl, fluoroacetyl, bromodi
  • amino acid refers to naturally occurring and synthetic ⁇ , ⁇ , ⁇ , or ⁇ amino acids, and includes but is not limited to, amino acids found in proteins, i.e. glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine.
  • the amino acid is in the L-configuration.
  • the amino acid can be a derivative of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, ⁇ - alanyl, ⁇ -valinyl, ⁇ -leucinyl, ⁇ -isoleuccinyl, ⁇ -prolinyl, ⁇ -phenylalaninyl, ⁇ -tryptophanyl, ⁇ -methioninyl, ⁇ -glycinyl, ⁇ -serinyl, ⁇ -threoninyl, ⁇ -cysteinyl
  • nucleoside composition that includes at least 85 or 90% by weight, in certain embodiments 95%, 98 % , 99%, or 100% by weight, of the designated nucleoside, the designated diastereomer of such nucleoside, or the designated enantiomer of such nucleoside.
  • the compounds are substantially free of other compounds, other nucleosides, other diastereomers, or other enantiomers that are not designated.
  • nucleoside composition refers to a nucleoside composition that includes at least 85, 90%, 95%, 98%, 99%, to 100% by weight, of the nucleoside, the remainder comprising other chemical species or enantiomers.
  • Solvate refers to a compound provided herein or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
  • Isotopic composition refers to the amount of each isotope present for a given atom
  • naturally occuring isotopic composition or abundance for a given atom Atoms containing their natural isotopic composition may also be referred to herein as "non-enriched" atoms. Unless otherwise designated, the atoms of the compounds recited herein are meant to represent any stable isotope of that atom. For example, unless otherwise stated, when a position is designated specifically as “H” or “hydrogen”, the position is understood to have hydrogen at its natural isotopic composition.
  • Isotopic enrichment refers to the percentage of incorporation of an amount of a specific isotope at a given atom in a molecule in the place of that atom's natural isotopic abundance. For example, deuterium enrichment of 1% at a given position means that 1% of the molecules in a given sample contain deuterium at the specified position. Because the naturally occurring distribution of deuterium is about 0.0156%, deuterium enrichment at any position in a compound synthesized using non-enriched starting materials is about 0.0156%.
  • the isotopic enrichment of the compounds provided herein can be determined using conventional analytical methods known to one of ordinary skill in the art, including mass spectrometry and nuclear magnetic resonance spectroscopy.
  • “Isotopically enriched” refers to an atom having an isotopic composition other than the natural isotopic composition of that atom. “Isotopically enriched” may also refer to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom.
  • alkyl As used herein, “alkyl,” “cycloalkyl,” “alkenyl,” “cycloalkenyl,” “alkynyl,” “aryl,” “alkoxy,” “alkoxycarbonyl,” “amino,” “carboxyl,” “alkylamino,” “arylamino,” “thioalkyoxy,” “heterocyclyl,” “heteroaryl,” “alkylheterocyclyl,” “alkylheteroaryl,” “acyl,” “aralkyl,” “alkaryl,” “purine,” “pyrimidine,” “carboxyl,” and “amino acid” groups optionally comprise deuterium at one or more positions where hydrogen atoms are present, and wherein the deuterium composition of the atom or atoms is other than the natural isotopic composition.
  • alkyl cycloalkyl
  • alkenyl cycloalkenyl
  • alkynyl aryl
  • alkoxy alkoxycarbonyl
  • carbboxyl alkylamino
  • arylamino thioalkyoxy
  • heterocyclyl heteroaryl
  • alkylheterocyclyl alkylheteroaryl
  • acyl amino acid
  • amino acid amino acid
  • alkyl alkyl
  • cycloalkyl alkenyl
  • cycloalkenyl alkynyl
  • aryl alkyl
  • heterocyclyl alkynyl
  • heteroaryl alkynyl
  • proliferative disorder or disease refers to unwanted cell proliferation of one or more subset of cells in a multicellular organism resulting in harm (i.e., discomfort or decreased life expectancy) to the multicellular organisms.
  • a proliferative disorder or disease can occur in different types of animals and humans.
  • proliferative disorder or disease includes neoplastic disorders and other proliferative disorders.
  • neoplastic disorder or disease refers to a tumor resulting from abnormal or uncontrolled cellular growth.
  • neoplastic disorders include, but are not limited to, hematopoietic disorders, such as the myeloproliferative disorders, thrombocythemia, essential thrombocytosis (ET), angiogenic myeloid metaplasia, myelofibrosis (MF), myelofibrosis with myeloid metaplasia (MMM), chronic idiopathic myelofibrosis (IMF), polycythemia vera (PV), the cytopenias, and pre-malignant myelodysplastic syndromes; cancers, such as glioma cancers, lung cancers, breast cancers, colorectal cancers, prostate cancers, gastric cancers, esophageal cancers, colon cancers, pancreatic cancers, ovarian cancers, and hematologic malignancies.
  • hematopoietic disorders such as the
  • hematologic malignancy refers to cancer of the body's blood-forming and immune system-the bone marrow and lymphatic tissue.
  • hematological malignancies include, for instance, myelodysplasia, lymphomas, leukemias, lymphomas (non-Hodgkin's lymphoma), Hodgkin's disease (also called Hodgkin's lymphoma), and myeloma, such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic neutrophilic leukemia (CNL), acute undifferentiated leukemia (AUL), anaplastic large-cell lymphoma (ALCL), prolymphocytic leukemia (PML), juvenile myelomonocyctic leukemia (JMML), adult T-cell ALL, AML
  • leukemia refers to malignant neoplasms of the blood-forming tissues, including, but not limited to, chronic lymphocytic leukemia, chronic myelocytic leukemia, acute lymphoblastic leukemia, acute myeloid leukemia and acute myeloblastic leukemia.
  • the leukemia can be relapsed, refractory, or resistant to conventional therapy.
  • relapsed refers to a situation where a subject or a mammal, who has had a remission of cancer after therapy, has a return of cancer cells.
  • refractory or resistant refers to a circumstance where a subject or a mammal, even after intensive treatment, has residual cancer cells in his body.
  • drug resistance refers to the condition when a disease does not respond to the treatment of a drug or drugs. Drug resistance can be either intrinsic, which means the disease has never been responsive to the drug or drugs, or it can be acquired, which means the disease ceases responding to a drug or drugs that the disease had previously responded to. In certain embodiments, drug resistance is intrinsic. In certain embodiments, the drug resistance is acquired.
  • EC50 refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.
  • Emax refers to an amount, concentration or dosage of a particular test compound that achieves a 100% inhibition of a maximal response in an assay that measures such response.
  • the term “Eo” refers to an amount, concentration or dosage of a particular test compound that achieves a 0% inhibition of a maximal response in an assay that measures such response.
  • the terms “subject” and “patient” are used interchangeably herein.
  • the terms “subject” and “subjects” refer to an animal, such as a mammal including a non- primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey such as a cynomolgous monkey, a chimpanzee and a human), and for example, a human.
  • the subject is a farm animal (e.g., a horse, a cow, a pig, etc.) or a pet (e.g., a dog or a cat).
  • the subject is a human.
  • the terms “drug,” “therapeutic agent,” and “chemotherapeutic agent” refer to any agent(s), compound, or pharmaceutical composition thereof, which can be used in the treatment or prevention of a disorder or one or more symptoms thereof.
  • the term “therapeutic agent” includes a compound provided herein.
  • a therapeutic agent is an agent which is known to be useful for, or has been or is currently being used for the treatment or prevention of a disorder or one or more symptoms thereof.
  • “Therapeutically effective amount” refers to an amount of a compound or composition that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease.
  • a “therapeutically effective amount” can vary depending on, inter alia, the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
  • Treating” or “treatment” of any disease or disorder refers, in one embodiment, to ameliorating a disease or disorder that exists in a subject.
  • “treating” or “treatment” includes ameliorating at least one physical parameter, which may be indiscernible by the subject.
  • “treating” or “treatment” includes modulating the disease or disorder, either physically (e.g., stabilization of a discernible symptom) or physiologically (e.g., stabilization of a physical parameter) or both.
  • “treating” or “treatment” includes delaying the onset of the disease or disorder.
  • prophylactic agent and “prophylactic agents” as used refer to any agent(s) which can be used in the prevention of a disorder or one or more symptoms thereof.
  • the term “prophylactic agent” includes a compound provided herein.
  • the term “prophylactic agent” does not refer to a compound provided herein.
  • a prophylactic agent is an agent which is known to be useful for, or has been or is currently being used to prevent or impede the onset, development, progression and/or severity of a disorder.
  • prophylactically effective amount refers to the amount of a therapy (e.g., prophylactic agent) which is sufficient to result in the prevention or reduction of the development, recurrence or onset of one or more symptoms associated with a disorder or to enhance or improve the prophylactic effect(s) of another therapy (e.g., another prophylactic agent).
  • a therapy e.g., prophylactic agent
  • another therapy e.g., another prophylactic agent
  • Prodrug compounds of a variety of therapeutic agents can be formed using methods available in the art and those disclosed herein.
  • the therapeutic agent can be derivatized to include a reactive group for attachment of the phosphate moiety.
  • Such therapeutic agent includes but is not limited to nucleosides and nucleoside analogues including acyclic nucleosides.
  • a "phosphoramidate or cyclic phosphate compound of a therapeutic agent” includes a therapeutic agent derivatized to include a phosphoramidate or cyclic phosphate group.
  • the therapeutic agent is, for example, an anti-cancer agent that includes, or has been derivatized to include, a reactive group, such as a hydroxyl, for attachment of the phosphoroamidate and cyclic phosphate moiety.
  • Such therapeutic agents include, but are not limited to nucleosides and nucleoside analogs including acyclic nucleosides.
  • a nucleoside analog refers to a structurally modified nucleoside.
  • the nucleosides or nucleoside analogs are derivitized at the 5'- and/or the 3 '-position by removal of a hydrogen from an hydroxyl group to include a phosphate moiety comprising a phosphoramidate and a cyclic phosphate group.
  • the nucleosides or nucleoside analogs are derivitized at the 5 '- and/or the 2'-position by removal of a hydrogen from an hydroxyl group to include a phosphate moiety comprising a phosphoramidate and a cyclic phosphate group.
  • Modified phosphate derivatives comprising phosphoramidate and cyclic phosphate derivatives of nucleoside analogues comprising natural and non-natural nucleosides described herein can be formed as described herein and used for the treatment of cancer.
  • the derivative moiety can be at the 3 ' position.
  • the derivative moiety can be at the 5' position.
  • the derivative moiety can be at the 3', 5' positions linked together to form a cyclic derivative.
  • the prodrug moiety can be at the 2', 5' positions linked together to form a cyclic derivative.
  • each of X and Y is independently hydrogen, -OR a , -NR a R , an N-linked or O- linked amino acid residue, or an N-linked or O-linked residue of an amino acid derivative;
  • R a and R at each occurrence is independently hydrogen, alkyl, aryl, arylalkyl, or heteroarylalkyl;
  • R is a nucleobase
  • R 1 is hydrogen, halo, cyano, alkynyl or— OR 6 ;
  • R 6 is hydrogen, alkyl, aryl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aroxycarbonyl, alkylaminocarbonyl, arylaminocarbonyl, or alkylarylaminocarbonyl.
  • R 1 is hydrogen, halo or—OR 6 .
  • R z is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid; W is NH 2 , halo, OMe, or OH; T is NH 2 , F, CI or hydrogen;
  • R 1 is hydrogen, halo, OH, protected OH, cyano or alkynyl;
  • Q is OR 3 ;
  • E is CR 4 R 5 ;
  • L is H, p-Me, p-OMe, p-Cl, or 3,4-Cl; n is 1 ; m is 1 or 2; p is absent or O; R y is alkyl, alkenyl, alkynyl, alkoxycarbonyl, or hydroxyalkyl, each independently optionally substituted;
  • R 2 is hydrogen or alkyl
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl, alkylcarbonyl or aralkyl
  • R 4 and R 5 are selected from: i) R 4 and R 5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl, or alkylheteroaryl, wherein the alkyl is optionally substituted by alkoxy; or ii) R 4 and R 5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring; R 6 with one of X and Y, together with the atoms to which they are attached, combine to form a six-membered heterocyclic ring where R 6 and the one of X and Y together represent a single divalent -0-; or
  • R 6 with one of X and Y, together with the atoms to which they are attached, combine to form a 7-12-membered heterocyclic ring;
  • R y is other than alkyl.
  • R z is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
  • W is NH 2 , CI, OMe or OH
  • T is NH 2 , F, CI or hydrogen
  • R 1 is hydrogen, OH, F, cyano or alkynyl
  • E is independently CR 4 R 5 ;
  • L is H, p-Me, p-OMe, p-Cl, or 3,4-Cl; n is 1 ; m is 1 or 2; p is absent or O
  • R y is alkyl, alkenyl, alkynyl, alkoxycarbonyl, or hydroxyalkyl, each independently optionally substituted; hydrogen or alkyl;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl or aralkyl
  • R 4 and R 5 are selected from: i) R 4 and R 5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl or alkylheteroaryl, wherein alkyl is optionally substituted by alkoxy; or
  • R 4 and R 5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring;
  • R y is other than alkyl.
  • R is not
  • T is not hydrogen; W is not OH;
  • R 1 is not cyano or alkynyl
  • R 3 is not aralkyl.
  • R y is alkyl, alkenyl, or alkynyl. In one embodiment, R y is alkyl.
  • R y is alkenyl. In one embodiment, R y is alkynyl. In one embodiment, R y is hydroxyalkyl. In one embodiment, R y is 2-hydroxylmethylprop-2- yi.
  • Q is OR , wherein R is as defined herein.
  • Q is OCH 3 , OCH 2 CH 3 , OCH(CH 3 ) 2 or OCH 2 phenyl.
  • Q is OCH 3 , OCH 2 CH 3 , or OCH(C3 ⁇ 4) 2 .
  • R z is hydrogen
  • R 3 is C3 ⁇ 4, CH 2 CH 3 , CH(C3 ⁇ 4) 2 or CH 2 phenyl.
  • E is CR 4 R 5 , wherein R 4 and R 5 are each as defined herein.
  • R 4 or R 5 is hydrogen.
  • R 4 or R 5 is alkyl or cycloalkyl.
  • R 4 or R 5 is lower alkyl.
  • R 4 or R 5 is methyl.
  • R 4 or R 5 is isobutyl.
  • R 4 or R 5 is C3-C5 cycloalkyl.
  • R 4 or R 5 is cyclopentyl.
  • one of R 4 and R 5 is hydrogen and the other is alkyl or cycloalkyl.
  • one of R 4 and R 5 is hydrogen and the other is lower alkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is methyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is isobutyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is cyclopentyl.
  • Z is an oleic acid chain.
  • W is NH 2 and T is CI or F. In one embodiment, W is CI or OMe and T is NH 2 . In one embodiment, W is OMe and T is NH 2 . In one embodiment, W is CI and T is NH 2 .
  • R 1 is hydroxyl or fluoro. In one embodiment, R 1 is hydroxyl. In one embodiment, R 1 is fluoro.
  • R 2 is hydrogen or alkyl. In one embodiment, R 2 is lower alkyl. In one embodiment, R 2 is methyl. In one embodiment, R 2 is hydrogen.
  • R 6 is hydrogen
  • R is
  • the compound of formula (I) is selected such that when R is ? , then R y is other than alkyl.
  • the compound of formula (I) is selected such that when X and Y are both RV S ⁇ s ⁇ t
  • the compound of formula (I) is selected such that when R is
  • Y when X is OH, Y is embodiment, when X is OH, Y is o 1 In one embodiment, when X is OH, Y is
  • Y when X is OH, Y is p-N0 2 PhCH 2 0— . In one embodiment, when X is OH, Y is— NHCH 2 Ph.
  • Y when X is OH, Y is— N(CH 3 )CH 2 Ph. In one embodiment, when X is OH, Y is— NHR 2 .
  • Y when X is OH, Y is-NH(CH 2 )5N(CH 3 )2. In one embodiment, when X is OH, Y is— BH 3 .
  • Y when X is O , Y is °2 N one embodiment, when X is , Y is -N0 2 PhCH 2 0— .
  • Y is— NHCH 2 Ph.
  • Y is p-N0 2 PhCH 2 0-.
  • Y is— NHR 2 .
  • Y is -NH(CH 2 ) 5 N(CH 3 ) 2
  • Y is— BH 3 .
  • Y is m .
  • Y is— NHCH 2 Ph.
  • Y is— N(CH 3 )CH 2 Ph.
  • Y when X is o ' , Y is f ⁇ -s In one embodiment, when X is o 1 , Y is °2 N In one embodiment, when X is 0 , Y is
  • Y is— NHCH 2 Ph.
  • Y is -NH(CH 2 ) 5 N(CH 3 )2.
  • Y is H 2 N 0H .
  • Y is p-N0 2 PhCH 2 0— .
  • Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3
  • Y is— NHCH 2 Ph.
  • Y is -N(CH 3 )CH 2 Ph.
  • Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is °2 , Y is -NH(CH 2 ) 5 N(CH 3
  • Y is H 2 N 0 H .
  • Y when X is ⁇ / ⁇ , Y is p-N0 2 PhCH 2 0- embodiment, when X is , Y is— NHCH 2 Ph.
  • Y is -N(CH 3 )CH 2 Ph.
  • Y is— NHR .
  • Y is -NH(CH 2 ) 5 N(CH 3 ) 2 .
  • Y is p-N0 2 PhCH 2 0— . In one embodiment, when X is p-N0 2 PhCH 2 0— Y is— NHCH 2 Ph.
  • Y is— N(CH 3 )CH 2 Ph. In one embodiment, when X is p- N0 2 PhCH 2 0- Y is— NHR 2 .
  • Y when X is p-N0 2 PhCH 2 0— , Y is— NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is p-N0 2 PhCH 2 0— , Y is— BH 3 .
  • Y is OR 3 .
  • Y is H 2 N ° 0H .
  • Y when X is p-N0 2 PhCH 2 0— Y is . In one embodiment, when X is— NHCH 2 Ph, Y is— NHCH 2 Ph. In one embodiment, when X is— NHCH 2 Ph, Y is -N(CH 3 )CH 2 Ph. In one embodiment, when X is— NHCH 2 Ph, Y is— NHR 2 . In one embodiment, when X is— NHCH 2 Ph, Y is— NH(CH 2 )5N(CH 3 ) 2 . In one embodiment, when X is— NHCH 2 Ph, Y is— BH 3 .
  • Y is H 2 N OH .
  • Y is .
  • Y is H 2 N 0 H .
  • Y is selected from
  • Y is and R y ig alky l
  • R y is hydroxyalkyl.
  • R y is 2-hydroxylmethylprop-2-yl.
  • X is— NHCH 2 Ph
  • Y is Q / ⁇ ° '
  • Q is OR 3 .
  • R 3 is alkyl.
  • Q is OCH 3 , OCH 2 CH 3 , or OCH(CH 3 ) 2 .
  • Y is -N(CH 3 )CH 2 Ph.
  • Y is— NHR 2 .
  • Y when X is -N(CH 3 )CH 2 Ph, Y is -NH(CH 2 ) 5 N(CH 3 ) 2 .
  • Y when X is -N(CH 3 )CH 2 Ph, Y is— BH 3 .
  • Y when X is— N(CH 3 )CH 2 Ph, Y is OR 3 . In one embodiment, when X is -N(CH 3 )CH 2 Ph, Y is .
  • Y is H 2 N 0 H .
  • Y when X is -N(CH 3 )CH 2 Ph, Y is H 2 N 0 H . In one embodiment, when X is— NHR 2 , Y is— NHR 2 .
  • Y when X is—NHR 2 , Y is -NH(CH 2 ) 5 N(CH 3 ) 2 . In one embodiment, when X is—NHR 2 , Y is— BH .
  • Y is OR 3 .
  • Y is OR 3 .
  • Y is OR 3 .
  • Y is H 2 N 0H .
  • Y is H 2 N 0H .
  • Y when X is— NH(CH 2 )5N(CH 3 )2, Y is— NH(CH 2 )5N(CH 3 )2. In one embodiment, when X is— NH(CH 2 )5N(CH 3 )2, Y is— BH 3 . embodiment, when X is -NH(CH 2 )5N(CH 3 )2, Y is
  • Y is OR 3 .
  • Y is OR 3 .
  • Y is . In one embodiment, when X is -NH(CH 2 )5N(CH 3 )2,
  • X when X is -NH(CH 2 )5N(CH 3 )2, In one embodiment, when X is— BH 3 , Y is— BH 3 .
  • Y is H 2 N 0 H .
  • Y is H 2 N ° 0 H . In one embodiment, when X is H 2 N ° 0H Y is H 2 N ° 0 H . In one embodiment, when X is H 2 N ° 0H Y is H 2 N ° 0 H . In one embodiment, when X is
  • Q is OCH 3 , OCH 2 CH 3 , or OCH(CH 3 ) 2 .
  • R when X is , R is hydrogen, R or R is hydrogen.
  • R 4 or R 5 is alkyl or cycloalkyl. In one embodiment, R 4 or R 5 is isobutyl. In one embodiment, R 4 or R 5 is C3-C5 cycloalkyl. In one embodiment, R 4 or R 5 is cyclopentyl.
  • Q is OR 3 , wherein R 3 and all other variables are as described elsewhere herein.
  • R when X is , Y is , R is hydrogen, R or R is hydrogen. In one embodiment, R 4 or R 5 is alkyl. In one embodiment, R 4 or R 5 is isobutyl. In one embodiment, Q is OR . In one embodiment, R is alkyl. In one embodiment Q, is OCH 3 , OCH 2 CH 3 , or OCH(CH 3 ) 2 .
  • X when X is , Y is , m is 1 , and Q is OR , wherein n, R 2 , R 3 , R 4 , R 5 , R 6 and all other variables are as described elsewhere herein.
  • Q is OCH 3 , OCH 2 CH 3 , or OCH(C3 ⁇ 4) 2 .
  • R is hydrogen, R or R is hydrogen.
  • R 4 or R 5 is alkyl or cycloalkyl. In one embodiment, R 4 or R 5 is isobutyl. In one embodiment, R 4 or R 5 is C3-C5 cycloalkyl. In one embodiment, R 4 or R 5 is cyclopentyl.
  • Q is OR 3 , wherein R 3 and all other variables are as described elsewhere herein.
  • R 2 is hydrogen
  • R r. 4 4 o composerrdon R r.5 is hydrogen.
  • R 4 or R 5 is alkyl.
  • R 4 or R 5 is isobutyl.
  • Q is OR 3 .
  • R 3 is alkyl.
  • Q is OCH 3 , OCH 2 CH 3 , or OCH(C3 ⁇ 4) 2 .
  • R y is 2-hydroxylmethylprop-2-yl, and other variables are as described elsewhere herein.
  • Q is OC3 ⁇ 4, OCH 2 CH 3 , or OCH(CH 3 ) 2 .
  • R when X is , Y is , R is hydrogen, R or R is hydrogen. In one embodiment, R 4 or R 5 is alkyl or cycloalkyl. In one embodiment, R 4 or R 5 is methyl. In one embodiment, R 4 or R 5 is C3-C 5 cycloalkyl. In one embodiment, R 4 or R 5 is cyclopentyl. In one embodiment, Q is OR 3 , wherein R 3 and all other variables are as described elsewhere herein. In one embodiment, when X is , Y is O 1 , R 2 is hydrogen, R 4 or R 5 is hydrogen. In one embodiment, R or R is alkyl. In one embodiment, R or R is methyl.
  • R 3 is alkyl.
  • Q is OCH 3 , OCH 2 CH 3 , or OCH(CH 3 ) 2 .
  • L is hydrogen
  • L is hydrogen
  • the amino acid when the phosphoramidate is substituted with at least one amino acid group, the amino acid is in the L-configuration. In one embodiment, the amino acid is alanine. In one embodiment, the amino acid is leucine.
  • the phosphoramidate compound provided herein possesses a chiral phosphorous center. In some embodiments, the phosphoramidate is diastereomerically enriched.
  • provided herein are:
  • compositions comprising a compound as described herein, e.g., of Formula I or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier, diluents or excipient;
  • a method for the treatment and/or prophylaxis of cancer, in one embodiment, leukemia comprising the administration of an effective amount of a compound as described herein, e.g., of Formulas I its pharmaceutically acceptable salt or composition.
  • R z is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
  • W is NH 2 , halo, or OMe
  • T is NH 2 , F, or CI
  • R 1 is hydrogen, halo, OH, or protected OH
  • E is CR 4 R 5 ;
  • n 1 ;
  • R 7 is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
  • R is hydrogen, alkyl, alkenyl, alkynyl or aralkyl; R 4 and R 5 are selected from:
  • R 4 and R 5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl or alkylheteroaryl, wherein the alkyl is optionally substituted by alkoxy; or
  • R 4 and R 5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
  • R 3 is not aralkyl.
  • R z is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
  • W is NH 2 or CI
  • T is NH 2 , F or CI
  • R 1 is hydrogen, OH or F
  • E is CR 4 R 5 ;
  • n 1 ;
  • R 7 is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl or aralkyl
  • R 4 and R 5 are selected as follows:
  • R 4 and R 5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl or alkylheteroaryl, wherein alkyl is optionally substituted by alkoxy; or
  • R 4 and R 5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
  • R 3 is not aralkyl.
  • E is CR 4 R 5 , wherein R 4 and R 5 are each as defined herein.
  • R 4 or R 5 is hydrogen.
  • R 4 or R 5 is alkyl, or cycloalkyl.
  • R 4 or R 5 is lower alkyl.
  • R 4 or R 5 is methyl.
  • R 4 or R 5 is isobutyl.
  • R 4 or R 5 is C3-C5 cycloalkyl.
  • R 4 or R 5 is cyclopentyl.
  • one of R 4 and R 5 is hydrogen and the other is alkyl or cycloalkyl.
  • one of R and R' is hydrogen and the other is lower alkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is methyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is isobutyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is cyclopentyl.
  • Z is an oleic acid chain.
  • W is NH 2 , T is CI or F. In one embodiment, W is CI, T is NH 2 . In one embodiment, W is OMe, T is NH2.
  • R 1 is hydrogen. In one embodiment, R 1 is hydroxy or halo. In one embodiment R 1 is fluoro. In one embodiment, R 2 is hydrogen or alkyl. In one embodiment, R 2 is lower alkyl. In one embodiment, R 2 is methyl. In one embodiment, R 2 is hydrogen.
  • R 3 is C3 ⁇ 4, CH 2 CH 3 , CH(CH 3 ) 2 or CH 2 phenyl.
  • Y is .
  • Y is OR , wherein R is as defined herein.
  • R is as defined herein.
  • Y is OCH 3 , OCH 2 CH 3 , or OCH(CI3 ⁇ 4) 2 .
  • Q is OCH(CH 3 ) 2 .
  • Q is OCH 2 CH 3 or OCH 2 phenyl.
  • the amino acid when the phosphoramidate is substituted with an amino acid group, the amino acid is in the L-configuration. In one embodiment, the amino acid is alanine. In one embodiment, the amino acid is leucine.
  • the phosphoramidate compound provided herein possesses a chiral phosphorous center. In some embodiments, the phosphoramidate is diastereomerically enriched.
  • Y is or OR 7 ;
  • R is R z is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
  • E is CR 4 R 5 ;
  • n 1 ;
  • R is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
  • R 3 is hydrogen, alkyl, alkenyl, alkynyl or aralkyl
  • R 4 and R 5 are selected from:
  • R 4 and R 5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl, or alkylheteroaryl, wherein alkyl is optionally substituted by alkoxy; or
  • R 4 and R 5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
  • R 3 is not aralkyl.
  • E is CR 4 R 5 , wherein R 4 and R 5 are each as defined herein.
  • R 4 or R 5 is hydrogen.
  • R 4 or R 5 is alkyl, or cycloalkyl.
  • R 4 or R 5 is lower alkyl.
  • R 4 or R 5 is methyl.
  • R 4 or R 5 is isobutyl.
  • R 4 or R 5 is C3-C5 cycloalkyl.
  • R 4 or R 5 is cyclopentyl.
  • one of R 4 and R 5 is hydrogen and the other is alkyl or cycloalkyl.
  • one of R 4 and R 5 is hydrogen and the other is lower alkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is methyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is isobutyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is cyclopentyl.
  • Z is an oleic acid chain.
  • R is hydrogen or alkyl. In one embodiment, R is lower alkyl. In one embodiment, R 2 is methyl. In one embodiment, R 2 is hydrogen.
  • R 3 is C3 ⁇ 4, CH 2 CH 3 , CH(CH 3 ) 2 or CH 2 phenyl.
  • Y is . In one embodiment, Y is OR , wherein R is as defined herein. In one embodiment, Y is OCH 3 , OCH 2 CH 3 , or OCH(CH 3 ) 2 . In one embodiment Q is OCH(CH 3 ) 2 . In one embodiment Q is OCH 2 CH 3 or OCH 2 phenyl.
  • R z is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
  • E is CR 4 R 5 ; n is 1 ;
  • R 7 is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
  • R is hydrogen, alkyl, alkenyl, alkynyl or aralkyl
  • R 4 and R 5 are selected from:
  • R 4 and R 5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
  • R 3 is not aralkyl.
  • E is CR 4 R 5 , wherein R 4 and R 5 are each as defined herein.
  • R 4 or R 5 is hydrogen.
  • R 4 or R 5 is alkyl, or cycloalkyl.
  • R 4 or R 5 is lower alkyl.
  • R 4 or R 5 is methyl.
  • R 4 or R 5 is isobutyl.
  • R 4 or R 5 is C3-C5 cycloalkyl.
  • R 4 or R 5 is cyclopentyl.
  • one of R 4 and R 5 is hydrogen and the other is alkyl or cycloalkyl.
  • one of R 4 and R 5 is hydrogen and the other is lower alkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is methyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is isobutyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R 4 and R 5 is hydrogen and the other is cyclopentyl.
  • Z is an oleic acid chain.
  • R 2 is hydrogen or alkyl. In one embodiment, R 2 is lower alkyl. In one embodiment, R 2 is methyl. In one embodiment, R 2 is hydrogen.
  • R 3 is C3 ⁇ 4, CH 2 CH 3 , CH(CH 3 ) 2 or CH 2 phenyl
  • Y is .
  • R 7 is as defined herein.
  • Y is OCH3, OCH 2 CH3, or OCH(CI3 ⁇ 4) 2 .
  • Q is OCH(CH 3 ) 2 .
  • Q is OCH 2 CH 3 or OCH 2 phenyl.
  • the amino acid when the phosphoramidate is substituted with an amino acid group, the amino acid is in the L-configuration. In one embodiment, the amino acid is alanine. In one embodiment, the amino acid is leucine.
  • the phosphoramidate compound provided herein possesses a chiral phosphorous center. In some embodiments, the phosphoramidate is diastereomerically enriched.
  • R 2 is hydrogen; R 3 is alkyl; R 4 is hydrogen and R 5 is alkyl or cycloalkyl; or R 4 is alkyl or cycloalkyl, and R 5 is hydrogen.
  • R 2 is hydrogen; R 3 is alkyl; R 4 is hydrogen and R 5 is alkyl or cycloalkyl; or R 4 is alkyl or cycloalkyl, and R 5 is hydrogen.
  • R, R 1 , and R 6 are as defined herein.
  • R 2 is hydrogen; R 3 is alkyl; R 4 is hydrogen and R 5 is alkyl, or R 4 is alkyl and R 5 is hydrogen.
  • the compound provided herein is a compound of one of the following Formulas:
  • R y , R z , and R 6 are as described in Formula I.
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the nucleosides that can be derivatized to include a phosphoramidate, e.g., at the 5' position include:
  • the compound provided herein is a compound of one of the following Formulas: In one embodiment, the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • the compound provided herein is a compound of one of the following Formulas:
  • ⁇ and 4' carbons of a nucleoside are chiral, their nonhydrogen substituents (the base and the CHOR groups, respectively) can be either cis (on the same side) or trans (on opposite sides) with respect to the sugar ring system.
  • the four optical isomers therefore are represented by the following configurations (when orienting the sugar moiety in a horizontal plane such that the oxygen atom is in the back): cis (with both groups “up”, which corresponds to the configuration of naturally occurring ⁇ -D nucleosides), cis (with both groups “down”, which is a nonnaturally occurring ⁇ -L configuration), trans (with the C2' substituent "up” and the C4' substituent "down”), and trans (with the C2' substituent "down” and the C4' substituent "up”).
  • the "D-nucleosides” are cis nucleosides in a natural configuration and the "L-nucleosides” are cis nucleosides in the non-naturally occurring configuration.
  • optically active materials examples include at least the following.
  • the desired enantiomer is then released from the diastereomer;
  • kinetic resolutions refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions;
  • enantiospecific synthesis from non-racemic precursors - a synthetic technique whereby the desired enantiomer is obtained from non- chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis;
  • the stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;
  • the barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
  • compositions of the present disclosure are provided that are substantially free of an undesignated enantiomer of a nucleoside or nucleoside derivative.
  • the compounds are substantially free of undesignated enantiomers.
  • the composition includes a compound that is at least 85 %, 90%, 95%, 98%, 99%, to 100% by weight of the designated compound, the remainder comprising other chemical species or enantiomers.
  • isotopically enriched compounds including but not limited to isotopically enriched nucleoside derivatives.
  • Isotopic enrichment of a drug can be used, for example, to (1) reduce or eliminate unwanted metabolites, (2) increase the half-life of the parent drug, (3) decrease the number of doses needed to achieve a desired effect, (4) decrease the amount of a dose necessary to achieve a desired effect, (5) increase the formation of active metabolites, if any are formed, and/or (6) decrese the production of deleterious metabolites in specific tissues and/or create a more effective drug and/or a safer drug for combination therapy, whether the combination therapy is intentional or not.
  • KIE Kinetic Isotope Effect
  • DKIE Deuterium Kinetic Isotope Effect
  • the magnitude of the DKIE can be expressed as the ratio between the rates of a given reaction in which a C-H bond is broken, and the same reaction where deuterium is substituted for hydrogen.
  • the DKIE can range from about 1 (no isotope effect) to very large numbers, such as 50 or more, meaning that the reaction can be fifty, or more, times slower when deuterium is substituted for hydrogen.
  • high DKIE values may be due in part to a phenomenon known as tunneling, which is a consequence of the uncertainty principle. Tunneling is ascribed to the small mass of a hydrogen atom, and occurs because transition states involving a proton can sometimes form in the absence of the required activation energy. Because deuterium has more mass than hydrogen, it statistically has a much lower probability of undergoing this phenomenon.
  • substitution of tritium ("T") for hydrogen results in yet a stronger bond than deuterium and gives numerically larger isotope effects.
  • substitution of isotopes for other elements including, but not limited to, 13 C or 14 C for carbon, 33 S, 34 S, or 36 S for sulfur, 15 N for nitrogen, and 17 0 or 18 0 for oxygen, may lead to a similar kinetic isotope effect.
  • the DKIE was used to decrease the hepatotoxicity of halothane by presumably limiting the production of reactive species such as trifluoroacetyl chloride.
  • this method may not be applicable to all drug classes.
  • deuterium incorporation can lead to metabolic switching.
  • the concept of metabolic switching asserts that xenogens, when sequestered by Phase I enzymes, may bind transiently and re-bind in a variety of conformations prior to the chemical reaction (e.g., oxidation). This hypothesis is supported by the relatively vast size of binding pockets in many Phase I enzymes and the promiscuous nature of many metabolic reactions. Metabolic switching can potentially lead to different proportions of known metabolites as well as altogether new metabolites. This new metabolic profile may impart more or less toxicity.
  • the animal body expresses a variety of enzymes for the purpose of eliminating foreign substances, such as therapeutic agents, from its circulation system.
  • enzymes include the cytochrome P450 enzymes ("CYPs"), esterases, proteases, reductases, dehydrogenases, and monoamine oxidases, to react with and convert these foreign substances to more polar intermediates or metabolites for renal excretion.
  • CYPs cytochrome P450 enzymes
  • esterases esterases
  • proteases proteases
  • reductases reductases
  • dehydrogenases dehydrogenases
  • monoamine oxidases monoamine oxidases
  • the resultant metabolites may be stable or unstable under physiological conditions, and can have substantially different pharmacokinetic, pharmacodynamic, and acute and long-term toxicity profiles relative to the parent compounds. For many drugs, such oxidations are rapid. These drugs therefore often require the administration of multiple or high daily doses.
  • isotopic enrichment at certain positions of a compound provided herein will produce a detectable KIE that will affect the pharmacokinetic, pharmacologic, and/or toxicological profiles of a compound provided herein in comparison with a similar compound having a natural isotopic composition.
  • the compounds provided herein can be, in some instances, prepared, isolated, or obtained by any method apparent to those of skill in the art. Those skilled in the art will recognize that compounds provided herein may be designed or prepared by reaction, e.g., at a hydroxy group of said anti-cancer drug, for example, via condensation or dehydration.
  • Step 1 To a stirred solution of compound 22a or 22b (9.97mmol) in THF (lOmL/mmol) at -10°C was added phosphorus oxychloride (11.96mmol). The reaction mixture was stirred from -10°C to room temperature overnight and then, a solution of reagent Bl (9.97mmol) and triethylamine (49.85mmol) in acetonitrile (2.4mL/mmol) was added at 0°C. The reaction mixture was stirred for 1 hour at 0°C.
  • Step 2 To a stirred suspension of compound 23a-c (2.40mmol) in DCM (50mL/mmol) at -80°C under nitrogen was added a solution of boron trichloride, 1.0M in DCM (12.02mmol). The reaction mixture was stirred from -80°C to -40°C for 4 hours. Then MeOH (2mL) was added at -80°C and the reaction mixture was concentrated under reduced pressure at 25°C. The crude residue was purified by RP18 chromatography (H 2 O/CH 3 CN) to afford the expected compound as solid. In some cases, the mixture of diastereoisomers was purified by chiral HPLC to afford the expected diastereoisomer.
  • Step 1 To a stirred solution of compound 22a (1.86mmol) in THF (lOmL/mmol) at -10°C under nitrogen was added phosphorus oxychloride (2.23mmol). The reaction mixture was stirred from -10°C to room temperature overnight and then, cooled down to 0°C. Then, a solution of reagent B3 (3.72mmol) and triethylamine (9.31mmol) in acetonitrile (2.4mL/mmol) was added at 0°C. The reaction mixture was stirred at room temperature overnight. The reaction mixture was then diluted with ethyl acetate and washed with a saturated NH 4 CI solution. The organic layer was filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (DCM/methanol: 0 to 3%) to afford the expected compound. Step 2: according to step 2 of general procedure A.
  • Step 1 To a solution of compound lb (2.20mmol) in THF (lOmL/mmol) at 0°C was added a solution of tert-butylmagnesium chloride, 1M in THF (4.60mmol). The reaction mixture was stirred for 1 hour at room temperature, then cooled to 0°C. To this reaction mixture was added a solution of the appropriate reagent CI (2.40mmol) in THF (lOmL/mmol). The reaction mixture was stirred for 1 hour at 0°C and at room temperature overnight. The reaction mixture was diluted with ethyl acetate and washed with a saturated NH 4 C1 solution, water and brine. The organic layer was dried, filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (DCM/methanol) to afford the expected intermediate as a mixture of diastereoisomers;
  • Step 2 To a solution of the previous intermediate (0.52mmol) in DCM (30mL/mmol) was added trifluoroacetic acid (10.27mmol). The reaction mixture was stirred at room temperature for 3 hours and purified by flash chromatography on silica gel (DCM/methanol: 0 to 20%) followed by RP18 chromatography ((H 2 0/CH 3 CN) to afford the expected compound as mixture of diastereoisomers. This mixture was purified by MS- preparative HPLC or by chiral HPLC to afford the 2 expected diastereoisomers as pure solid compounds.
  • General procedure D To a solution of the previous intermediate (0.52mmol) in DCM (30mL/mmol) was added trifluoroacetic acid (10.27mmol). The reaction mixture was stirred at room temperature for 3 hours and purified by flash chromatography on silica gel (DCM/methanol: 0 to 20%) followed by RP18 chromatography ((H 2 0/CH 3 CN) to afford the
  • Step 1 To a stirred solution of 4-nitrophenol (29.36mmol) and phosphorus oxychloride (14.68mmol) in diethyl ether (lOmL/mmol) at -80°C under nitrogen was added triethylamine (29.36mmol). The reaction mixture was stirred at room temperature overnight. Then, DCM (lOmL/mmol) and the appropriate L-alanine ester hydrochloride (14.68mmol) were added at 0°C under nitrogen. To the reaction mixture was added dropwise at 0°C triethylamine (29.36mmol). The reaction mixture was stirred at room temperature overnight, and then, filtrated. The filtrate was concentrated under reduced pressure.
  • reaction mixture was extracted with DCM and the organic layer was dried, filtered and concentrated under reduced pressure.
  • the crude residue was purified by flash chromatography on silica gel (DCM/methanol: 0 to 10%) to afford the expected compound as a mixture of diastereoisomers. This mixture was purified by MS -Preparative HPLC to afford the pure diastereoisomer.
  • Compound 24a was synthesized from compound 22a with reagent Bl and B2 according to scheme 7 and general procedure A.
  • Compound 24d was synthesized from compound 22b with reagents Bl and B5 according to scheme 7 and general procedure A.
  • Compound 24f was synthesized from compound 22c with reagents B6 and B5 according to scheme 9 and general procedure A step 1. In this case, the addition of phosphorus oxychloride was followed by the addition of Proton sponge. Step 2 was done according to step 2 of general procedure C.
  • Compound 28 was synthesized from 2'OCBz-fludarabine according to scheme 3bis and to the procedure D. In this case, additional step of deprotection using 3 ⁇ 4, Pd/C 5% in ethanol was used to afford the 2 expected diastereoisomers.
  • Compound 29 was synthesized according to scheme 10 and to the following procedure.
  • Therapeutic agents that can be derivatized to phosphate derivatives include any anti-cancer agent that includes, or has been derivatized to include a reactive group for attachment of the phosphate moiety, including but not limited to nucleosides and nucleoside analogues including acyclic nucleosides. Any of the compounds disclosed herein can be provided in the appropriate pharmaceutical composition and be administered by a suitable route of administration.
  • compositions containing at least one compound as described herein including a compound of general Formulas I, Ila, lib, or lie, if appropriate in the salt form, either used alone or in the form of a combination with one or more compatible and pharmaceutically acceptable carriers, such as excipients, diluents or adjuvants, or with another anti-cancer agent.
  • the second agent can be formulated or packaged with the compound provided herein.
  • the second agent will only be formulated with the compound provided herein when, according to the judgment of those of skill in the art, such co-formulation should not interfere with the activity of either agent or the method of administration.
  • the compound provided herein and the second agent are formulated separately. They can be packaged together, or packaged separately, for the convenience of the practitioner of skill in the art.
  • the active agents provided herein may be administered by any conventional route, in particular orally, parenterally, rectally or by inhalation (e.g. in the form of aerosols).
  • the compound provided herein is administered orally.
  • compositions for oral administration of tablets, pills, hard gelatin capsules, powders or granules.
  • the active product is mixed with one or more inert diluents or adjuvants, such as sucrose, lactose, or starch.
  • compositions can comprise substances other than diluents, for example a lubricant, such as magnesium stearate, or a coating intended for controlled release.
  • a lubricant such as magnesium stearate
  • compositions for oral administration of solutions which are pharmaceutically acceptable, suspensions, emulsions, syrups, and elixirs containing inert diluents, such as water or liquid paraffin.
  • solutions which are pharmaceutically acceptable, suspensions, emulsions, syrups, and elixirs containing inert diluents, such as water or liquid paraffin.
  • inert diluents such as water or liquid paraffin.
  • These compositions can also comprise substances other than diluents, for example wetting, sweetening or flavoring products.
  • compositions for parenteral administration can be emulsions or sterile solutions. Use may be made, as solvent or vehicle, of propylene glycol, a polyethylene glycol, vegetable oils, in particular olive oil, or injectable organic esters, for example ethyl oleate. These compositions can also contain adjuvants, in particular wetting, isotonizing, emulsifying, dispersing, and stabilizing agents. Sterilization can be carried out in several ways, for example using a bacteriological filter, by radiation or by heating. They can also be prepared in the form of sterile solid compositions which can be dissolved at the time of use in sterile water or any other injectable sterile medium.
  • compositions for rectal administration are suppositories or rectal capsules which contain, in addition to the active principle, excipients such as cocoa butter, semi-synthetic glycerides or polyethylene glycols.
  • the compositions can also be aerosols.
  • the compositions can be stable sterile solutions or solid compositions dissolved at the time of use in apyrogenic sterile water, in saline or any other pharmaceutically acceptable vehicle.
  • the active principle is finely divided and combined with a water-soluble solid diluent or vehicle, for example dextran, mannitol, or lactose.
  • compositions provided herein is a pharmaceutical composition or a single unit dosage form.
  • Pharmaceutical compositions and single unit dosage forms provided herein comprise a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic agents (e.g., a compound provided herein, or other prophylactic or therapeutic agent), and a typically one or more pharmaceutically acceptable carriers or excipients.
  • prophylactic or therapeutic agents e.g., a compound provided herein, or other prophylactic or therapeutic agent
  • typically one or more pharmaceutically acceptable carriers or excipients e.g., the term "pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier includes a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered.
  • adjuvant e.g., Freund's adjuvant (complete and incomplete)
  • excipient or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water can be used as a carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Examples of suitable pharmaceutical carriers are described in "Remington 's Pharmaceutical Sciences” by E.W. Martin.
  • Typical pharmaceutical compositions and dosage forms comprise one or more excipients.
  • Suitable excipients are well-known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol, and the like.
  • composition or dosage form Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a subject and the specific active ingredients in the dosage form.
  • the composition or single unit dosage form if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • Lactose free compositions can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmocopia (USP) SP (XXI) NF (XVI).
  • USP U.S. Pharmocopia
  • XXI U.S. Pharmocopia
  • NF NF
  • lactose free compositions comprise an active ingredient, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts.
  • Exemplary lactose free dosage forms comprise an active ingredient, microcrystalline cellulose, pre gelatinized starch, and magnesium stearate.
  • anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water is widely accepted 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. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80.
  • water and heat accelerate the decomposition of some compounds.
  • the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • Anhydrous pharmaceutical compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine can be anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions can 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, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • compositions and single unit dosage forms can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations, and the like.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc.
  • Such compositions and dosage forms will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent, in certain embodiments, in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject.
  • the formulation should suit the mode of administration.
  • the pharmaceutical compositions or single unit dosage forms are sterile and in suitable form for administration to a subject, for example, an animal subject, such as a mammalian subject, for example, a human subject.
  • a pharmaceutical composition is formulated to be compatible with its intended route of administration.
  • routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, intramuscular, subcutaneous, oral, buccal, sublingual, inhalation, intranasal, transdermal, topical, transmucosal, intra-tumoral, intra- synovial, and rectal administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal, or topical administration to human beings.
  • a pharmaceutical composition is formulated in accordance with routine procedures for subcutaneous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a subject, including suspensions (e.g., aqueous or non aqueous liquid suspensions, oil in water emulsions, or a water in oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a subject; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a subject.
  • suspensions e.g., aqueous or non aque
  • compositions, shape, and type of dosage forms provided herein will typically vary depending on their use.
  • a dosage form used in the initial treatment of viral infection may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the maintenance treatment of the same infection.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder.
  • compositions are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • Typical dosage forms comprise a compound provided herein, or a pharmaceutically acceptable salt, solvate or hydrate thereof lie within the range of from about 0.1 mg to about 1000 mg per day, given as a single once-a-day dose in the morning or as divided doses throughout the day taken with food.
  • Particular dosage forms can have about 0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 2.0, 2.5, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 100, 200, 250, 500, or 1000 mg of the active compound.
  • compositions that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets ⁇ e.g., chewable tablets), cap lets, capsules, and liquids ⁇ e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington 's Pharmaceutical Sciences, 20th ed., Mack Publishing, Easton PA (2000).
  • the oral dosage forms are solid and prepared under anhydrous conditions with anhydrous ingredients, as described in detail in the sections above.
  • anhydrous ingredients as described in detail in the sections above.
  • the scope of the compositions provided herein extends beyond anhydrous, solid oral dosage forms. As such, further forms are described herein.
  • Typical oral dosage forms are prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for administration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms ⁇ e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • 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, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
  • fillers suitable 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.
  • the binder or filler in pharmaceutical compositions is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL PH 101, AVICEL PH 103 AVICEL RC 581 , AVICEL PH 105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof.
  • a specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC 581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL PH 103TM and Starch 1500 LM.
  • Disintegrants are used in the compositions to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms.
  • the amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms 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, pre gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms 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, ethyl laureate, agar, and mixtures thereof.
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R.
  • lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
  • Active ingredients such as the compounds provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art.
  • Such dosage forms can be used to provide slow or controlled release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients provided herein.
  • single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled release.
  • controlled release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non controlled counterparts.
  • the use of an optimally designed controlled release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled release formulations include extended activity of the drug, reduced dosage frequency, and increased subject compliance.
  • controlled release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • the drug may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration.
  • a pump may be used (see, Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al, Surgery 55:507 (1980); Saudek et al, N. Engl. J. Med. 321:574 (1989)).
  • polymeric materials can be used.
  • a controlled release system can be placed in a subject at an appropriate site determined by a practitioner of skill, i.e., thus requiring only a fraction of the systemic dose ⁇ see, e.g., Goodson, Medical Applications of Controlled Release, vol. 2, pp. 115-138 (1984)). Other controlled release systems are discussed in the review by Langer ⁇ Science 2 9: 1527-1533 (1990)).
  • the active ingredient can be dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross- linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, ne
  • parenteral dosage forms can be administered to subjects by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses subjects' natural defenses against contaminants, parenteral dosage forms are typically sterile or capable of being sterilized prior to administration to a subject. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. Suitable vehicles that can be used to provide parenteral dosage forms are well known to those skilled in the art.
  • examples of a suitable vehicle include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
  • aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection
  • water miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glyco
  • Transdermal, topical, and mucosal dosage forms include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington 's Pharmaceutical Sciences, 16 th , 18th and 20 th eds., Mack Publishing, Easton PA (1980, 1990 & 2000); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985).
  • transdermal dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels.
  • transdermal dosage forms include "reservoir type” or “matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients ⁇ e.g., carriers and diluents
  • other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed herein are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane -1,3- diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are non toxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington 's Pharmaceutical Sciences, 16 , 18th and 20 eds., Mack Publishing, Easton PA (1980, 1990 & 2000).
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery enhancing or penetration enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • a method for treating a proliferative disease in a subject which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or a hydrate thereof.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate,
  • the therapeutically effective amount is ranging from about 0.1 to about 100 mg/kg/day, from about 0.1 to about 50 mg/kg/day, from about 0.1 to about 40 mg/kg/day, from about 0.1 to about 30 mg/kg/day, from about 0.1 to about 25 mg/kg/day, from about 0.1 to about 20 mg/kg/day, from about 0.1 to about 15 mg/kg/day, from about 0.1 to about 10 mg/kg/day, or from about 0.1 to about 5 mg/kg/day.
  • the therapeutically effective amount is ranging from about 0.1 to about 100 mg/kg/day.
  • the therapeutically effective amount is ranging from about 0.1 to about 50 mg/kg/day.
  • the therapeutically effective amount is ranging from about 0.1 to about 40 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 30 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 25 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 20 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 15 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 10 mg/kg/day. In still another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 5 mg/kg/day.
  • the administered dose can also be expressed in units other than mg/kg/day.
  • doses for parenteral administration can be expressed as mg/m 2 /day.
  • doses for parenteral administration can be expressed as mg/m 2 /day.
  • One of ordinary skill in the art would readily know how to convert doses from mg/kg/day to mg/m 2 /day to given either the height or weight of a subject or both (see, www.fda.gov/cder/cancer/animalframe.htm).
  • a dose of 1 mg/m 2 /day for a 65 kg human is approximately equal to 38 mg/kg/day.
  • the subject is a mammal. In certain embodiments, the subject is a human.
  • In one embodiment is a compound or a pharmaceutically acceptable salt, or solvate thereof, or stereoisomeric, tautomeric, or polymorphic form thereof for use in therapy.
  • the proliferative disease is a carcinoma, including, but not limited to, Kit-mediated carcinomas, adenocarcinoma, squamous cell carcinoma, adenosquamous carcinoma, teratocarcinoma, head and neck cancer, brain cancer, intracranial carcinoma, glioblastoma (including PDGFR-mediated glioblastoma), glioblastoma multiforme (including PDGFR-mediated glioblastoma multiforme), neuroblastoma, cancer of the larynx, multiple endocrine neoplasias 2A and 2B (MENS 2A and MENS 2B) (including RET-mediated MENS), thyroid cancer (including sporadic and familial medullary thyroid carcinoma), papillary thyroid carcinoma, parathyroid carcinoma (including any RET-mediated thyroid carcinoma), follicular thyroid cancer, anaplastic thyroid cancer, bronchial carcinoid, oat cell carcinoma, lung cancer, small-cell lung cancer (
  • the proliferative disease is sarcomas, including, but not limited to, PDGFR-mediated sarcomas, osteosarcoma, osteogenic sarcoma, bone cancer, glioma (including PDGFR-mediated and/or CSF-lR-mediated glioma), astrocytoma, vascular tumors (including VEGFR-mediated vascular tumors), Kaposi's sarcoma, carcinosarcoma, hemangiosar comas (including VEGFR3 -mediated hemangiosarcomas), and lymphangiosarcoma (including VEGFR3 -mediated lymphangiosarcoma).
  • PDGFR-mediated sarcomas including PDGFR-mediated and/or CSF-lR-mediated glioma
  • astrocytoma including PDGFR-mediated and/or CSF-lR-mediated glioma
  • vascular tumors including VEGFR-mediated vascular tumors
  • the proliferative disease is a hematologic malignancy. In certain embodiments, the proliferative disease is a relapsed hematologic malignancy. In certain embodiments, the proliferative disease is a refractory hematologic malignancy. In certain embodiments, the proliferative disease is a drug-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a multidrug-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a Bcr-Abl kinase inhibitor-resistant hematologic malignancy. In certain embodiments, the proliferative disease is an imatinib-resistant hematologic malignancy.
  • the proliferative disease is a dasatinib- resistant hematologic malignancy. In certain embodiments, the proliferative disease is a nilatinib-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a bosutinib-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a cytarabine-resistant hematologic malignancy.
  • the hematologic malignancy is myeloma, leukemia, myeloproliferative diseases, acute myeloid leukemia (AML) (including FLT3 mediated and/or KIT-mediated and/or CSFlR-mediated acute myeloid leukemia), chronic myeloid leukemias (CML) (including FLT3- mediated and/or PDGFR-mediated chronic myeloid leukemia), myelodysplastic leukemias (including FLT3 -mediated myelodysplastic leukemia), myelodysplastic syndrome (including FLT3 mediated and/or Kit-mediated myelodysplastic syndrome), idiopathic hypereosinophilic syndrome (HES) (including PDGFR-mediated HES), chronic eosinophilic leukemia (CEL) (including PDGFR-mediated CEL), chronic myelomonocytic leukemia (CMML), mast cell leukemia (including Kit-mediated mast cell leukemia), or system
  • AML
  • the hematologic malignancy is lymphoma, lymphoproliferative diseases, acute lymphoblastic leukemia (ALL), B-cell acute lymphoblastic leukemias, T-cell acute lymphoblastic leukemias, chronic lymphocytic leukemia (CLL), natural killer (NK) cell leukemia, B-cell lymphoma, T-cell lymphoma, or natural killer (NK) cell lymphoma.
  • ALL acute lymphoblastic leukemia
  • B-cell acute lymphoblastic leukemias B-cell acute lymphoblastic leukemias
  • T-cell acute lymphoblastic leukemias T-cell acute lymphoblastic leukemias
  • CLL chronic lymphocytic leukemia
  • NK natural killer
  • the hematologic malignancy is myelodysplastic syndrome (MDS).
  • MDS myelodysplastic syndrome
  • the hematologic malignancy is Langerhans cell histiocytosis (including CSF-lR-mediated and/or FLT3-mediated Langerhans cell histiocytosis), mast cell tumors, or mastocytosis.
  • the hematologic malignancy is leukemia. In certain embodiments, the hematologic malignancy is relapsed leukemia. In certain embodiments, the hematologic malignancy is refractory leukemia. In certain embodiments, the hematologic malignancy is drug-resistant leukemia. In certain embodiments, the hematologic malignancy is multidrug- resistant leukemia. In certain embodiments, the hematologic malignancy is a Bcr-Abl kinase inhibitor-resistant leukemia. In certain embodiments, the hematologic malignancy is imatinib- resistant leukemia. In certain embodiments, the hematologic malignancy is dasatinib-resistant leukemia.
  • the hematologic malignancy is nilatinib-resistant leukemia. In certain embodiments, the hematologic malignancy is bosutinib-resistant leukemia. In certain embodiments, the hematologic malignancy is cytarabine-resistant leukemia. In certain embodiments, the leukemia is acute leukemia. In certain embodiments, the leukemia is relapsed acute leukemia. In certain embodiments, the leukemia is refractory acute leukemia. In certain embodiments, the leukemia is drug-resistant acute leukemia. In certain embodiments, the leukemia is multidrug-resistant acute leukemia.
  • the leukemia is a Bcr-Abl kinase inhibitor-resistant acute leukemia. In certain embodiments, the leukemia is imatinib-resistant acute leukemia. In certain embodiments, the leukemia is dasatinib-resistant acute leukemia. In certain embodiments, the leukemia is nilatinib-resistant acute leukemia. In certain embodiments, the leukemia is bosutinib-resistant acute leukemia. In certain embodiments, the leukemia is cytarabine-resistant acute leukemia. In certain embodiments, the leukemia is a hereditary leukemia. In certain embodiments, the hereditary leukemia is severe congenital neutropenia (SCN).
  • SCN severe congenital neutropenia
  • the hereditary leukemia is familial platelet disorder with acute myelogenous leukemia (FDP/AML).
  • the leukemia is caused by LEF1.
  • the leukemia is mediated by LEF1.
  • the leukemia is caused by GSK3.
  • the leukemia is ALL. In certain embodiments, the leukemia is relapsed ALL. In certain embodiments, the leukemia is refractory ALL. In certain embodiments, the leukemia is drug-resistant ALL. In certain embodiments, the leukemia is multidrug-resistant ALL. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant ALL. In certain embodiments, the leukemia is imatinib-resistant ALL. In certain embodiments, the leukemia is dasatinib-resistant ALL. In certain embodiments, the leukemia is nilatinib-resistant ALL. In certain embodiments, the leukemia is bosutinib-resistant ALL. In certain embodiments, the leukemia is cytarabine-resistant ALL.
  • ALL is leukemia that originates in the blast cells of the bone marrow (B- cells), thymus (T-cells), or lymph nodes.
  • ALL is categorized according to the French- American- British (FAB) Morphological Classification Scheme as LI - mature-appearing lymphoblasts (T- cells or pre-B-cells), L2 - immature and pleomorphic (variously shaped) lymphoblasts (T-cells or pre-B-cells), and L3 - lymphoblasts (B-cells; Burkitt's cells).
  • ALL originates in the blast cells of the bone marrow (B-cells).
  • ALL originates in the thymus (T-cells).
  • ALL originates in the lymph nodes.
  • ALL is LI type characterized by mature-appearing lymphoblasts (T-cells or pre-B-cells).
  • ALL is L2 type characterized by immature and pleomorphic (variously shaped) lymphoblasts (T-cells or pre-B-cells).
  • ALL is L3 type characterized by lymphoblasts (B-cells; Burkitt's cells).
  • the leukemia is AML. In certain embodiments, the leukemia is relapsed AML. In certain embodiments, the leukemia is refractory AML. In certain embodiments, the leukemia is drug-resistant AML. In certain embodiments, the leukemia is multidrug-resistant AML. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant AML. In certain embodiments, the leukemia is imatinib-resistant AML. In certain embodiments, the leukemia is dasatinib-resistant AML. In certain embodiments, the leukemia is nilatinib-resistant AML.
  • the leukemia is bosutinib-resistant AML. In certain embodiments, the leukemia is cytarabine-resistant AML. In certain embodiments, AML has a RAS mutation. In certain embodiments, the RAS mutation is NRAS, KRAS, or HRAS. In certain embodiments, the RAS mutation is NRAS. In certain embodiments, the RAS mutation is KRAS. In certain embodiments, the RAS mutation is HRAS.
  • AML is undifferentiated AML (MO), myeloblastic leukemia (Ml), myeloblastic leukemia (M2), promyelocytic leukemia (M3 or M3 variant [M3V]), myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]), monocytic leukemia (M5), erythro leukemia (M6), or megakaryoblastic leukemia (M7).
  • AML is undifferentiated AML (MO).
  • AML is myeloblastic leukemia (Ml).
  • AML is myeloblastic leukemia (M2).
  • AML is promyelocytic leukemia (M3 or M3 variant [M3V]). In yet another embodiment, AML is myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]). In yet another embodiment, AML is monocytic leukemia (M5). In yet another embodiment, AML is erythroleukemia (M6). In still another embodiment, AML is megakaryoblastic leukemia (M7). In certain embodiments, the leukemia is chronic leukemia. In certain embodiments, the leukemia is relapsed chronic leukemia. In certain embodiments, the leukemia is refractory chronic leukemia.
  • the leukemia is drug-resistant chronic leukemia. In certain embodiments, the leukemia is multidrug-resistant chronic leukemia. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant chronic leukemia. In certain embodiments, the leukemia is imatinib-resistant chronic leukemia. In certain embodiments, the leukemia is dasatinib-resistant chronic leukemia. In certain embodiments, the leukemia is nilatinib-resistant chronic leukemia. In certain embodiments, the leukemia is bosutinib-resistant chronic leukemia. In certain embodiments, the leukemia is cytarabine-resistant chronic leukemia.
  • the leukemia is CLL. In certain embodiments, the leukemia is relapsed CLL. In certain embodiments, the leukemia is refractory CLL. In certain embodiments, the leukemia is drug-resistant CLL. In certain embodiments, the leukemia is multidrug-resistant CLL. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant CLL. In certain embodiments, the leukemia is imatinib-resistant CLL. In certain embodiments, the leukemia is dasatinib-resistant CLL. In certain embodiments, the leukemia is nilatinib-resistant CLL. In certain embodiments, the leukemia is bosutinib-resistant CLL. In certain embodiments, the leukemia is cytarabine-resistant CLL.
  • the leukemia is CML. In certain embodiments, the leukemia is relapsed CML. In certain embodiments, the leukemia is refractory CML. In certain embodiments, the leukemia is drug-resistant CML. In certain embodiments, the leukemia is multidrug-resistant CML. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant CML. In certain embodiments, the leukemia is imatinib-resistant CML. In certain embodiments, the leukemia is dasatinib-resistant CML. In certain embodiments, the leukemia is nilatinib-resistant CML.
  • the leukemia is bosutinib-resistant CML. In certain embodiments, the leukemia is cytarabine-resistant CML. In certain embodiments, the leukemia is juvenile CML. In certain embodiments, the leukemia is juvenile CML with one or more NF-1 mutations.
  • the leukemia is T-cell leukemia.
  • the T-cell leukemia is peripheral T-cell leukemia, T-cell lymphoblastic leukemia, cutaneous T-cell leukemia, and adult T-cell leukemia.
  • the T-cell leukemia is peripheral T-cell leukemia.
  • the T-cell leukemia is T-cell lymphoblastic leukemia.
  • the T-cell leukemia is cutaneous T-cell leukemia.
  • the T-cell leukemia is adult T-cell leukemia.
  • the leukemia is Philadelphia positive.
  • the Philadelphia positive leukemia is Philadelphia positive AML, including, but not limited to, undifferentiated AML (MO), myeloblastic leukemia (Ml), myeloblastic leukemia (M2), promyelocytic leukemia (M3 or M3 variant [M3V]), myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]), monocytic leukemia (M5), erythroleukemia (M6), or megakaryoblastic leukemia (Ml).
  • the Philadelphia positive leukemia is Philadelphia positive ALL.
  • the proliferative disease is cancer, including, but not limited to, head and neck cancer (originating lip, oral cavity, oropharynx, hypopharynx, larynx, nasopharynx, nasal cavity, paranasal sinuses, or salivary glands), lung cancer (including small cell lung cancer and non-small cell lung cancer), gastrointestinal tract cancers (including esophageal cancer), gastric cancer, colorectal cancer, anal cancer, pancreatic cancer, liver cancer, gallbladder cancer, extrahepatic bile duct cancer, cancer of the ampulla of vater, breast cancer, gynecologic cancers (including cancer of uterine cervix), cancer of the uterine body, vaginal cancer, vulvar cancer, ovarian cancer, gestational trophoblastic cancer neoplasia, testicular cancer, urinary tract cancers (including renal cancer), urinary blader cancer, prostate cancer, penile cancer, urethral cancer, neurodecane,
  • cancers are basal cell carcinoma, squamous cell carcinoma, chondrosarcoma (a cancer arising in cartilage cells), mesenchymal-chondrosarcoma, soft tissue sarcomas (including malignant tumors that may arise in any of the mesodermal tissues (muscles, tendons, vessels that carry blood or lymph, joints and fat)), soft tissue sarcomas (include alveolar soft-part sarcoma), angiosarcoma, fibrosarcoma, leiomyosarcoma, liposarcoma, malignant fibrous histiocytoma, hemangiopericytoma, mesenchymoma, schwannoma, peripheral neuroectodermal tumours, rhabdomyosarcoma, synovial sarcoma, gestational trophoblastic tumor (malignancy in which the tissues formed in the uterus following conception become cancerous), Hodgkin's lymphoma, and
  • the proliferative disease is a nonmalignant proliferation disease, including, but not limited to, atherosclerosis (including PDGFR-mediated atherosclerosis), restenosis following vascular angioplasty (including PDGFR-mediated restenosis), and fibroproliferative disorders (including obliterative bronchiolitis and idiopathic myelofibrosis).
  • atherosclerosis including PDGFR-mediated atherosclerosis
  • restenosis following vascular angioplasty including PDGFR-mediated restenosis
  • fibroproliferative disorders including obliterative bronchiolitis and idiopathic myelofibrosis.
  • the proliferative disease is an inflammatory disease or disorder related to immune dysfunction, immunodeficiency, or immunomodulation, including, but not limited to, autoimmune diseases, tissue transplant rejection, graft-versus-host disease, wound healing, kidney disease, multiple sclerosis, thyroiditis, type 1 diabetes, sarcoidosis, allergic rhinitis, inflammatory bowel diseases (including Crohn's disease and ulcerative colitis (UC)), systemic lupus erythematosis (SLE), arthritis, osteoarthritis, rheumatoid arthritis, osteoporosis, asthma, and chronic obstructive pulmonary disease (COPD).
  • autoimmune diseases tissue transplant rejection, graft-versus-host disease, wound healing, kidney disease, multiple sclerosis, thyroiditis, type 1 diabetes, sarcoidosis, allergic rhinitis, inflammatory bowel diseases (including Crohn's disease and ulcerative colitis (UC)), systemic lupus erythematosis (SLE), arthritis,
  • the proliferative disease is an infectious disease.
  • the infectious disease is fungal infection.
  • the infectious disease is a superficial mycose (e.g., Tinea versicolor).
  • the infectious disease is a cutaneous mycose (e.g., epidermis).
  • the infectious disease is a subcutaneous mycose.
  • the infectious disease is a systemic mycose.
  • the proliferative disease is leukemia, adult T-cell leukemia, promyelocytic leukemia, pre-B cell leukemia, lymphoma, Mantle cell lymphoma, breast cancer, pancreatic cancer, prostate cancer, head and neck cancer, ovarian cancer, melanoma, glioma, liver cancer, renal cancer, colorectal cancer, rhabdomyosarcoma, tongue cancer, stomach cancer, multiple myeloma, bladder cancer, thyroid cancer, epidermoid carcinoma, lung cancer, NSC lung cancer, or large cell lung cancer.
  • the proliferative disease is adult T-cell leukemia, promyelocytic leukemia, pre-B cell leukemia, lymphoma, mantle cell lymphoma, pancreatic cancer, prostate cancer, head and neck cancer, ovarian cancer, melanoma, glioma, liver cancer, renal cancer, colorectal cancer, rhabdomyosarcoma, tongue cancer, stomach cancer, multiple myeloma, bladder cancer, thyroid cancer, epidermoid carcinoma, NSC lung cancer, or large cell lung cancer.
  • the proliferative disease is leukemia, adult T-cell leukemia, promyelocytic leukemia, pre-B cell leukemia, lymphoma, mantle cell lymphoma, breast cancer, head and neck cancer, ovarian cancer, colorectal cancer, tongue cancer, multiple myeloma, or large cell lung cancer.
  • the cancers which can be treated by the compounds described herein include, but are not limited to, Acute Lymphoblastic Leukemia; Acute Myeloid Leukemia; Adrenocortical Carcinoma; AIDS-Related Lymphoma; AIDS-Related Malignancies; Anal Cancer; Astrocytoma; Bile Duct Cancer; Bladder Cancer; Bone Cancer, Osteosarcoma/Malignant Fibrous Histiocytoma; Brain Stem Glioma; Brain Tumor, Cerebellar Astrocytoma; Brain Tumor, Cerebral Astrocytoma/Malignant Glioma; Brain Tumor, Ependymoma; Brain Tumor, Medulloblastoma; Brain Tumor, Supratentorial Primitive Neuroectodermal Tumors; Brain Tumor, Visual Pathway and Hypothalamic Glioma; Breast Cancer; Bronchial Adenomas/Carcinoids; Carcinoid
  • a cancer potentially associated with mutant IDH enzyme activity is brain cancer, such as an astrocytic tumor (e.g., pilocytic astrocytoma, subependymal giant-cell astrocytoma, diffuse astrocytoma, pleomorphic xanthoastrocytoma, anaplastic astrocytoma, astrocytoma, giant cell glioblastoma, glioblastoma, secondary glioblastoma, primary adult glioblastoma, and primary pediatric glioblastoma); oligodendroglial tumor (e.g., oligodendroglioma, and anaplastic oligodendroglioma); oligoastrocytic tumor (e.g., oligoastrocytoma, and anaplastic oligoastrocytoma); ependymoma (e.g., myxopapillary ependymoma),
  • the brain cancer is glioma, glioblastoma multiforme, paraganglioma, or suprantentorial primordial neuroectodermal tumors (sPNET).
  • a cancer potentially associated with mutant IDH enzyme activity is leukemia, such as acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML), myeloproliferative neoplasm (MPN), post-MPN AML, post-MDS AML, del(5q)-associated high risk MDS or AML, blast-phase chronic myelogenous leukemia, angioimmunoblastic lymphoma and acute lymphoblastic leukemia.
  • AML acute myeloid leukemia
  • MDS myelodysplastic syndrome
  • CML chronic myelogenous leukemia
  • MPN myeloproliferative neoplasm
  • post-MPN AML post-MPN AML
  • a cancer potentially associated with mutant IDH enzyme activity is skin cancer, including melanoma.
  • a cancer potentially associated with mutant IDH enzyme activity is prostate cancer, breast cancer, thyroid cancer, colon cancer, or lung cancer.
  • a cancer potentially associated with mutant IDH enzyme activity is sarcoma, including central chondrosarcoma, central and periosteal chondroma, and fibrosarcoma.
  • a cancer potentially associated with mutant IDH enzyme activity is cholangiocarcinoma.
  • the subject to be treated with one of the methods provided herein has not been treated with anticancer therapy for the proliferative disease to be treated prior to the administration of a compound provided herein, e.g., a compound of Formulas I, II, or Ila, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof
  • the subject to be treated with one of the methods provided herein has been treated with anticancer therapy for the proliferative disease to be treated prior to the administration of a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof
  • the subject to be treated with one of the methods provided herein has developed drug resistance to the anticancer therapy.
  • the methods provided herein encompass treating a subject regardless of patient's age, although some diseases or disorders are more common in certain age groups. Further provided herein is a method for treating a subject who has undergone surgery in an attempt to treat the disease or condition at issue, as well as the one who have not. Because the subjects with cancer have heterogeneous clinical manifestations and varying clinical outcomes, the treatment given to a particular subject may vary, depending on his/her prognosis. The skilled clinician will be able to readily determine without undue experimentation, specific secondary agents, types of surgery, and types of non-drug based standard therapy that can be effectively used to treat an individual subject with cancer.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, CIV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration.
  • parenteral e.g., intramuscular, intraperitoneal, intravenous, CIV, intracistemal injection or infusion, subcutaneous injection, or implant
  • parenteral e.g., intramuscular, intra
  • a compound provided herein e.g., an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, may be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, carriers, adjuvants and vehicles, appropriate for each route of administration.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered orally.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered parenterally.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered intravenously.
  • a compound provided herein e.g., a compound of a diastereomer, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered intramuscularly.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered subcutaneously.
  • a compound provided herein e.g., a compound of Formulas I, Ha, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered topically.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, can be delivered as a single dose such as, e.g., a single bolus injection, or oral tablets or pills; or over time such as, e.g., continuous infusion over time or divided bolus doses over time.
  • a single dose such as, e.g., a single bolus injection, or oral tablets or pills
  • time such as, e.g., continuous infusion over time or divided bolus doses over time.
  • stable disease for solid tumors generally means that the perpendicular diameter of measurable lesions has not increased by 25% or more from the last measurement.
  • Stable disease or lack thereof is determined by methods known in the art such as evaluation of patient symptoms, physical examination, visualization of the tumor that has been imaged using X-ray, CAT, PET, or MRI scan and other commonly accepted evaluation modalities.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, can be administered once daily (QD), or divided into multiple daily doses such as twice daily (BID), and three times daily (TID). In addition, the administration can be continuous, i.e., every day, or intermittently.
  • intermittent administration of a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administration for one to six days per week, administration in cycles ⁇ e.g., daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week), or administration on alternate days.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is cyclically administered to a patient. Cycling therapy involves the administration of an active agent for a period of time, followed by a rest for a period of time, and repeating this sequential administration. Cycling therapy can reduce the development of resistance to one or more of the therapies, avoid or reduce the side effects of one of the therapies, and/or improves the efficacy of the treatment.
  • a combination comprising an effective amount of a compound as provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, or a tautomeric, or polymorphic form thereof and one, two, three or more other therapeutic agents, e.g. anti-cancer agents.
  • a compound as provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, or a tautomeric, or polymorphic form thereof and one, two, three or more other therapeutic agents, e.g. anti-cancer agents.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, can also be combined or used in combination with other therapeutic agents useful in the treatment and/or prevention of a disease described herein.
  • the term "in combination” includes the use of more than one therapy (e.g., one or more prophylactic and/or therapeutic agents).
  • a first therapy e.g., a prophylactic or therapeutic agent such as a compound provided herein
  • a first therapy can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy (e.g., a prophylactic or therapeutic agent) to
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is independent of the route of administration of a second therapy.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered orally.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered intravenously.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered orally or intravenously, and the second therapy can be administered orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery by catheter or stent, subcutaneously, intraadiposally, intraarticularly, intrathecally, or in a slow release dosage form.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, and a second therapy are administered by the same mode of administration, orally or by IV.
  • a compound provided herein e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered by one mode of administration, e.g., by IV, whereas the second agent (an anticancer agent) is administered by another mode of administration, e.g., orally.
  • each method provided herein may independently, further comprise the step of administering a second therapeutic agent.
  • the second therapeutic agent is an anticancer agent.
  • the anticancer agent is an antimetabolite, including, but not limited to, 5-fluoro uracil, methotrexate, cytarabine (also known as cytosine arabinoside or Ara-C), and HDAC (high dose cytarabine) and fludarabine.
  • the anticancer agent is an antimicrotubule agent, including, but not limited to, vinca alkaloids (e.g., vincristine and vinblastine) taxanes (e.g., paclitaxel and docetaxel), and epothilones and their derivatives (e.g., ixabepilone).
  • the anticancer agent is an alkylating agent, including, but not limited to, cyclophosphamide, melphalan, carmustine, and nitrosoureas (e.g., bischloroethylnitrosurea and hydroxyurea).
  • the anticancer agent is a platinum agent, including, but not limited to, cisplatin, carbop latin, oxalip latin, satraplatin (JM-216), and CI-973.
  • the anticancer agent is an anthracycline, including, but not limited to, doxrubicin and daunorubicin.
  • the anticancer agent is an antitumor antibiotic, including, but not limited to, mitomycin, idarubicin, adriamycin, and daunomycin (also known as daunorubicin).
  • the anticancer agent is a topoisomerase inhibitor, e.g., etoposide and camptothecins.
  • the anticancer agent is selected from the group consisting of adriamycin, busulfan, cytarabine, cyclophosphamide, dexamethasone, fludarabine, fluorouracil, hydroxyurea, interferons, oblimersen, platinum derivatives, taxol, topotecan, and vincristine.
  • the anticancer agent is a Bcr-Abl kinase inhibitor.
  • the Bcr-Abl kinase inhibitor is imatinib, BMS354825 (dasatinib), AMN107 (nilotinib), AP23464, AZD0530, CGP76030, ON012380, INN-0406 (NS-187), SKI-606 (bosutimb), VX- 680, or pyrrolo[2,3-d]pyrimidines including PD166326, PD173955 and PD180970.
  • the Bcr-Abl kinase inhibitor is imatinib.
  • the Bcr-Abl kinase inhibitor is dasatinib. In yet another embodiment, the Bcr-Abl kinase inhibitor is nilotinib. In yet another embodiment, the Bcr-Abl kinase inhibitor is AP23464. In yet another embodiment, the Bcr-Abl kinase inhibitor is AZD0530. In yet another embodiment, the Bcr-Abl kinase inhibitor is CGP76030. In yet another embodiment, the Bcr-Abl kinase inhibitor is SKI- 606. In yet another embodiment, the Bcr-Abl kinase inhibitor is ON012380.
  • the Bcr-Abl kinase inhibitor is INN-0406 (NS-187). In yet another embodiment, the Bcr-Abl kinase inhibitor is a pyrrolo[2,3-d]pyrimidine. In another embodiment, the Bcr-Abl kinase inhibitor is VX-680. In another embodiment, the Bcr-Abl kinase inhibitor is PD166326. In yet another embodiment, the Bcr-Abl kinase inhibitor is PD173955. In still another embodiment, the Bcr-Abl kinase inhibitor is PD 180970.
  • the anticancer agent is a FLT3 kinase inhibitor.
  • the FLT3 kinase inhibitor is PKC 412, MLN 578, CEP-701, CT 53518, CT-53608, CT-52923, D-64406, D-65476, AGL-2033, AG1295, AG1296, KN-1022, PKC-412, SU5416, SU5614, SU11248, L-00021649, or CHIR-258.
  • the FLT3 kinase inhibitor is PKC 412.
  • the FLT3 kinase inhibitor is MLN 578.
  • the FLT3 kinase inhibitor is CEP-701.
  • the FLT3 kinase inhibitor is CT 53518. In yet another embodiment, the FLT3 kinase inhibitor is CT- 53608. In yet another embodiment, the FLT3 kinase inhibitor is CT-52923. In yet another embodiment, the FLT3 kinase inhibitor is D-64406. In yet another embodiment, the FLT3 kinase inhibitor is D-65476. In yet another embodiment, the FLT3 kinase inhibitor is AGL-2033. In yet another embodiment, the FLT3 kinase inhibitor is AG1295. In yet another embodiment, the FLT3 kinase inhibitor is AG1296. In yet another embodiment, the FLT3 kinase inhibitor is KN-1022.
  • the FLT3 kinase inhibitor is KN-1022. In yet another embodiment, the FLT3 kinase inhibitor is SU5416. In yet another embodiment, the FLT3 kinase inhibitor is SU5614. In yet another embodiment, the FLT3 kinase inhibitor is SU1 1248. In yet another embodiment, the FLT3 kinase inhibitor is L-00021649. In still another embodiment, the FLT3 kinase inhibitor is CFIIR-258.
  • therapies or anticancer agents that may be used in combination with a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, include surgery, radiotherapy (e.g., gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, biologic response modifiers (e.g., interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, alkylating drugs (mechlorethamine, chlorambucil, cyclophospham
  • the other anticancer agent is selected from the group consisting of vascular endothelial growth factor (VEGF) receptor inhibitors, topoisomerase II inhibitors, smoothen inhibitors, alkylating agents, anti-tumor antibiotics, anti-metabolites, retinoids, immunomodulatory agents including but not limited to anti-cancer vaccines, CTLA-4, LAG-3, PD-1 antagonists and BET bromodomain inhibitors.
  • VEGF vascular endothelial growth factor
  • vascular endothelial growth factor (VEGF) receptor inhibitors include, but are not limited to, bevacizumab (sold under the trademark AVASTIN by Genentech/Roche), axitinib, (N-methyl-2-[[3-[([pound])-2-pyridin-2-ylethenyl]-l H-indazol-6- yl]sulfanyl]benzamide, also known as AG013736, and described in PCT Publication No.
  • topoisomerase II inhibitors include but are not limited to, etoposide (also known as VP-16 and Etoposide phosphate, sold under the tradenames TOPOSAR, VEPESID and ETOPOPHOS), and teniposide (also known as VM-26, sold under the tradename VUMON).
  • etoposide also known as VP-16 and Etoposide phosphate, sold under the tradenames TOPOSAR, VEPESID and ETOPOPHOS
  • teniposide also known as VM-26, sold under the tradename VUMON.
  • alkylating agents include but are not limited to, 5-azacytidine (sold under the trade name VIDAZA), decitabine (sold under the trade name of DECOGEN), temozolomide (sold under the trade names TEMODAR and TEMODAL by Schering- Plough/Merck), dactinomycin (also known as actinomycin-D and sold under the tradename COSMEGEN), melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, sold under the tradename ALKERAN), altretamine (also known as hexamethylmelamine (HMM), sold under the tradename HEXALEN), carmustine (sold under the tradename BCNU), bendamustine (sold under the tradename TREANDA), busulfan (sold under the tradenames Busulfex(R) and Myleran(R)), carboplatin (sold under the tradename Paraplatin(R)), lomustine (also known as
  • anti-tumor antibiotics include, but are not limited to, doxorubicin (sold under the tradenames Adriamycin(R) and Rubex(R)), bleomycin (sold under the tradename lenoxane(R)), daunorubicin (also known as dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, sold under the tradename Cerubidine(R)), daunorubicin liposomal (daunorubicin citrate liposome, sold under the tradename DaunoXome(R)), mitoxantrone (also known as DHAD, sold under the tradename Novantrone(R)), epirubicin (sold under the tradename Ellence(TM)), idarubicin (sold under the tradenames Idamycin(R), Idamycin PFS(R)), and mitomycin C (sold under the tradename Mutamycin(R)).
  • doxorubicin sold under the tradename
  • anti-metabolites include, but are not limited to, claribine (2- chlorodeoxyadenosine, sold under the tradename leustatin(R)), 5-fluorouracil (sold under the tradename Adrucil(R)), 6-thioguanine (sold under the tradename Purinethol(R)), pemetrexed (sold under the tradename Alimta(R)), cytarabine (also known as arabinosylcytosine (Ara-C), sold under the tradename Cytosar-U(R)), cytarabine liposomal (also known as Liposomal Ara-C, sold under the tradename DepoCyt(TM)), decitabine (sold under the tradename Dacogen(R)), hydroxyurea (sold under the tradenames Hydrea(R), Droxia(TM) and Mylocel(TM)), fludarabine (sold under the tradename Fludara(R)), floxuridine (s
  • retinoids examples include, but are not limited to, alitretinoin (sold under the tradename Panretin(R)), tretinoin (all-trans retinoic acid, also known as ATRA, sold under the tradename Vesanoid(R)), Isotretinoin (13-c/s-retinoic acid, sold under the tradenames Accutane(R), Amnesteem(R), Claravis(R), Clarus(R), Decutan(R), Isotane(R), Izotech(R), Oratane(R), Isotret(R), and Sotret(R)), and bexarotene (sold under the tradename Targretin(R)).
  • PD-1 antagonist means any chemical compound or biological molecule that blocks binding of PD-L1 expressed on a cancer cell to PD-1 expressed on an immune cell (T cell, B cell or NKT cell) and preferably also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1.
  • Alternative names or synonyms for PD-1 and its ligands include: PDCD1, PD1, CD279 and SLEB2 for PD-1 ; PDCD1L1, PDL1, B7H1, B7-4, CD274 and B7-H for PD-L1 ; and PDCD1L2, PDL2, B7-DC, Btdc and CD273 for PD-L2.

Abstract

Provided herein are compounds, compositions, and methods for the treatment of cancer, including leukemia. In one embodiment, compounds and compositions of nucleoside derivatives are disclosed, which can be administered either alone or in combination with other anti-cancer agents.

Description

NUCLEOSIDE DERIVATIVES FOR THE TREATMENT OF CANCER
FIELD
Provided herein are nucleoside derivatives, pharmaceutical compositions comprising the compounds, processes of preparation thereof, and methods of use thereof for treating cancer.
BACKGROUND
Cancer is a disease characterized primarily by an uncontrolled divisions of abnormal cells derived from a given normal tissue and the invasion of adjacent tissues by these malignant cells. Blood or lymphatic transportation can spread cancer cells to other parts of the body leading to regional lymph nodes and to distant sites (metastasis). Cancer is a complex, multistep process that begins with minor preneoplastic changes, which may under certain conditions progress to neoplasia. There are more than 100 different types of cancer, which can be grouped into broader categories. The main categories include: carcinoma, sarcoma, leukemia, lymphoma and myeloma, and central nervous system cancers. The incidence of cancer continues to climb as the general population ages, as new cancers develop, and as susceptible populations (e.g., people infected with AIDS or excessively exposed to sunlight) grow. A tremendous demand therefore exists for new methods and compositions that can be used to treat patients with cancer.
Hematologic or hematopoietic malignancies are cancers of the blood or bone marrow, including leukemia and lymphoma. Leukemia is a type of cancer of the blood characterized by abnormal accumulation of immature white blood cells. There are four types of leukemia: acute lymphocytic leukemia (ALL), acute myelogenous leukemia (AML), chronic lymphocytic leukemia (CLL), and chronic myelogenous leukemia (CML). Acute leukemia is a rapidly progressing disease that results in the accumulation of immature, functionless cells in the marrow and blood. The marrow often stops producing enough normal red cells, white cells and platelets. On the other hand, chronic leukemia progresses more slowly and allows greater numbers of more mature, functional cells to be made.
Leukemia can affect people at any age. The cause of most cases of leukemia is not known. Extraordinary doses of radiation and certain cancer therapies are possible causes. About 90% of leukemia are diagnosed in adults. In 2000 approximately 256,000 children and adults around the world developed some form of leukemia, and 209,000 died from it. Cases of Chronic leukemia account for 4.5 percent more cases than acute leukemia. The most common types of leukemia in adults are acute myelogenous leukemia (AML), with estimated 14,590 new cases in 2013, and chronic lymphocytic leukemia (CLL), with about 15,680 new cases in 2013. Chronic myelogenous leukemia (CML) was estimated to affect about 5,920 persons in 2013 (data from the Leukemia and Lymphoma Society, Facts 2013, August 2013).
The dramatic improvement in blood cancer treatment in the latter part of the 20th century is largely the result of chemotherapy. In addition, there are more than 50 drugs individually used to treat these disorders and a number of potential new therapies are under investigation in clinical trials. While current chemotherapy can result in complete remissions, the long term disease-free survival rate for leukemia, in particular AML, is low. For example, the overall relative survival rate for AML was estimated to be about 59% from 2003 to 2009. Therefore, there is a clear and unmet need for effective therapeutics for treatment of blood cancers, including leukemia.
SUMMARY OF THE DISCLOSURE
Derivatives, in one embodiment phosphoramidate derivatives and cyclic phosphate derivatives, of a variety of therapeutic agents are provided, as well as pharmaceutical compositions and methods of treatment of a variety of cancer including hematopoietic malignancies. The therapeutic agent is, for example, an anti-cancer agent that includes, or has been derivatized to include, a reactive group, such as a hydroxyl, for attachment of the phosphoroamidate and cyclic phosphate moiety. Such therapeutic agents include, but are not limited to nucleosides and nucleoside analogs including acyclic nucleosides. A nucleoside analog refers to a structurally modified nucleoside. In one embodiment, the nucleosides or nucleoside analogs are derivitized at the 5'- and/or the 3 '-position by removal of a hydrogen from an hydroxyl group to incorporate a phosphoramidate and cyclic phosphate group. In one embodiment, the nucleosides or nucleoside analogs are derivitized at the 5'- and/or the 2'-position by removal of a hydrogen from an hydroxyl group to incorporate a phosphoramidate and cyclic phosphate group. In one embodiment, phosphorodiamidates of nucleosides and nucleoside analogs are provided. In other embodiments, the compound is a -S'-pivaloyl-2-thioethyl phosphoroamidate or S- hydroxypivaloyl-2-thioethyl phosphoroamidate. In another embodiment, <S'-pivaloyl-2- thioethyl disulfide phosphoroamidate or ^^-dimethyl-S-oxopropanoate 2-thioethyl benzyl phosphoroamidate or benzylphosphoramidate of nucleosides and nucleoside analogs are provided. In other embodiments, the derivative is l-methyl-2-nitro-lH- imidazol-5-methoxymethyl N- benzylphosphoramidate or 5-nitrofuran-2-methoxymethyl N-benzyl phosphoramidate. In one embodiment the derivative is a 3', 5'-cyclic phosphate. In another embodiment the derivative is a 2', 5'-cyclic phosphate.
Phosphoroamidate and cyclic phosphate compounds of a variety of anti-cancer agents are provided. The anti-cancer agents include, but are not limited to, modified nucleosides. In another embodiment, the anti-cancer agent is clofarabine, cytarabine, isocladribine, cladribine, fludarabine or nelarabine. In another embodiment, the anti-cancer agent is clofarabine, cytarabine, isocladribine, cladribine or fludarabine. In another embodiment, the anti-cancer agent is clofarabine or cytarabine.
DETAILED DESCRIPTION OF THE INVENTION
Definitions
To facilitate understanding of the disclosure set forth herein, a number of terms are defined below.
Generally, the nomenclature used herein and the laboratory procedures in organic chemistry, medicinal chemistry, and pharmacology described herein are those well known and commonly employed in the art. Unless defined otherwise, all technical and scientific terms used herein generally have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs.
The term "alkyl", as used herein, unless otherwise specified, refers to a saturated straight or branched hydrocarbon. In one embodiment, the alkyl group is a primary, secondary, or tertiary hydrocarbon. In one embodiment, the alkyl group includes one to ten carbon atoms, i.e.,
Figure imgf000005_0001
to C10 alkyl. In one embodiment, the alkyl group is methyl, CF3, CCI3, CFCI2, CF2C1, ethyl, CH2CF3, CF2CF3, propyl, isopropyl, butyl, isobutyl, sec-butyl, t- butyl, pentyl, isopentyl, neopentyl, hexyl, isohexyl, 3-methylpentyl, 2,2-dimethylbutyl, or 2,3-dimethylbutyl. The term includes both substituted and unsubstituted alkyl groups, including halogenated alkyl groups, preferably unsubstituted or halogenated alkyl groups. In one embodiment, the alkyl group is a fluorinated alkyl group. Non-limiting examples of moieties with which the alkyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
The term "lower alkyl", as used herein, and unless otherwise specified, refers to a saturated straight or branched hydrocarbon having one to six carbon atoms, i.e., Ci to C6 alkyl. In one embodiment, the lower alkyl group is a primary, secondary, or tertiary hydrocarbon. The term includes both substituted and unsubstituted moieties, preferably unsubstituted
The term "cycloalkyl", as used herein, unless otherwise specified, refers to a saturated cyclic hydrocarbon. In one embodiment, the cycloalkyl group may be a saturated, and/or bridged, and/or non-bridged, and/or a fused bicyclic group. In one embodiment, the cycloalkyl group includes three to ten carbon atoms, i.e., C3 to C10 cycloalkyl. In some embodiments, the cycloalkyl has from 3 to 15 (C3-15), from 3 to 10 (C3-10), or from 3 to 7 (C3-7) carbon atoms. In one embodiment, the cycloalkyl group is cyclopropyl, cyclobutyl, cyclopentyl, cyclopentylmethyl, cyclohexyl, cyclohexylmethyl, cycloheptyl, bicyclo[2.1.1]hexyl, bicyclo[2.2.1 ]heptyl, decalinyl, or adamantyl. The term includes both substituted and unsubstituted cycloalkyl groups, including halogenated cycloalkyl groups. Non-limiting examples of moieties with which the cycloalkyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference. In preferred embodiments, the cycloalkyl is unsubstituted or substituted with one or more halogens.
"Alkenyl" refers to monovalent olefinically unsaturated hydrocarbon groups, in certain embodiment, having up to about 11 carbon atoms, from 2 to 8 carbon atoms, or from 2 to 6 carbon atoms, which can be straight-chained or branched and having at least 1 or from 1 to 2 sites of olefinic unsaturation. The term "alkenyl" embraces radicals having a "CJV or "trans" configuration or a mixture thereof, or alternatively, a "Z" or "E" configuration or a mixture thereof, as appreciated by those of ordinary skill in the art. For example, C2-6 alkenyl refers to a linear unsaturated monovalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenyl is a linear monovalent hydrocarbon radical of 2 to 20 (C2-20), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched monovalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of alkenyl groups include, but are not limited to, ethenyl, propen-l-yl, propen-2-yl, allyl, butenyl, and 4-methylbutenyl. The term includes both substituted and unsubstituted alkenylene groups, including halogenated cycloalkyl groups, preferably unsubstituted or halogenated cycloalkyl. Non-limiting examples of moieties with which the alkenylene group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
The term "cycloalkenyl", as used herein, unless otherwise specified, refers to an unsaturated cyclic hydrocarbon and includes both substituted and unsubstituted cycloalkenyl groups, preferably unsubstituted. Non-limiting examples of moieties with which the cycloalkenyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
The term "alkenylene" refers to a linear or branched divalent hydrocarbon radical, which contains one or more, in one embodiment, one, two, three, four, or five, in another embodiment, one or two, carbon-carbon double bond(s). The term "alkenylene" embraces radicals having a "c/V or "trans'" configuration or a mixture thereof, or alternatively, a "Z" or "E" configuration or a mixture thereof, as appreciated by those of ordinary skill in the art. For example, C2-6 alkenylene refers to a linear unsaturated divalent hydrocarbon radical of 2 to 6 carbon atoms or a branched unsaturated divalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkenylene is a linear divalent hydrocarbon radical of 2 to 20 (C2-2o), 2 to 15 (C2-15), 2 to 10 (C2-10), or 2 to 6 (C2-6) carbon atoms, or a branched divalent hydrocarbon radical of 3 to 20 (C3-20), 3 to 15 (C3-15), 3 to 10 (C3-10), or 3 to 6 (C3-6) carbon atoms. Examples of alkenylene groups include, but are not limited to, ethenylene, allylene, propenylene, butenylene, and 4-methylbutenylene. The term includes both substituted and unsubstituted groups, including halogenated groups, preferably unsubstituted. Non-limiting examples of moieties with which the alkenylene group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxy carbonyl, aryloxy carbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al , Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
"Alkynyl" refers to acetylenically unsaturated hydrocarbon groups, in certain embodiments, having up to about 11 carbon atoms or from 2 to 6 carbon atoms which can be straight-chained or branched and having at least 1 or from 1 to 2 sites of alkynyl unsaturation. Non-limiting examples of alkynyl groups include acetylenic, ethynyl (- C≡CH), propargyl (-CI¾C≡CH), and the like. The term also includes both substituted and unsubstituted alkynyl groups, preferably unsubstituted. Non-limiting examples of moieties with which the alkynyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et al, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
The term "aryl", as used herein, and unless otherwise specified, refers to phenyl, biphenyl, or naphthyl. The term includes both substituted and unsubstituted moieties. An aryl group can be substituted with any described moiety, including, but not limited to, one or more moieties selected from halogen (fluoro, chloro, bromo, or iodo), alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, cycloakenyl, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
"Alkoxy" refers to the group -OR' where R' is alkyl or cycloalkyl where alkyl and cycloalkyl are as defined herein. Alkoxy groups include, by way of example, methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, tert-butoxy, sec-butoxy, n-pentoxy, n-hexoxy, 1,2-dimethylbutoxy, and the like.
"Aryloxy" refers to the group -OR' where R' is aryl or heteroaryl where aryl and heteroaryl are as defined herein.
"Alkoxycarbonyl" refers to a radical -C(0)-alkoxy where alkoxy is as defined herein.
"Amino" refers to the radical -NH2.
"Carboxyl" or "carboxy" refers to the radical -C(0)OH. "Carbonyl" refers to the radical - C(O)-.
The term "alkylamino," "arylamino," or "alkylarylamino" refers to an amino group that has one or two alkyl substituents (-NHR' or -NR'R', where R' is alkyl as defined herein), one or two aryl substituents (-NHR' or - NR'R', where R' is aryl as defined herein), or one alkyl substituent and one aryl substituent (-NR'R", where one of R' and R" is alkyl as defined herein and the other is aryl as defined herein), respectively. In one embodiment, the alkyl substituent is lower alkyl. In another embodiment, the alkyl or lower alkyl is unsubstituted.
"Halogen" or "halo" refers to fluoro, chloro, bromo, or iodo.
"Oxo" refers to =0 or— >Ό. "Thioalkoxy" refers to the group -SR' where R' is alkyl or cycloalkyl each of which is as defined herein. "Thioaryloxy" refers to the group -SR' where R' is aryl or heteroaryl each of which is as defined herein.
"Alkyldisulfanyl" refers to the group R'-S-S- where R' is alkyl or cycloalkyl each of which is as defined herein.
The term "heterocyclyl" or "heterocyclic" refers to a monovalent monocyclic non- aromatic ring system and/or multicyclic ring system that contains at least one non- aromatic ring, wherein one or more of the non-aromatic ring atoms are heteroatoms independently selected from O, S, or N; and the remaining ring atoms are carbon atoms. In certain embodiments, the heterocyclyl or heterocyclic group has from 3 to 20, from 3 to 15, from 3 to 10, from 3 to 8, from 4 to 7, or from 5 to 6 ring atoms. Heterocyclyl groups are bonded to the rest of the molecule through the non-aromatic ring. In certain embodiments, the heterocyclyl is a monocyclic, bicyclic, tricyclic, or tetracyclic ring system, which may include a fused or bridged ring system, and in which the nitrogen or sulfur atoms may be optionally oxidized, the nitrogen atoms may be optionally quaternized, and some rings may be partially or fully saturated, or aromatic. The heterocyclyl may be attached to the main structure at any heteroatom or carbon atom which results in the creation of a stable compound. Examples of such heterocyclic radicals include, but are not limited to, azepinyl, benzodioxanyl, benzodioxolyl, benzofuranonyl, benzopyranonyl, benzopyranyl, benzotetrahydrofuranyl, benzotetrahydrothienyl, benzothiopyranyl, benzoxazinyl, β-carbolinyl, chromanyl, chromonyl, cinnolinyl, coumarinyl, decahydroisoquinolinyl, dihydrobenzisothiazinyl, dihydrobenzisoxazinyl, dihydrofuryl, dihydroisoindolyl, dihydropyranyl, dihydropyrazolyl, dihydropyrazinyl, dihydropyridinyl, dihydropyrimidinyl, dihydropyrrolyl, dioxolanyl, 1 ,4-dithianyl, furanonyl, imidazolidinyl, imidazolinyl, indolinyl, isobenzotetrahydrofuranyl, isobenzotetrahydrothienyl, isochromanyl, isocoumarinyl, isoindolinyl, isothiazolidinyl, isoxazolidinyl, morpholinyl, octahydroindolyl, octahydroisoindolyl, oxazolidinonyl, oxazolidinyl, oxiranyl, piperazinyl, piperidinyl, 4-piperidonyl, pyrazolidinyl, pyrazolinyl, pyrrolidinyl, pyrrolinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydropyranyl, tetrahydrothienyl, thiamorpholinyl, thiazolidinyl, tetrahydroquinolinyl, and 1 ,3,5- trithianyl. The term also includes both substituted and unsubstituted heterocyclyl groups. Non-limiting examples of moieties with which the heterocyclyl group can be substituted include halogen (fluoro, chloro, bromo, or iodo), oxo, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaroxyl, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991, hereby incorporated by reference.
The term "heteroaryl" refers to a monovalent monocyclic aromatic group and/or multicyclic aromatic group that contain at least one aromatic ring, wherein at least one aromatic ring contains one or more heteroatoms independently selected from O, S, and N in the ring. Heteroaryl groups are bonded to the rest of the molecule through the aromatic ring. Each ring of a heteroaryl group can contain one or two O atoms, one or two S atoms, and/or one to four N atoms, provided that the total number of heteroatoms in each ring is four or less and each ring contains at least one carbon atom. In certain embodiments, the heteroaryl has from 5 to 20, from 5 to 15, or from 5 to 10 ring atoms. Examples of monocyclic heteroaryl groups include, but are not limited to, furanyl, imidazolyl, isothiazolyl, isoxazolyl, oxadiazolyl, oxadiazolyl, oxazolyl, pyrazinyl, pyrazolyl, pyridazinyl, pyridyl, pyrimidinyl, pyrrolyl, thiadiazolyl, thiazolyl, thienyl, tetrazolyl, triazinyl, and triazolyl. Examples of bicyclic heteroaryl groups include, but are not limited to, benzofuranyl, benzimidazolyl, benzoisoxazolyl, benzopyranyl, benzothiadiazolyl, benzothiazolyl, benzothienyl, benzotriazolyl, benzoxazolyl, furopyridyl, imidazopyridinyl, imidazothiazolyl, indolizinyl, indolyl, indazolyl, isobenzofuranyl, isobenzothienyl, isoindolyl, isoquinolinyl, isothiazolyl, naphthyridinyl, oxazolopyridinyl, phthalazinyl, pteridinyl, purinyl, pyridopyridyl, pyrrolopyridyl, quinolinyl, quinoxalinyl, quinazolinyl, thiadiazolopyrimidyl, and thienopyridyl. Examples of tricyclic heteroaryl groups include, but are not limited to, acridinyl, benzindolyl, carbazolyl, dibenzofuranyl, perimidinyl, phenanthrolinyl, phenanthridinyl, phenarsazinyl, phenazinyl, phenothiazinyl, phenoxazinyl, and xanthenyl. A heteroaryl group can be substituted with any described moiety, including, but not limited to, one or more moieties selected from halogen (fluoro, chloro, bromo, or iodo), oxo, alkyl, haloalkyl, alkenyl, alkynyl, cycloalkyl, cycloakenyl, hydroxyl, amino, alkylamino, arylamino, alkylarylamino, alkoxy, aryloxy, thioalkoxy, thioaryloxy, alkyldisulfanyl, acyl, hydroxylcarbonyl, alkoxycarbonyl, aryloxycarbonyl, aminocarbonyl, alkylaminocarbonyl, arylaminocarbonyl, alkylarylaminocarbonyl, acyloxy, acylthio, nitro, cyano, sulfonic acid, sulfate, phosphonic acid, phosphate, or phosphonate, either unprotected, or protected as necessary, as known to those skilled in the art, for example, as taught in Greene, et ah, Protective Groups in Organic Synthesis, John Wiley and Sons, Second Edition, 1991.
The term "alkylaryl" refers to an aryl group with an alkyl substituent. The term "aralkyl" or "arylalkyl" includes an alkyl group with an aryl substituent.
The term "alkylheterocyclyl" refers to a heterocyclyl group with an alkyl substituent. The term "alkylheterocyclyl" includes an alkyl group with a heterocyclyl substituent.
The term "alkylheteroaryl" refers to a heteroaryl group with an alkyl substituent. The term "alkylheteroaryl" includes an alkyl group with a heteroaryl substituent.
The term "protecting group" as used herein and unless otherwise defined refers to a group that is added to an oxygen, nitrogen, or phosphorus atom to prevent its further reaction or for other purposes. A wide variety of oxygen and nitrogen protecting groups are known to those skilled in the art of organic synthesis. "Pharmaceutically acceptable salt" refers to any salt of a compound provided herein which retains its biological properties and which is not toxic or otherwise undesirable for pharmaceutical use. Such salts may be derived from a variety of organic and inorganic counter-ions well known in the art. Such salts include, but are not limited to: (1) acid addition salts formed with organic or inorganic acids such as hydrochloric, hydrobromic, sulfuric, nitric, phosphoric, sulfamic, acetic, trifluoroacetic, trichloroacetic, propionic, hexanoic, cyclopentylpropionic, glycolic, glutaric, pyruvic, lactic, malonic, succinic, sorbic, ascorbic, malic, maleic, fumaric, tartaric, citric, benzoic, 3-(4- hydroxybenzoyl)benzoic, picric, cinnamic, mandelic, phthalic, lauric, methanesulfonic, ethanesulfonic, 1,2-ethane-disulfonic, 2-hydroxyethanesulfonic, benzenesulfonic, 4- chlorobenzenesulfonic, 2-naphthalenesulfonic, 4-toluenesulfonic, camphoric, camphorsulfonic, 4-methylbicyclo[2.2.2]-oct-2-ene-l-carboxylic, glucoheptonic, 3- phenylpropionic, trimethylacetic, feri-butylacetic, lauryl sulfuric, gluconic, benzoic, glutamic, hydroxynaphthoic, salicylic, stearic, cyclohexylsulfamic, quinic, muconic acid and the like acids; or (2) salts formed when an acidic proton present in the parent compound either (a) is replaced by a metal ion, e.g., an alkali metal ion, an alkaline earth ion or an aluminum ion, or alkali metal or alkaline earth metal hydroxides, such as sodium, potassium, calcium, magnesium, aluminum, lithium, zinc, and barium hydroxide, ammonia, or (b) coordinates with an organic base, such as aliphatic, alicyclic, or aromatic organic amines, such as ammonia, methylamine, dimethylamine, diethylamine, picoline, ethanolamine, diethanolamine, triethanolamine, ethylenediamine, lysine, arginine, ornithine, choline, Ν,Ν'-dibenzylethylene-diamine, chloroprocaine, diethanolamine, procaine, N-benzylphenethylamine, N-methylglucamine piperazine, tris(hydroxymethyl)- aminomethane, tetramethylammonium hydroxide, and the like.
Pharmaceutically acceptable salts further include, by way of example only and without limitation, sodium, potassium, calcium, magnesium, ammonium, tetraalkylammonium, and the like, and when the compound contains a basic functionality, salts of non-toxic organic or inorganic acids, such as hydrohalides, e.g. hydrochloride and hydrobromide, sulfate, phosphate, sulfamate, nitrate, acetate, trifluoroacetate, trichloroacetate, propionate, hexanoate, cyclopentylpropionate, glycolate, glutarate, pyruvate, lactate, malonate, succinate, sorbate, ascorbate, malate, maleate, fumarate, tartarate, citrate, benzoate, 3-(4- hydroxybenzoyl)benzoate, picrate, cinnamate, mandelate, phthalate, laurate, methanesulfonate (mesylate), ethanesulfonate, 1 ,2-ethane-disulfonate, 2- hydroxyethanesulfonate, benzenesulfonate (besylate), 4-chlorobenzenesulfonate, 2- naphthalenesulfonate, 4-toluenesulfonate, camphorate, camphorsulfonate, 4- methylbicyclo[2.2.2]-oct-2-ene-l -carboxylate, glucoheptonate, 3-phenylpropionate, trimethylacetate, fert-butylacetate, lauryl sulfate, gluconate, benzoate, glutamate, hydroxynaphthoate, salicylate, stearate, cyclohexylsulfamate, quinate, muconate, and the like.
The term "purine" or "pyrimidine" base refers to, but is not limited to, adenine, guanine, adenine, hypoxanthine, 7-deazaguanine, 7-deazaadenine, 2,6-diaminopurine, 6- chloropurine, N6-alkylpurines, N^acylpurines (wherein acyl is C(0)(alkyl, aryl, alkylaryl, or arylalkyl), N6-benzylpurine, 6-halopurine, N6-vinylpurine, N6-acetylenic purine, N6- hydroxyalkyl purine, N6-alkylaminopurine, N^thioalkyl purine, N2-alkylpurines, N2-alkyl- 6-thiopurines, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, including 6-azacytosine, 2- and/or 4-mercaptopyrmidine, uracil, 5-halouracil, including 5-fluorouracil, C5-alkylpyrimidines, C5-benzylpyrimidines, C5-halopyrimidines, C5- vinylpyrimidine, C5-acetylenic pyrimidine, C5-acyl pyrimidine, C5-hydroxyalkyl purine, C5-amidopyrimidine, C5-cyanopyrimidine, C5-iodopyrimidine, C6-iodo-pyrimidine, C5-Br- vinyl pyrimidine, C6-Br-vinyl pyrimidine, C5-nitropyrimidine, C5-amino-pyrimidine, 5- azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolopyrimidinyl. Functional oxygen and nitrogen groups on the base can be protected as necessary or desired. Suitable protecting groups are well known to those skilled in the art, and include trimethylsilyl, dimethylhexylsilyl, /-butyldimethylsilyl, and t- butyldiphenylsilyl, trityl, alkyl groups, acyl groups such as acetyl and propionyl, methanesulfonyl, and p-toluenesulfonyl.
The term "acyl" or "O-linked ester" refers to a group of the formula O C(0)R', wherein R' is alkyl or cycloalkyl (including lower alkyl), carboxylate reside of amino acid, aryl including phenyl, alkaryl, arylalkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl. For example, R' can be substituted alkyl (including lower alkyl), aryl including phenyl optionally substituted with chloro, bromo, fluoro, iodo, Ci to C4 alkyl or Ci to C4 alkoxy, sulfonate esters such as alkyl or arylalkyl sulphonyl including methanesulfonyl, the mono, di or triphosphate ester, trityl or monomethoxy-trityl, substituted benzyl, alkaryl, arylalkyl including benzyl, alkoxyalkyl including methoxymethyl, aryloxyalkyl such as phenoxymethyl. Aryl groups in the esters optimally comprise a phenyl group. In particular, acyl groups include acetyl, trifluoroacetyl, methylacetyl, cyclpropylacetyl, propionyl, butyryl, hexanoyl, heptanoyl, octanoyl, neo-heptanoyl, phenylacetyl, 2-acetoxy-2-phenylacetyl, diphenylacetyl, a- methoxy-a-trifluoromethyl-phenylacetyl, bromoacetyl, 2-nitro-benzeneacetyl, 4-chloro- benzeneacetyl, 2-chloro-2,2-diphenylacetyl, 2-chloro-2 -phenylacetyl, trimethylacetyl, chlorodifluoroacetyl, perfluoroacetyl, fluoroacetyl, bromodifluoroacetyl, methoxyacetyl, 2-thiopheneacetyl, chlorosulfonylacetyl, 3-methoxyphenylacetyl, phenoxyacetyl, tert- butylacetyl, trichloroacetyl, monochloro-acetyl, dichloroacetyl, 7H-dodecafluoro- heptanoyl, perfluoro-heptanoyl, 7H-dodeca-fluoroheptanoyl, 7-chlorododecafluoro- heptanoyl, 7-chloro-dodecafluoro-heptanoyl, 7H-dodecafluoroheptanoyl, 7H-dodeca- fluoroheptanoyl, nona-fluoro-3,6-dioxa-heptanoyl, nonafluoro-3,6-dioxaheptanoyl, perfluoroheptanoyl, methoxybenzoyl, methyl 3-amino-5-phenylthiophene-2-carboxyl, 3,6- dichloro-2-methoxy-benzoyl, 4-(l, l,2,2-tetrafluoro-ethoxy)-benzoyl, 2-bromo-propionyl, omega-aminocapryl, decanoyl, n-pentadecanoyl, stearyl, 3-cyclopentyl-propionyl, 1 - benzene-carboxyl, O-acetylmandelyl, pivaloyl acetyl, 1 -adamantane-carboxyl, cyclohexane-carboxyl, 2,6-pyridinedicarboxyl, cyclopropane-carboxyl, cyclobutane- carboxyl, perfluorocyclohexyl carboxyl, 4-methylbenzoyl, chloromethyl isoxazolyl carbonyl, perfluorocyclohexyl carboxyl, crotonyl, 1 -m ethyl- 1 H-indazole-3 -carbonyl, 2- propenyl, isovaleryl, 1 -pyrrolidinecarbonyl, 4-phenylbenzoyl.
The term "amino acid" refers to naturally occurring and synthetic α, β, γ, or δ amino acids, and includes but is not limited to, amino acids found in proteins, i.e. glycine, alanine, valine, leucine, isoleucine, methionine, phenylalanine, tryptophan, proline, serine, threonine, cysteine, tyrosine, asparagine, glutamine, aspartate, glutamate, lysine, arginine and histidine. In one embodiment, the amino acid is in the L-configuration. Alternatively, the amino acid can be a derivative of alanyl, valinyl, leucinyl, isoleuccinyl, prolinyl, phenylalaninyl, tryptophanyl, methioninyl, glycinyl, serinyl, threoninyl, cysteinyl, tyrosinyl, asparaginyl, glutaminyl, aspartoyl, glutaroyl, lysinyl, argininyl, histidinyl, β- alanyl, β-valinyl, β-leucinyl, β-isoleuccinyl, β-prolinyl, β-phenylalaninyl, β-tryptophanyl, β-methioninyl, β-glycinyl, β-serinyl, β-threoninyl, β-cysteinyl, β-tyrosinyl, β-asparaginyl, β-glutaminyl, β-aspartoyl, β-glutaroyl, β-lysinyl, β-argininyl, or β-histidinyl.
The term "substantially free of or "substantially in the absence of with respect to a nucleoside composition refers to a nucleoside composition that includes at least 85 or 90% by weight, in certain embodiments 95%, 98 % , 99%, or 100% by weight, of the designated nucleoside, the designated diastereomer of such nucleoside, or the designated enantiomer of such nucleoside. In one embodiment, in the methods and compounds provided herein, the compounds are substantially free of other compounds, other nucleosides, other diastereomers, or other enantiomers that are not designated.
Similarly, the term "isolated" with respect to a nucleoside composition refers to a nucleoside composition that includes at least 85, 90%, 95%, 98%, 99%, to 100% by weight, of the nucleoside, the remainder comprising other chemical species or enantiomers.
"Solvate" refers to a compound provided herein or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of solvent bound by non-covalent intermolecular forces. Where the solvent is water, the solvate is a hydrate.
"Isotopic composition" refers to the amount of each isotope present for a given atom, and "natural isotopic composition" refers to the naturally occuring isotopic composition or abundance for a given atom. Atoms containing their natural isotopic composition may also be referred to herein as "non-enriched" atoms. Unless otherwise designated, the atoms of the compounds recited herein are meant to represent any stable isotope of that atom. For example, unless otherwise stated, when a position is designated specifically as "H" or "hydrogen", the position is understood to have hydrogen at its natural isotopic composition.
"Isotopic enrichment" refers to the percentage of incorporation of an amount of a specific isotope at a given atom in a molecule in the place of that atom's natural isotopic abundance. For example, deuterium enrichment of 1% at a given position means that 1% of the molecules in a given sample contain deuterium at the specified position. Because the naturally occurring distribution of deuterium is about 0.0156%, deuterium enrichment at any position in a compound synthesized using non-enriched starting materials is about 0.0156%. The isotopic enrichment of the compounds provided herein can be determined using conventional analytical methods known to one of ordinary skill in the art, including mass spectrometry and nuclear magnetic resonance spectroscopy.
"Isotopically enriched" refers to an atom having an isotopic composition other than the natural isotopic composition of that atom. "Isotopically enriched" may also refer to a compound containing at least one atom having an isotopic composition other than the natural isotopic composition of that atom.
As used herein, "alkyl," "cycloalkyl," "alkenyl," "cycloalkenyl," "alkynyl," "aryl," "alkoxy," "alkoxycarbonyl," "amino," "carboxyl," "alkylamino," "arylamino," "thioalkyoxy," "heterocyclyl," "heteroaryl," "alkylheterocyclyl," "alkylheteroaryl," "acyl," "aralkyl," "alkaryl," "purine," "pyrimidine," "carboxyl," and "amino acid" groups optionally comprise deuterium at one or more positions where hydrogen atoms are present, and wherein the deuterium composition of the atom or atoms is other than the natural isotopic composition.
Also as used herein, "alkyl," "cycloalkyl," "alkenyl," "cycloalkenyl," "alkynyl," "aryl," "alkoxy," "alkoxycarbonyl," "carboxyl," "alkylamino," "arylamino," "thioalkyoxy," "heterocyclyl," "heteroaryl," "alkylheterocyclyl," "alkylheteroaryl," "acyl," "aralkyl," "alkaryl," "purine," "pyrimidine," "carboxyl," and "amino acid" groups optionally comprise carbon-13 at an amount other than the natural isotopic composition. Further, "alkyl," "cycloalkyl," "alkenyl," "cycloalkenyl," "alkynyl," "aryl," "heterocyclyl," and "heteroaryl" groups that are included as examples of moieties with which a certain functional group can be substituted can further be substituted.
The term "proliferative disorder or disease" refers to unwanted cell proliferation of one or more subset of cells in a multicellular organism resulting in harm (i.e., discomfort or decreased life expectancy) to the multicellular organisms. A proliferative disorder or disease can occur in different types of animals and humans. For example, as used herein, "proliferative disorder or disease" includes neoplastic disorders and other proliferative disorders.
The term "neoplastic disorder or disease" or "cancer" refers to a tumor resulting from abnormal or uncontrolled cellular growth. Examples of neoplastic disorders include, but are not limited to, hematopoietic disorders, such as the myeloproliferative disorders, thrombocythemia, essential thrombocytosis (ET), angiogenic myeloid metaplasia, myelofibrosis (MF), myelofibrosis with myeloid metaplasia (MMM), chronic idiopathic myelofibrosis (IMF), polycythemia vera (PV), the cytopenias, and pre-malignant myelodysplastic syndromes; cancers, such as glioma cancers, lung cancers, breast cancers, colorectal cancers, prostate cancers, gastric cancers, esophageal cancers, colon cancers, pancreatic cancers, ovarian cancers, and hematologic malignancies.
The term "hematologic malignancy" refers to cancer of the body's blood-forming and immune system-the bone marrow and lymphatic tissue. Examples of hematological malignancies include, for instance, myelodysplasia, lymphomas, leukemias, lymphomas (non-Hodgkin's lymphoma), Hodgkin's disease (also called Hodgkin's lymphoma), and myeloma, such as acute lymphocytic leukemia (ALL), acute myeloid leukemia (AML), acute promyelocytic leukemia (APL), chronic lymphocytic leukemia (CLL), chronic myeloid leukemia (CML), chronic neutrophilic leukemia (CNL), acute undifferentiated leukemia (AUL), anaplastic large-cell lymphoma (ALCL), prolymphocytic leukemia (PML), juvenile myelomonocyctic leukemia (JMML), adult T-cell ALL, AML with trilineage myelodysplasia (AML/TMDS), mixed lineage leukemia (MLL), myelodysplasia syndromes (MDSs), myeloproliferative disorders (MPD), and multiple myeloma (MM).
The term "leukemia" refers to malignant neoplasms of the blood-forming tissues, including, but not limited to, chronic lymphocytic leukemia, chronic myelocytic leukemia, acute lymphoblastic leukemia, acute myeloid leukemia and acute myeloblastic leukemia. The leukemia can be relapsed, refractory, or resistant to conventional therapy.
The term "relapsed" refers to a situation where a subject or a mammal, who has had a remission of cancer after therapy, has a return of cancer cells.
The term "refractory or resistant" refers to a circumstance where a subject or a mammal, even after intensive treatment, has residual cancer cells in his body.
The term "drug resistance" refers to the condition when a disease does not respond to the treatment of a drug or drugs. Drug resistance can be either intrinsic, which means the disease has never been responsive to the drug or drugs, or it can be acquired, which means the disease ceases responding to a drug or drugs that the disease had previously responded to. In certain embodiments, drug resistance is intrinsic. In certain embodiments, the drug resistance is acquired.
As used herein, the term "EC50" refers to a dosage, concentration or amount of a particular test compound that elicits a dose-dependent response at 50% of maximal expression of a particular response that is induced, provoked or potentiated by the particular test compound.
As used herein, the term "Emax" refers to an amount, concentration or dosage of a particular test compound that achieves a 100% inhibition of a maximal response in an assay that measures such response.
As used herein, the term "Eo" refers to an amount, concentration or dosage of a particular test compound that achieves a 0% inhibition of a maximal response in an assay that measures such response. As used herein, the terms "subject" and "patient" are used interchangeably herein. The terms "subject" and "subjects" refer to an animal, such as a mammal including a non- primate (e.g., a cow, pig, horse, cat, dog, rat, and mouse) and a primate (e.g., a monkey such as a cynomolgous monkey, a chimpanzee and a human), and for example, a human. In another embodiment, the subject is a farm animal (e.g., a horse, a cow, a pig, etc.) or a pet (e.g., a dog or a cat). In one embodiment, the subject is a human.
As used herein, the terms "drug," "therapeutic agent," and "chemotherapeutic agent" refer to any agent(s), compound, or pharmaceutical composition thereof, which can be used in the treatment or prevention of a disorder or one or more symptoms thereof. In certain embodiments, the term "therapeutic agent" includes a compound provided herein. In one embodiment, a therapeutic agent is an agent which is known to be useful for, or has been or is currently being used for the treatment or prevention of a disorder or one or more symptoms thereof.
"Therapeutically effective amount" refers to an amount of a compound or composition that, when administered to a subject for treating a disease, is sufficient to effect such treatment for the disease. A "therapeutically effective amount" can vary depending on, inter alia, the compound, the disease and its severity, and the age, weight, etc., of the subject to be treated.
"Treating" or "treatment" of any disease or disorder refers, in one embodiment, to ameliorating a disease or disorder that exists in a subject. In another embodiment, "treating" or "treatment" includes ameliorating at least one physical parameter, which may be indiscernible by the subject. In yet another embodiment, "treating" or "treatment" includes modulating the disease or disorder, either physically (e.g., stabilization of a discernible symptom) or physiologically (e.g., stabilization of a physical parameter) or both. In yet another embodiment, "treating" or "treatment" includes delaying the onset of the disease or disorder.
As used herein, the terms "prophylactic agent" and "prophylactic agents" as used refer to any agent(s) which can be used in the prevention of a disorder or one or more symptoms thereof. In certain embodiments, the term "prophylactic agent" includes a compound provided herein. In certain other embodiments, the term "prophylactic agent" does not refer to a compound provided herein. For example, a prophylactic agent is an agent which is known to be useful for, or has been or is currently being used to prevent or impede the onset, development, progression and/or severity of a disorder. As used herein, the phrase "prophylactically effective amount" refers to the amount of a therapy (e.g., prophylactic agent) which is sufficient to result in the prevention or reduction of the development, recurrence or onset of one or more symptoms associated with a disorder or to enhance or improve the prophylactic effect(s) of another therapy (e.g., another prophylactic agent). Compounds
Prodrug compounds of a variety of therapeutic agents can be formed using methods available in the art and those disclosed herein. The therapeutic agent can be derivatized to include a reactive group for attachment of the phosphate moiety. Such therapeutic agent includes but is not limited to nucleosides and nucleoside analogues including acyclic nucleosides.
As used herein, a "phosphoramidate or cyclic phosphate compound of a therapeutic agent" includes a therapeutic agent derivatized to include a phosphoramidate or cyclic phosphate group. The therapeutic agent is, for example, an anti-cancer agent that includes, or has been derivatized to include, a reactive group, such as a hydroxyl, for attachment of the phosphoroamidate and cyclic phosphate moiety. Such therapeutic agents include, but are not limited to nucleosides and nucleoside analogs including acyclic nucleosides. A nucleoside analog refers to a structurally modified nucleoside. In one embodiment, the nucleosides or nucleoside analogs are derivitized at the 5'- and/or the 3 '-position by removal of a hydrogen from an hydroxyl group to include a phosphate moiety comprising a phosphoramidate and a cyclic phosphate group. In another embodiment, the nucleosides or nucleoside analogs are derivitized at the 5 '- and/or the 2'-position by removal of a hydrogen from an hydroxyl group to include a phosphate moiety comprising a phosphoramidate and a cyclic phosphate group. Modified phosphate derivatives comprising phosphoramidate and cyclic phosphate derivatives of nucleoside analogues comprising natural and non-natural nucleosides described herein can be formed as described herein and used for the treatment of cancer. In one embodiment, the derivative moiety can be at the 3 ' position. In another embodiment, the derivative moiety can be at the 5' position. In another embodiment, the derivative moiety can be at the 3', 5' positions linked together to form a cyclic derivative. In one embodiment, the prodrug moiety can be at the 2', 5' positions linked together to form a cyclic derivative.
In one embodiment, provided herein is a compound of Formula I:
X ; ;
R60 H
(I)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein
each of X and Y is independently hydrogen, -ORa, -NRaR , an N-linked or O- linked amino acid residue, or an N-linked or O-linked residue of an amino acid derivative;
where Ra and R at each occurrence is independently hydrogen, alkyl, aryl, arylalkyl, or heteroarylalkyl;
R is a nucleobase;
R1 is hydrogen, halo, cyano, alkynyl or— OR6; and
R6 is hydrogen, alkyl, aryl, alkylcarbonyl, arylcarbonyl, alkoxycarbonyl, aroxycarbonyl, alkylaminocarbonyl, arylaminocarbonyl, or alkylarylaminocarbonyl.
In one embodiment R1 is hydrogen, halo or—OR6.
In one embodiment, when one of X and Y is hydroxyl, the other is other than hydroxyl. In one embodiment, provided herein is a compound of Formula I:
Figure imgf000023_0001
(I)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein
Figure imgf000023_0002
p-N02PhCH20- — NHCH2Ph, -N(CH3)CH2Ph, — NF£R , -NH(CH2)5 — BH3,
Figure imgf000023_0003
Figure imgf000024_0001
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid; W is NH2, halo, OMe, or OH; T is NH2, F, CI or hydrogen;
R1 is hydrogen, halo, OH, protected OH, cyano or alkynyl; Q is OR3; E is CR4R5;
L is H, p-Me, p-OMe, p-Cl, or 3,4-Cl; n is 1 ; m is 1 or 2; p is absent or O; Ry is alkyl, alkenyl, alkynyl, alkoxycarbonyl, or hydroxyalkyl, each independently optionally substituted;
R2 is hydrogen or alkyl;
R3 is hydrogen, alkyl, alkenyl, alkynyl, alkylcarbonyl or aralkyl;
R4 and R5 are selected from: i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl, or alkylheteroaryl, wherein the alkyl is optionally substituted by alkoxy; or ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring; R6 with one of X and Y, together with the atoms to which they are attached, combine to form a six-membered heterocyclic ring where R6 and the one of X and Y together represent a single divalent -0-; or
R6 with one of X and Y, together with the atoms to which they are attached, combine to form a 7-12-membered heterocyclic ring;
wherein, when R is
Figure imgf000026_0001
, then at least one
Ry is other than alkyl.
In one embodiment, provided herein is a compound of Formula I:
X i -
R60 H
(I)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, polymorphic form thereof, wherein
Figure imgf000026_0002
p-N02PhCH20- — NHCH2Ph, -N(C¾)CH2Ph, — NHR , -NH(CH2)5N(CH3)2, — BH3,
Figure imgf000027_0001

Figure imgf000028_0001
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
W is NH2, CI, OMe or OH;
T is NH2, F, CI or hydrogen;
R1 is hydrogen, OH, F, cyano or alkynyl;
Q is OR3;
E is independently CR4R5;
L is H, p-Me, p-OMe, p-Cl, or 3,4-Cl; n is 1 ; m is 1 or 2; p is absent or O
Ry is alkyl, alkenyl, alkynyl, alkoxycarbonyl, or hydroxyalkyl, each independently optionally substituted; hydrogen or alkyl;
R3 is hydrogen, alkyl, alkenyl, alkynyl or aralkyl;
R4 and R5 are selected from: i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl or alkylheteroaryl, wherein alkyl is optionally substituted by alkoxy; or
ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring;
wherein, when R
Figure imgf000029_0001
Ry is other than alkyl. embodiment of each one of the above embodiments,
Figure imgf000029_0002
R
Figure imgf000030_0001
is not,
T is not hydrogen; W is not OH;
R1 is not cyano or alkynyl; and
R3 is not aralkyl.
The variables X, Y, Z, E, L, Q, W, T, n, m, p, R, Ry, Rz, R1, R2, R3, R4, R5, and R6 as provided herein are defined as follows. All combinations of such embodiments are within the scope of this disclosure.
In one embodiment, Ry is alkyl, alkenyl, or alkynyl. In one embodiment, Ry is alkyl.
In one embodiment, Ry is alkenyl. In one embodiment, Ry is alkynyl. In one embodiment, Ry is hydroxyalkyl. In one embodiment, Ry is 2-hydroxylmethylprop-2- yi.
3 * 3 * ·
In one embodiment, Q is OR , wherein R is as defined herein. In one embodiment Q is OCH3, OCH2CH3, OCH(CH3)2 or OCH2phenyl. In one embodiment Q is OCH3, OCH2CH3, or OCH(C¾)2.
In one embodiment, Rz is hydrogen.
In one embodiment, R3 is C¾, CH2CH3, CH(C¾)2 or CH2phenyl.
In one embodiment, E is CR4R5, wherein R4 and R5 are each as defined herein. In one embodiment, R4 or R5 is hydrogen. In one embodiment, R4 or R5 is alkyl or cycloalkyl. In one embodiment, R4 or R5 is lower alkyl. In one embodiment, R4 or R5 is methyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, R4 or R5 is C3-C5 cycloalkyl. In one embodiment, R4 or R5 is cyclopentyl. In one embodiment, one of R4 and R5 is hydrogen and the other is alkyl or cycloalkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is lower alkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is methyl. In one embodiment, one of R4 and R5 is hydrogen and the other is isobutyl. In one embodiment, one of R4 and R5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is cyclopentyl.
In one embodiment, Z is an oleic acid chain.
In one embodiment, W is NH2 and T is CI or F. In one embodiment, W is CI or OMe and T is NH2. In one embodiment, W is OMe and T is NH2. In one embodiment, W is CI and T is NH2.
In one embodiment, R1 is hydroxyl or fluoro. In one embodiment, R1 is hydroxyl. In one embodiment, R1 is fluoro.
In one embodiment, R2 is hydrogen or alkyl. In one embodiment, R2 is lower alkyl. In one embodiment, R 2 is methyl. In one embodiment, R 2 is hydrogen.
In one embodiment, R6 is hydrogen.
Figure imgf000031_0001
In one embodiment, R is
Figure imgf000032_0001
In certain embodiments, the compound of formula (I) is selected such that when R is
Figure imgf000032_0002
? , then Ry is other than alkyl.
In certain embodiments, the compound of formula (I) is selected such that when X and Y are both RV S^^^ s ^ t|ien j^y js Q^er faan aikyi
In certain embodiments, the compound of formula (I) is selected such that when R is
Figure imgf000032_0003
one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
Figure imgf000032_0004
embodiment, when X is OH, Y is o 1 In one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
Figure imgf000033_0001
In one embodiment, when X is OH, Y is p-N02PhCH20— . In one embodiment, when X is OH, Y is— NHCH2Ph.
In one embodiment, when X is OH, Y is— N(CH3)CH2Ph. In one embodiment, when X is OH, Y is— NHR2.
In one embodiment, when X is OH, Y is-NH(CH2)5N(CH3)2. In one embodiment, when X is OH, Y is— BH3.
In one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
In one embodiment, when X is OH, Y is
Figure imgf000033_0002
In one embodiment, when X is OH, Y is
Figure imgf000034_0001
HN
In one embodiment, when X is OH, Y is OR
In one embodiment, when X is OH, Y is
Figure imgf000034_0002
HN
In one embodiment, when X is OH, Y is OR3
HN >
(CH2)3 ,0
In one embodiment, when X is OH, Y is H2N OH
HN
(CH2)3
In one embodiment, when X is OH, Y is H2N 0 H
HN
(CH2)3
I n one emb .od .iment, wh hen X is OH, Y is H2N ° 0H
N-|
In one embodiment, when X is o > γ 1S Ry^S^ In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000035_0001
In one embodiment, when X is O , Y is °2N
Figure imgf000035_0002
one embodiment, when X is , Y is -N02PhCH20— .
In one embodiment, when X is , Y is— NHCH2Ph.
In one embodiment, when X is
Figure imgf000035_0003
J is— N(CH3)CH2Ph.
In one embodiment, when X
Figure imgf000035_0004
. In one embodiment, when X is )5N(CH3)2.
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is .
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is In one embodiment, when X is
In one embodiment, when X is
Figure imgf000037_0001
.
R2 HN
- (CH2)3 embodiment, when X is % O , Y is H2N ° 0H
embodiment, when X is
Figure imgf000037_0002
.
,0.
In one embodiment, when X is RV ss . Y is I
In one embodiment, when X is
Figure imgf000037_0003
In one embodiment, when X is B Ky^ b ° ^ , Y i -s Ϊ 0 I
Figure imgf000037_0004
In one embodiment, when X is RV s , Y is °2N ^
T
-o
In one embodiment, when X is R s , Y is
In one embodiment, when X is Ry
Figure imgf000037_0005
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000038_0001
, Y is p-N02PhCH20-.
In one embodiment, when X is h
In one embodiment, when X is H2Ph
In one embodiment, when X is
In one embodiment, when X is 5N(CH3)
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is In one embodiment, when X is
Figure imgf000038_0002
In one embodiment, when X is
Figure imgf000039_0001
, Y is
HN
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is embodiment, when X is
In one embodiment, when X is
Figure imgf000039_0002
In one embodiment, when X
In one embodiment, when X
In one embodiment, when X
In one embodiment, when X In one embodiment, when X
In one embodiment, when X
In one embodiment, when X
In one embodiment, when X
Figure imgf000040_0001
-°y
In one embodiment, when X is Y is -N(CH3)CH2Ph. In one embodiment, when X is -°y
Y is— NHR2.
In one embodiment, when X is -°y
Y is -NH(CH2)5N(CH3)2
,0.
In one embodiment, when X is y Y is— BH3.
In one embodiment, when X is
Figure imgf000040_0002
Y is In one embodiment, when X
In one embodiment, when X
Figure imgf000041_0001
I
HN HN
In one embodiment, when X is
Figure imgf000041_0002
, Y is
HN T
In one embodiment, when X In one embodiment, when X
In one embodiment, when X
Figure imgf000041_0003
HN
(CH2)3
In one embodiment, when X is
Figure imgf000041_0004
, Y is H 2 ° OH HN
(CH2)3
In one embodiment, when X is \ I , YY I iSs H22NN C 0 .
In one embodiment, when X is
Figure imgf000042_0001
o o
In one embodiment when X is m , Y is m . embodiment, when X is In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000042_0002
Figure imgf000042_0003
o
In one embodiment, when X is c *° my f , Y is— NHCH2Ph.
o
In one embodiment, when X is cA*° mY ' , Y is— N(CH3)CH2Ph. O
embodiment, when X is m , Y is— NHR
In one embodiment, when X )5N(CH3)2.
In one embodiment, when X
In one embodiment, when X i
Figure imgf000043_0001
s m Ϋ , Y is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000043_0002
, Y is
HN
O
Q½°y
In one embodiment, when X is m , Y is OR3 In one embodiment, when X
embodiment, when X
In one embodiment, when X
Figure imgf000044_0001
HN
(CH2)3
O / o embodiment, when X is m , Y is H2N 0 H
In one embodiment, when X is
Figure imgf000044_0002
, Y is .
Y
In one embodiment, when X is o 1 , Y is o 1 y
In one embodiment, when X is O I Y is °2N x
In one embodiment, when X is o ' , Y is
Figure imgf000044_0003
f Λ-s In one embodiment, when X is o 1 , Y is °2N In one embodiment, when X is 0 , Y is
Figure imgf000045_0001
Figure imgf000045_0002
In one embodiment, when X is 0 , Y is— NHCH2Ph.
In one embodiment, when X is o 1 J is— N(CH3)CH2Ph. In one embodiment, when X is
Figure imgf000045_0003
, Y is— NHR .
In one embodiment, when X is o 1 , Y is -NH(CH2)5N(CH3)2.
In one embodiment, when X is
In one embodiment, when X
Figure imgf000045_0004
Figure imgf000045_0005
In one embodiment, when X is
Figure imgf000045_0006
In one embodiment, when X is
Figure imgf000046_0001
HN r embodiment, when X is V oV 1V , Y is OR3
T HN
QY°Y°
embodiment, when X is o 1 , Y is OR3
In one embodiment, when X is QY o°r 1°- , Y is OR3
HN
(CH2)3 In one embodiment, when X is
Figure imgf000046_0002
, Y is H2N W 0H
> HN
(CH2)3
In one embodiment, when X is Y o Y 1V , Y is H2N ° 0H
Figure imgf000046_0003
In one embodiment, when X is
Figure imgf000046_0004
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000047_0001
w J
ne embodiment, when X is °2N H u 0
In o , Y is V 0
In one embodiment, when X is H20— .
In one embodiment, when X is Ph.
In one embodiment, when X is H2Ph.
embodiment, when X is
In one embodiment, when X is )5N(CH3)2.
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000047_0002
, Y is In one embodiment, when X is
In one embodiment, when X is
Figure imgf000048_0001
, Y is
Figure imgf000048_0002
embodiment, when X is 2
Figure imgf000048_0003
In one embodiment, when X is °2N , Y is
Figure imgf000048_0004
In one embodiment, when X is °2N ^ , Y is OR3
In one embodiment, when X is
Figure imgf000048_0005
H
Figure imgf000049_0001
HN
^ „ (CH2)3
N λ
In one embodiment, when X is °2N H 0 , Y is H2N 0H .
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000049_0002
°2 , Y is p-N02PhCH20— .
In one embodiment, when X is Y is— NHCH2Ph.
In one embodiment, when X is Y is -N(CH3)CH2Ph.
embodiment, when X is Y is— NHR
In one embodiment, when X is
Figure imgf000049_0003
°2 , Y is -NH(CH2)5N(CH3)2. In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000050_0001
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
embodiment, when X is
Figure imgf000051_0001
°2 , Y is p-N02PhCH20-
In one embodiment, when X is °2 , Y is— NHCH2Ph.
embodiment, when X is °2 , Y is -N(CH3)CH2Ph.
In one embodiment, when X is °2 Y is— NHR
In one embodiment, when X is °2
Figure imgf000051_0002
, Y is -NH(CH2)5N(CH3)2. In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000052_0001
In one embodiment, when X is °2N .
In one embodiment, when X is °2N
Figure imgf000053_0001
.
HN
^ , (CH2)3
In one embodiment, when X is °2N , Y is H2N 0 H .
In one embodiment, when X is
Figure imgf000053_0002
o o
In one embodiment, when X is ^ /\ , Y is p-N02PhCH20- embodiment, when X is
Figure imgf000053_0003
, Y is— NHCH2Ph.
o o
In one embodiment, when X is x , Y is -N(CH3)CH2Ph.
o o
In one embodiment, when X is / \ , Y is— NHR .
o o
In one embodiment, when X is \ , Y is -NH(CH2)5N(CH3)2.
o o
In one embodiment, when X is / \ " r , Y is— B¾ In one embodiment, when X
In one embodiment, when X
In one embodiment, when X
In one embodiment, when X
In one embodiment, when X
embodiment, when X
In one embodiment, when X
Figure imgf000054_0001
HN
(CH2)3
0 0 ( O
In one embodiment, when X I iSs / \X ' , , YY I iSs HH22NN 00hH . embodiment, when X is
In one embodiment, when X iiss
Figure imgf000055_0001
.
In one embodiment, when X is p-N02PhCH20— Y is p-N02PhCH20— . In one embodiment, when X is p-N02PhCH20— Y is— NHCH2Ph.
In one embodiment, when X is p-N02PhCH20— , Y is— N(CH3)CH2Ph. In one embodiment, when X is p- N02PhCH20- Y is— NHR2.
In one embodiment, when X is p-N02PhCH20— , Y is— NH(CH2)5N(CH3)2. In one embodiment, when X is p-N02PhCH20— , Y is— BH3.
In one embodiment, when X is p-N02PhCH20— , Y is
In one embodiment, when X is p-N02PhCH20— , Y is
In one embodiment, when X is p-N02PhCH20— , Y is
Figure imgf000055_0002
In one embodiment, when X is p-N02PhCH20— , Y is
In one embodiment, when X is p-N02PhCH20— , Y is
In one embodiment, when X is p-NChPhCFbO— , Y is
Figure imgf000056_0001
HN
In one embodiment, when X is p-NChPhCFbO— , Y is OR3 .
HN >
(CH2)3
In one embodiment, when X is p-N02PhCH20— , Y is H2N ° 0H .
In one embodiment, when X is p-N02PhCH20— Y is .
In one embodiment, when X is p-N02PhCH20— Y is
Figure imgf000056_0002
. In one embodiment, when X is— NHCH2Ph, Y is— NHCH2Ph. In one embodiment, when X is— NHCH2Ph, Y is -N(CH3)CH2Ph. In one embodiment, when X is— NHCH2Ph, Y is— NHR2. In one embodiment, when X is— NHCH2Ph, Y is— NH(CH2)5N(CH3)2. In one embodiment, when X is— NHCH2Ph, Y is— BH3.
In one embodiment, when X is— NHCH2Ph, Y is
In one embodiment, when X is— NHCH2Ph, Y is
In one embodiment, when X is— NHCH2Ph, Y is
embodiment, when X is— NHCH Ph, Y is
Figure imgf000057_0001
HN
In one embodiment, when X is— NHCH2Ph, Y is OR
HN
In one embodiment, when X is— NHCH Ph, Y is OR3
HN
In one embodiment, when X is— NHCH2Ph, Y is OR3 . HN
(CH2)3
.0
In one embodiment, when X is— NHCH2Ph, Y is H2N OH .
In one embodiment, when X is— NHCH2Ph, Y is
Figure imgf000058_0001
.
HN
(CH2)3
In one embodiment, when X is— NHCH2Ph, Y is H2N 0 H .
In one embodiment, when X is— NHCH2Ph, Y is selected from
Figure imgf000058_0002
Figure imgf000058_0003
In one embodiment, when X is— NHCH2Ph, Y is
Figure imgf000058_0004
and Ry ig alky l In one embodiment, Ry is hydroxyalkyl. In one embodiment, Ry is 2-hydroxylmethylprop-2-yl.
O O
In one embodiment, when X is— NHCH2Ph, Y is Q /\ ° ' , and Q is OR3. In one embodiment, R3 is alkyl. In one embodiment, Q is OCH3, OCH2CH3, or OCH(CH3)2.
In one embodiment, when X is -N(CH3)CH2Ph, Y is -N(CH3)CH2Ph.
In one embodiment, when X is— N(CH3)CH2Ph, Y is— NHR2.
In one embodiment, when X is -N(CH3)CH2Ph, Y is -NH(CH2)5N(CH3)2. embodiment, when X is -N(CH3)CH2Ph, Y is— BH3.
embodiment, when X is— N(CH3)CH2Ph, Y is
embodiment, when X is— N(CH3)CH2Ph, Y is
In one embodiment, when X is— N(CH3)CH2Ph, Y is
embodiment, when X is -N(CH3)CH2Ph, Y "
Figure imgf000059_0001
In one embodiment, when X is— N(CH3)CH2Ph, Y is OR3
HN
In one embodiment, when X is— N(CH3)CH2Ph, Y is OR3
HN
In one embodiment, when X is— N(CH3)CH2Ph, Y is OR3 . In one embodiment, when X is -N(CH3)CH2Ph, Y is
Figure imgf000060_0001
.
HN
(CH2)3 ί O
In one embodiment, when X is -N(CH3)CH2Ph, Y is H2N 0 H .
HN
(CH2)3
In one embodiment, when X is -N(CH3)CH2Ph, Y is H2N 0 H . In one embodiment, when X is— NHR2, Y is— NHR2.
In one embodiment, when X is—NHR2, Y is -NH(CH2)5N(CH3)2. In one embodiment, when X is—NHR2, Y is— BH .
In one embodiment, when X is—NHR2, Y is
In one embodiment, when X is—NHR2, Y is
In one embodiment, when X is— NHR , Y is
Figure imgf000060_0002
In one embodiment, when X is— NHR2, Y is
Figure imgf000061_0001
HN
In one embodiment, when X is— NHR , Y is OR3.
T HN
In one embodiment, when X is— NHR , Y is OR3.
HN
In one embodiment, when X is—NHR", Y is OR3 .
HN
(CH2)3
.0
In one embodiment, when X is—NHR2, Y is H2N 0H .
HN
(CH2)3
- P
"r
In one embodiment, when X is—NHR", Y is H2N 0H .
Figure imgf000061_0002
In one embodiment, when X is— NH(CH2)5N(CH3)2, Y is— NH(CH2)5N(CH3)2. In one embodiment, when X is— NH(CH2)5N(CH3)2, Y is— BH3. embodiment, when X is -NH(CH2)5N(CH3)2, Y is
In one embodiment, when X is— NH(CH2)5N(CH3)2, Y is
In one embodiment, when X is— NH(CH2)5N(CH3)2, Y is
embodiment, when X IS -NH(CH2)5N(CH3)2, Y IS
Figure imgf000062_0001
HN
In one embodiment, when X is— NH(CH2)5N(CH3)2, Y is OR3 .
HN
In one embodiment, when X is -NH(CH2)5N(CH3)2, Y is OR3
HN
In one embodiment, when X is— NH(CH2)5N(CH3)2, Y is OR3.
In one embodiment, when X is -NH(CH2)5N(CH3)2, Y is
Figure imgf000062_0002
. In one embodiment, when X is -NH(CH2)5N(CH3)2,
In one embodiment, when X is -NH(CH2)5N(CH3)2,
Figure imgf000063_0001
In one embodiment, when X is— BH3, Y is— BH3.
In one embodiment, when X is— BH3, Y is
In one embodiment, when X is— BH3, Y is
In one embodiment, when X is— BH3, Y is
In one embodiment, when X is— BH3, Y is
Figure imgf000063_0002
HN
In one embodiment, when X is— BH3, Y is OR HN
In one embodiment, when X is— B¾, Y is OR3
In one embodiment, when X is— B¾, Y is
Figure imgf000064_0001
HN
(CH2)3
In one embodiment, when X is— B¾, Y is H2N 0 H .
HN
(CH2)3 ' .0 embodiment, when X is— B¾, Y is H2N 0 H
HN
(CH2)3 .0 In one embodiment, when X is— BH3, Y is H2N c
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000064_0002
γ is In one embodiment, when X i
In one embodiment, when X i
In one embodiment, when X i
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000065_0001
embodiment, when X i
In one embodiment, when X i
In one embodiment, when X is
In one embodiment, when X is In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000066_0001
7
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000067_0001
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000068_0001
HN HN
(CH2)3
In one embodiment, when X is P , Y is H2N 0 H
In one embodiment, when X is H
In one embodiment, when X is
Figure imgf000068_0002
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000069_0001
HN HN
In one embodiment, when X is OR3 5 γ ls OR3 •y^
HN HN
In one embodiment, when X is OR3 5 γ is OR3
■y*, - ^
HN HN
In one embodiment, when X is OR3 5 γ is OR3 .
HN HN T
In one embodiment, when X is OR3 γ is OR3 .
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000070_0001
HN HN
(CH2)3 (CH2)3
In one embodiment, when X is H2N ° 0H Y is H2N ° 0 H . In one embodiment, when X is
Figure imgf000071_0001
>
HN HN
(CH2)3 (CH2)3
C o C ,0
In one embodiment, when X is H2N 0H , Y is H2N 'H 0: H .
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000071_0002
R5, R6, and all other variables are as described elsewhere herein. In one embodiment, Q is OCH3, OCH2CH3, or OCH(CH3)2.
In one embodiment, when X is
Figure imgf000071_0003
, R is hydrogen, R or R is hydrogen. In one embodiment, R4 or R5 is alkyl or cycloalkyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, R4 or R5 is C3-C5 cycloalkyl. In one embodiment, R4 or R 5 is cyclopentyl. In one embodiment, Q is OR 3 , wherein R 3 and all other variables are as described elsewhere herein.
In one embodiment, when X is
Figure imgf000071_0004
, Y is , R is hydrogen, R or R is hydrogen. In one embodiment, R4 or R5 is alkyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, Q is OR . In one embodiment, R is alkyl. In one embodiment Q, is OCH3, OCH2CH3, or OCH(CH3)2.
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000072_0001
, Y is , m is 1 , and Q is OR , wherein n, R2, R3, R4, R5, R6 and all other variables are as described elsewhere herein. In one embodiment, Q is OCH3, OCH2CH3, or OCH(C¾)2.
In one embodiment, when X is
Figure imgf000072_0002
, Y is , m is 1, R is hydrogen, R or R is hydrogen. In one embodiment, R4 or R5 is alkyl or cycloalkyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, R4 or R5 is C3-C5 cycloalkyl. In one embodiment, R4 or R5 is cyclopentyl. In one embodiment, Q is OR3, wherein R3 and all other variables are as described elsewhere herein.
In one embodiment, when X is
Figure imgf000072_0003
, Y is , m is 1, R2 is hydrogen, R r.44 o„r„ R r.5 is hydrogen. In one embodiment, R4 or R5 is alkyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, Q is OR3. In one embodiment, R3 is alkyl. In one embodiment Q, is OCH3, OCH2CH3, or OCH(C¾)2.
In one embodiment, when X is
Figure imgf000072_0004
In one embodiment, when R is
Figure imgf000073_0001
; X and Y both are 5 then one Ry is other than alkyl, and other variables are as described elsewhere herein.
In one embodiment, when R is
Figure imgf000073_0002
X and Y both are
hydroxyalkyl. In one embodiment, Ry is 2-hydroxylmethylprop-2-yl, and other variables are as described elsewhere herein.
In one embodiment, when X is
In one embodiment, when X is
Figure imgf000073_0003
, and Q is OR , wherein n, R ,
R3, R4, R5, R6 and all other variables are as described elsewhere herein. In one embodiment, Q is OC¾, OCH2CH3, or OCH(CH3)2.
In one embodiment, when X is
Figure imgf000073_0004
, Y is , R is hydrogen, R or R is hydrogen. In one embodiment, R4 or R5 is alkyl or cycloalkyl. In one embodiment, R4 or R5 is methyl. In one embodiment, R4 or R5 is C3-C5 cycloalkyl. In one embodiment, R4 or R5 is cyclopentyl. In one embodiment, Q is OR3, wherein R3 and all other variables are as described elsewhere herein. In one embodiment, when X is
Figure imgf000074_0001
, Y is O 1 , R2 is hydrogen, R4 or R5 is hydrogen. In one embodiment, R or R is alkyl. In one embodiment, R or R is methyl. IInn oonnee eemmbbooddiimmeenntt,, QQ iiss OORR33.. IInn one embodiment, R3 is alkyl. In one embodiment Q, is OCH3, OCH2CH3, or OCH(CH3)2.
In one embodiment when X is
Figure imgf000074_0002
Y IS 0,N or
Ο,
In
Figure imgf000074_0003
one embodiment, when X is Y lS In one embodiment, L is hydrogen.
In one embodiment, when X is
Figure imgf000074_0004
Y is °2N In one embodiment, L is hydrogen.
In one embodiment, when X is
Figure imgf000074_0005
Y IS °2N In one embodiment, L is hydrogen. In one embodiment, when X is
In one embodiment, when X is
Figure imgf000075_0001
In one embodiment, L is hydrogen.
In some embodiments, when the phosphoramidate is substituted with at least one amino acid group, the amino acid is in the L-configuration. In one embodiment, the amino acid is alanine. In one embodiment, the amino acid is leucine.
In some embodiments, the phosphoramidate compound provided herein possesses a chiral phosphorous center. In some embodiments, the phosphoramidate is diastereomerically enriched.
In some embodiments, provided herein are:
(a) compounds as described herein, e.g., of Formula I and pharmaceutically acceptable salts and compositions thereof;
(b) compounds as described herein, e.g., of Formula I and pharmaceutically acceptable salts and compositions thereof for use in therapy;
(c) compounds as described herein, e.g., of Formula I and pharmaceutically acceptable salts and compositions thereof for use in the treatment and/or prophylaxis of cancer including leukemia;
(d) processes for the preparation of compounds as described herein, e.g., of Formula I as described in more detail elsewhere herein;
(e) pharmaceutical compositions comprising a compound as described herein, e.g., of Formula I or a pharmaceutically acceptable salt thereof together with a pharmaceutically acceptable carrier, diluents or excipient;
(f) a method for the treatment and/or prophylaxis of cancer, in one embodiment, leukemia, comprising the administration of an effective amount of a compound as described herein, e.g., of Formulas I its pharmaceutically acceptable salt or composition.
(g) combinations comprising an effective amount of a compound as described herein, e.g., of Formulas I or its pharmaceutically acceptable salt, and one, two, three or more other therapeutic agents, e.g. anti-cancer agents.
In one embodiment, provided herein is a compound of Formula Ila:
Figure imgf000076_0001
(Ila)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein
Figure imgf000076_0002
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
W is NH2, halo, or OMe;
T is NH2, F, or CI;
R1 is hydrogen, halo, OH, or protected OH;
Q is OR3;
E is CR4R5;
n is 1 ;
R7 is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
R is hydrogen, alkyl, alkenyl, alkynyl or aralkyl; R4 and R5 are selected from:
i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl or alkylheteroaryl, wherein the alkyl is optionally substituted by alkoxy; or
ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
In one embodiment, R3 is not aralkyl.
In one embodiment, provided herein is a compound of Formula Ila:
Figure imgf000077_0001
(Ila)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein
Figure imgf000077_0002
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
W is NH2 or CI;
T is NH2, F or CI;
R1 is hydrogen, OH or F;
Q is OR3;
E is CR4R5;
n is 1 ; R7 is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
R3 is hydrogen, alkyl, alkenyl, alkynyl or aralkyl;
R4 and R5 are selected as follows:
i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl or alkylheteroaryl, wherein alkyl is optionally substituted by alkoxy; or
ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
In one embodiment R 3 is not aralkyl.
The variables X, Y, Z, E, Q, W, T, n, R, Ry, Rz, R1, R2, R3, R4, R5, and R7 as provided herein are defined as herein.
In one embodiment, E is CR4R5, wherein R4 and R5 are each as defined herein. In one embodiment, R4 or R5 is hydrogen. In one embodiment, R4 or R5 is alkyl, or cycloalkyl. In one embodiment, R4 or R5 is lower alkyl. In one embodiment, R4 or R5 is methyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, R4 or R5 is C3-C5 cycloalkyl. In one embodiment, R4 or R5 is cyclopentyl. In one embodiment, one of R4 and R5 is hydrogen and the other is alkyl or cycloalkyl. In one embodiment, one of R and R' is hydrogen and the other is lower alkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is methyl. In one embodiment, one of R4 and R5 is hydrogen and the other is isobutyl. In one embodiment, one of R4 and R5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is cyclopentyl.
In one embodiment, Z is an oleic acid chain.
In one embodiment, W is NH2, T is CI or F. In one embodiment, W is CI, T is NH2. In one embodiment, W is OMe, T is NH2.
In one embodiment, R1 is hydrogen. In one embodiment, R1 is hydroxy or halo. In one embodiment R1 is fluoro. In one embodiment, R2 is hydrogen or alkyl. In one embodiment, R2 is lower alkyl. In one embodiment, R2 is methyl. In one embodiment, R2 is hydrogen.
In one embodiment, R3 is C¾, CH2CH3, CH(CH3)2 or CH2phenyl.
Figure imgf000079_0001
In one embodiment, Y is . In one embodiment Y is OR , wherein R is as defined herein. In one embodiment Y is OCH3, OCH2CH3, or OCH(CI¾)2. In one embodiment Q is OCH(CH3)2. In one embodiment Q is OCH2CH3 or OCH2phenyl.
In some embodiments, when the phosphoramidate is substituted with an amino acid group, the amino acid is in the L-configuration. In one embodiment, the amino acid is alanine. In one embodiment, the amino acid is leucine.
In some embodiments, the phosphoramidate compound provided herein possesses a chiral phosphorous center. In some embodiments, the phosphoramidate is diastereomerically enriched.
In one embodiment, provided herein is a compound of Formula lib:
Figure imgf000079_0002
(lib)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein
Y is or OR7;
R is
Figure imgf000079_0003
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
Q is OR3;
E is CR4R5;
n is 1 ;
R is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
R3 is hydrogen, alkyl, alkenyl, alkynyl or aralkyl;
R4 and R5 are selected from:
i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl, or alkylheteroaryl, wherein alkyl is optionally substituted by alkoxy; or
ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
In one embodiment R3 is not aralkyl.
The variables X, Y, Z, E, Q, W, T, n, R, Ry, Rz, R1, R2, R3, R4, R5, and R7 as provided herein are defined as herein.
In one embodiment, E is CR4R5, wherein R4 and R5 are each as defined herein. In one embodiment, R4 or R5 is hydrogen. In one embodiment, R4 or R5 is alkyl, or cycloalkyl. In one embodiment, R4 or R5 is lower alkyl. In one embodiment, R4 or R5 is methyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, R4 or R5 is C3-C5 cycloalkyl. In one embodiment, R4 or R5 is cyclopentyl. In one embodiment, one of R4 and R5 is hydrogen and the other is alkyl or cycloalkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is lower alkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is methyl. In one embodiment, one of R4 and R5 is hydrogen and the other is isobutyl. In one embodiment, one of R4 and R5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is cyclopentyl.
In one embodiment, Z is an oleic acid chain. In one embodiment, R is hydrogen or alkyl. In one embodiment, R is lower alkyl. In one embodiment, R2 is methyl. In one embodiment, R2 is hydrogen.
In one embodiment, R3 is C¾, CH2CH3, CH(CH3)2 or CH2phenyl.
In one embodiment, Y is
Figure imgf000081_0001
. In one embodiment Y is OR , wherein R is as defined herein. In one embodiment, Y is OCH3, OCH2CH3, or OCH(CH3)2. In one embodiment Q is OCH(CH3)2. In one embodiment Q is OCH2CH3 or OCH2phenyl.
In one embodiment, provided herein is a compound of Formula lie:
Figure imgf000081_0002
(lie)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, polymorphic form thereof, wherein
Figure imgf000081_0003
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
Q is OR3;
E is CR4R5; n is 1 ;
R7 is alkyl, alkenyl, or alkynyl, each of which is optionally substituted;
R is hydrogen, alkyl, alkenyl, alkynyl or aralkyl;
R4 and R5 are selected from:
i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl, or alkylheteroaryl, wherein alkyl is optionally substituted by alkoxy; or
ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring.
In one embodiment R3 is not aralkyl.
The variables X, Y, Z, E, Q, W, T, n, R, Ry, Rz, R1, R2, R3, R4, R5, and R7 as provided herein are defined as herein.
In one embodiment, E is CR4R5, wherein R4 and R5 are each as defined herein. In one embodiment, R4 or R5 is hydrogen. In one embodiment, R4 or R5 is alkyl, or cycloalkyl. In one embodiment, R4 or R5 is lower alkyl. In one embodiment, R4 or R5 is methyl. In one embodiment, R4 or R5 is isobutyl. In one embodiment, R4 or R5 is C3-C5 cycloalkyl. In one embodiment, R4 or R5 is cyclopentyl. In one embodiment, one of R4 and R5 is hydrogen and the other is alkyl or cycloalkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is lower alkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is methyl. In one embodiment, one of R4 and R5 is hydrogen and the other is isobutyl. In one embodiment, one of R4 and R5 is hydrogen and the other is C3-C5 cycloalkyl. In one embodiment, one of R4 and R5 is hydrogen and the other is cyclopentyl.
In one embodiment, Z is an oleic acid chain.
In one embodiment, R2 is hydrogen or alkyl. In one embodiment, R2 is lower alkyl. In one embodiment, R2 is methyl. In one embodiment, R2 is hydrogen.
In one embodiment, R3 is C¾, CH2CH3, CH(CH3)2 or CH2phenyl In one embodiment, Y is
Figure imgf000083_0001
. In one embodi ·ment Y i -s OR7 , wherein R 7 is as defined herein. In one embodiment, Y is OCH3, OCH2CH3, or OCH(CI¾)2. In one embodiment Q is OCH(CH3)2. In one embodiment, Q is OCH2CH3 or OCH2phenyl.
In some embodiments, when the phosphoramidate is substituted with an amino acid group, the amino acid is in the L-configuration. In one embodiment, the amino acid is alanine. In one embodiment, the amino acid is leucine.
In some embodiments, the phosphoramidate compound provided herein possesses a chiral phosphorous center. In some embodiments, the phosphoramidate is diastereomerically enriched.
In one embodiment, provided herein is a compound of Formula lid:
Figure imgf000083_0002
(lid)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein R and Rxare as defined herein.
In one embodiment, provided herein is a compound of Formula He:
Figure imgf000083_0003
(He) or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein R, R1, R2, R6, E, n, and Q are as defined herein.
In one embodiment, R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl or cycloalkyl; or R4 is alkyl or cycloalkyl, and R5 is hydrogen.
In one embodiment, provided herein is a compound of Formula Ilf:
Figure imgf000084_0001
(Ilf)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein R, R1, R2, R6, E, m, n, and Q are as defined herein. In one embodiment, R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl or cycloalkyl; or R4 is alkyl or cycloalkyl, and R5 is hydrogen.
In one embodiment, provided herein is a compound of Formula Ilg:
Figure imgf000084_0002
(Ilg)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein R, R1, and R6 are as defined herein.
In one embodiment, provided herein is a compound of Formula IHi:
Figure imgf000085_0001
(Ilh)
or a pharmaceutically acceptable salt, or solvate thereof, or a stereoisomeric, tautomeric, or polymorphic form thereof, wherein R, R1, R2, R6, E, n, and Q are as defined herein. In one embodiment, R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl, or R4 is alkyl and R5 is hydrogen.
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000085_0002
wherein E, Q, n, Rz, R2, and R6 are as described in Formula I. In another embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000086_0001
wherein Ry, Rz, and R6 are as described in Formula I.
In another embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000086_0002
Figure imgf000087_0001
wherein E, Q, R2, Rz, and R6 are as described in Formula I.
In another embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000087_0002
wherein E, Q, R , and Rz are as described in Formula II.
In another embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000088_0001
wherein Q and Rz are as described in Formula II.
In another embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000088_0002
In another embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000089_0001
wherein Q, and Rz are as described in Formula lib.
In one embodiment, the nucleosides that can be derivatized to include a phosphoramidate, e.g., at the 5' position, include:
Figure imgf000089_0002
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000090_0001
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000091_0001
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000092_0001
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000093_0001
Figure imgf000094_0001
93
Figure imgf000095_0001
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000096_0001
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000097_0001
Figure imgf000098_0001
In one embodiment, the compound provided herein is a compound of one of the following Formulas:
Figure imgf000098_0002
Figure imgf000099_0001
It is appreciated that compounds provided herein have several chiral centers and may exist in and be isolated in optically active and racemic forms. Some compounds may exhibit polymorphism. It is to be understood that any racemic, optically-active, diastereomeric, polymorphic, or stereoisomeric form, or mixtures thereof, of a compound provided herein, which possess the useful properties described herein is within the scope of the invention. It is well known in the art how to prepare optically active forms (for example, by resolution of the racemic form by recrystallization techniques, by synthesis from optically-active starting materials, by chiral synthesis, or by chromatographic separation using a chiral stationary phase).
In particular, since the Γ and 4' carbons of a nucleoside are chiral, their nonhydrogen substituents (the base and the CHOR groups, respectively) can be either cis (on the same side) or trans (on opposite sides) with respect to the sugar ring system. The four optical isomers therefore are represented by the following configurations (when orienting the sugar moiety in a horizontal plane such that the oxygen atom is in the back): cis (with both groups "up", which corresponds to the configuration of naturally occurring β-D nucleosides), cis (with both groups "down", which is a nonnaturally occurring β-L configuration), trans (with the C2' substituent "up" and the C4' substituent "down"), and trans (with the C2' substituent "down" and the C4' substituent "up"). The "D-nucleosides" are cis nucleosides in a natural configuration and the "L-nucleosides" are cis nucleosides in the non-naturally occurring configuration.
Likewise, most amino acids are chiral (designated as L or D, wherein the L enantiomer is the naturally occurring configuration) and can exist as separate enantiomers.
Examples of methods to obtain optically active materials are known in the art, and include at least the following.
i) physical separation of crystals - a technique whereby macroscopic crystals of the individual enantiomers are manually separated. This technique can be used if crystals of the separate enantiomers exist, i.e., the material is a conglomerate, and the crystals are visually distinct; simultaneous crystallization - a technique whereby the individual enantiomers are separately crystallized from a solution of the racemate, possible only if the latter is a conglomerate in the solid state;
enzymatic resolutions - a technique whereby partial or complete separation of a racemate by virtue of differing rates of reaction for the enantiomers with an enzyme;
enzymatic asymmetric synthesis - a synthetic technique whereby at least one step of the synthesis uses an enzymatic reaction to obtain an enantiomerically pure or enriched synthetic precursor of the desired enantiomer;
chemical asymmetric synthesis - a synthetic technique whereby the desired enantiomer is synthesized from an achiral precursor under conditions that produce asymmetry (i.e., chirality) in the product, which may be achieved using chiral catalysts or chiral auxiliaries; diastereomer separations - a technique whereby a racemic compound is reacted with an enantiomerically pure reagent (the chiral auxiliary) that converts the individual enantiomers to diastereomers. The resulting diastereomers are then separated by chromatography or crystallization by virtue of their now more distinct structural differences and the chiral auxiliary later removed to obtain the desired enantiomer;
first- and second-order asymmetric transformations - a technique whereby diastereomers from the racemate equilibrate to yield a preponderance in solution of the diastereomer from the desired enantiomer or where preferential crystallization of the diastereomer from the desired enantiomer perturbs the equilibrium such that eventually in principle all the material is converted to the crystalline diastereomer from the desired enantiomer. The desired enantiomer is then released from the diastereomer;
kinetic resolutions - this technique refers to the achievement of partial or complete resolution of a racemate (or of a further resolution of a partially resolved compound) by virtue of unequal reaction rates of the enantiomers with a chiral, non-racemic reagent or catalyst under kinetic conditions;
enantiospecific synthesis from non-racemic precursors - a synthetic technique whereby the desired enantiomer is obtained from non- chiral starting materials and where the stereochemical integrity is not or is only minimally compromised over the course of the synthesis; chiral liquid chromatography - a technique whereby the enantiomers of a racemate are separated in a liquid mobile phase by virtue of their differing interactions with a stationary phase. The stationary phase can be made of chiral material or the mobile phase can contain an additional chiral material to provoke the differing interactions;
chiral gas chromatography - a technique whereby the racemate is volatilized and enantiomers are separated by virtue of their differing interactions in the gaseous mobile phase with a column containing a fixed non-racemic chiral adsorbent phase;
extraction with chiral solvents - a technique whereby the enantiomers are separated by virtue of preferential dissolution of one enantiomer into a particular chiral solvent;
transport across chiral membranes - a technique whereby a racemate is placed in contact with a thin membrane barrier. The barrier typically separates two miscible fluids, one containing the racemate, and a driving force such as concentration or pressure differential causes preferential transport across the membrane barrier. Separation occurs as a result of the non-racemic chiral nature of the membrane which allows only one enantiomer of the racemate to pass through.
In some embodiments, compositions of the present disclosure are provided that are substantially free of an undesignated enantiomer of a nucleoside or nucleoside derivative. In one embodiment, in the methods and compounds of this invention, the compounds are substantially free of undesignated enantiomers. In some embodiments, the composition includes a compound that is at least 85 %, 90%, 95%, 98%, 99%, to 100% by weight of the designated compound, the remainder comprising other chemical species or enantiomers.
Isotopically Enriched Compounds
Also provided herein are isotopically enriched compounds, including but not limited to isotopically enriched nucleoside derivatives.
Isotopic enrichment (for example, deuteration) of pharmaceuticals to improve pharmacokinetics ("PK"), pharmacodynamics ("PD"), and toxicity profiles, has been demonstrated previously with some classes of drugs. See, for example, Lijinsky et. al, Food Cosmet. Toxicol, 20: 393 (1982); Lijinsky et. al, J. Nat. Cancer Inst., 69: 1127 (1982); Mangold et. al, Mutation Res. 308: 33 (1994); Gordon et. al, Drug Metab. Dispos., 15: 589 (1987); Zello et. al, Metabolism, 43: 487 (1994); Gately et. al, J. Nucl. Med., 27: 388 (1986); Wade D, Chem. Biol. Interact. 117: 191 (1999).
Isotopic enrichment of a drug can be used, for example, to (1) reduce or eliminate unwanted metabolites, (2) increase the half-life of the parent drug, (3) decrease the number of doses needed to achieve a desired effect, (4) decrease the amount of a dose necessary to achieve a desired effect, (5) increase the formation of active metabolites, if any are formed, and/or (6) decrese the production of deleterious metabolites in specific tissues and/or create a more effective drug and/or a safer drug for combination therapy, whether the combination therapy is intentional or not. Replacement of an atom for one of its isotopes often will result in a change in the reaction rate of a chemical reaction. This phenomenon is known as the Kinetic Isotope Effect ("KIE"). For example, if a C-H bond is broken during a rate-determining step in a chemical reaction (i.e. the step with the highest transition state energy), substitution of a deuterium for that hydrogen will cause a decrease in the reaction rate and the process will slow down. This phenomenon is known as the Deuterium Kinetic Isotope Effect ("DKIE"). (See, e.g, Foster et al, Adv. DrugRes., vol. 14, pp. 1 -36 (1985); Kushner et al. , Can. J. Physiol. Pharmacol, vol. 77, pp. 79-88 (1999)).
The magnitude of the DKIE can be expressed as the ratio between the rates of a given reaction in which a C-H bond is broken, and the same reaction where deuterium is substituted for hydrogen. The DKIE can range from about 1 (no isotope effect) to very large numbers, such as 50 or more, meaning that the reaction can be fifty, or more, times slower when deuterium is substituted for hydrogen. Without being limited to a particular theory, high DKIE values may be due in part to a phenomenon known as tunneling, which is a consequence of the uncertainty principle. Tunneling is ascribed to the small mass of a hydrogen atom, and occurs because transition states involving a proton can sometimes form in the absence of the required activation energy. Because deuterium has more mass than hydrogen, it statistically has a much lower probability of undergoing this phenomenon.
Tritium ("T") is a radioactive isotope of hydrogen, used in research, fusion reactors, neutron generators and radiopharmaceuticals. Tritium is a hydrogen atom that has 2 neutrons in the nucleus and has an atomic weight close to 3. It occurs naturally in the environment in very low concentrations, most commonly found as T20. Tritium decays slowly (half-life = 12.3 years) and emits a low energy beta particle that cannot penetrate the outer layer of human skin. Internal exposure is the main hazard associated with this isotope, yet it must be ingested in large amounts to pose a significant health risk. As compared with deuterium, a lesser amount of tritium must be consumed before it reaches a hazardous level. Substitution of tritium ("T") for hydrogen results in yet a stronger bond than deuterium and gives numerically larger isotope effects. Similarly, substitution of isotopes for other elements, including, but not limited to, 13C or 14C for carbon, 33S, 34S, or 36S for sulfur, 15N for nitrogen, and 170 or 180 for oxygen, may lead to a similar kinetic isotope effect.
For example, the DKIE was used to decrease the hepatotoxicity of halothane by presumably limiting the production of reactive species such as trifluoroacetyl chloride. However, this method may not be applicable to all drug classes. For example, deuterium incorporation can lead to metabolic switching. The concept of metabolic switching asserts that xenogens, when sequestered by Phase I enzymes, may bind transiently and re-bind in a variety of conformations prior to the chemical reaction (e.g., oxidation). This hypothesis is supported by the relatively vast size of binding pockets in many Phase I enzymes and the promiscuous nature of many metabolic reactions. Metabolic switching can potentially lead to different proportions of known metabolites as well as altogether new metabolites. This new metabolic profile may impart more or less toxicity.
The animal body expresses a variety of enzymes for the purpose of eliminating foreign substances, such as therapeutic agents, from its circulation system. Examples of such enzymes include the cytochrome P450 enzymes ("CYPs"), esterases, proteases, reductases, dehydrogenases, and monoamine oxidases, to react with and convert these foreign substances to more polar intermediates or metabolites for renal excretion. Some of the most common metabolic reactions of pharmaceutical compounds involve the oxidation of a carbon-hydrogen (C-H) bond to either a carbon-oxygen (C-O) or carbon-carbon (C- C) pi-bond. The resultant metabolites may be stable or unstable under physiological conditions, and can have substantially different pharmacokinetic, pharmacodynamic, and acute and long-term toxicity profiles relative to the parent compounds. For many drugs, such oxidations are rapid. These drugs therefore often require the administration of multiple or high daily doses.
Therefore, isotopic enrichment at certain positions of a compound provided herein will produce a detectable KIE that will affect the pharmacokinetic, pharmacologic, and/or toxicological profiles of a compound provided herein in comparison with a similar compound having a natural isotopic composition. Preparation of Compounds
The compounds provided herein can be, in some instances, prepared, isolated, or obtained by any method apparent to those of skill in the art. Those skilled in the art will recognize that compounds provided herein may be designed or prepared by reaction, e.g., at a hydroxy group of said anti-cancer drug, for example, via condensation or dehydration.
In other instances, compounds provided herein can be prepared according to the Exemplary Preparation Schemes provided below. In certain embodiments, compounds provided herein can be prepared according to Schemes 1-6. Reaction conditions, steps, and reactants not provided in the Exemplary Preparation Schemes would be apparent to, and known by, those skilled in the art and, thus, are within the scope of this disclosure.
Figure imgf000107_0001
Scheme 2
Figure imgf000108_0001
6
6a: R= -CH3 and -CH3 with B†= 2-chloro-6-aminopurine 6b: R= -CH2CH3 and R-, = -CH:
r
tBuMgCI
Figure imgf000108_0002
7 8
7a: X= -OH, B= B2, R= -CH3 and R,= -CH3 8a.,: X= -OH, B= cytosine, R= -CH3 and R^ -CH3 (diastereomer 1 )
7b: X= -OH, B= B2, R= -CH2CH3 and R, = -CH2CH3 8a2: X= -OH, B= cytosine, R= -CH3 and R^ -CH3 (diastereomer 2)
7c: X= -F, B= BL R= -CH3 and R,= -CH3 8b.,: X= -OH, B= cytosine, R= -CH2CH3 and R-| = -CH2CH3 (diastereomer 1 )
8b2: X= -OH, B= cytosine, R= -CH2CH3 and R, = -CH2CH3 (diastereomer 2)
8a,: X= -F, B= B^ R= -CH3 and R,= -CH3 (diastereomer 1 )
8c2: X= -F, B= B-i , R= -CH3 and R-| = -CH3 (diastereomer 2)
Figure imgf000109_0001
108
Figure imgf000110_0001
Figure imgf000111_0001
Scheme 5 Tr
Figure imgf000112_0001
5c 16
Scheme 6
Figure imgf000113_0001
20 21
Scheme 7
Figure imgf000114_0001
Scheme 8:
1/ tBuMgCI, 1.0M in THF
Figure imgf000115_0001
Scheme 9:
Figure imgf000115_0002
B6: Benzyl (2S)-2-aminopropanoate 24f: R2=
Figure imgf000115_0003
Scheme 10:
Figure imgf000115_0004
General procedure A
Step 1 : To a stirred solution of compound 22a or 22b (9.97mmol) in THF (lOmL/mmol) at -10°C was added phosphorus oxychloride (11.96mmol). The reaction mixture was stirred from -10°C to room temperature overnight and then, a solution of reagent Bl (9.97mmol) and triethylamine (49.85mmol) in acetonitrile (2.4mL/mmol) was added at 0°C. The reaction mixture was stirred for 1 hour at 0°C. Then, reagent B2 (29.91mmol) and DMAP (29.91 mmol) were added at 0°C and the reaction mixture was stirred from 0°C to room temperature for 2 hours. The reaction mixture was then diluted with ethyl acetate, washed with HC1 IN solution, water and brine. The organic layer was filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (DCM/methanol: 0 to 5%) to afford the expected compound.
Step 2: To a stirred suspension of compound 23a-c (2.40mmol) in DCM (50mL/mmol) at -80°C under nitrogen was added a solution of boron trichloride, 1.0M in DCM (12.02mmol). The reaction mixture was stirred from -80°C to -40°C for 4 hours. Then MeOH (2mL) was added at -80°C and the reaction mixture was concentrated under reduced pressure at 25°C. The crude residue was purified by RP18 chromatography (H2O/CH3CN) to afford the expected compound as solid. In some cases, the mixture of diastereoisomers was purified by chiral HPLC to afford the expected diastereoisomer.
General procedure B
Step 1 : To a stirred solution of compound 22a (1.86mmol) in THF (lOmL/mmol) at -10°C under nitrogen was added phosphorus oxychloride (2.23mmol). The reaction mixture was stirred from -10°C to room temperature overnight and then, cooled down to 0°C. Then, a solution of reagent B3 (3.72mmol) and triethylamine (9.31mmol) in acetonitrile (2.4mL/mmol) was added at 0°C. The reaction mixture was stirred at room temperature overnight. The reaction mixture was then diluted with ethyl acetate and washed with a saturated NH4CI solution. The organic layer was filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (DCM/methanol: 0 to 3%) to afford the expected compound. Step 2: according to step 2 of general procedure A.
General procedure C
Step 1 : To a solution of compound lb (2.20mmol) in THF (lOmL/mmol) at 0°C was added a solution of tert-butylmagnesium chloride, 1M in THF (4.60mmol). The reaction mixture was stirred for 1 hour at room temperature, then cooled to 0°C. To this reaction mixture was added a solution of the appropriate reagent CI (2.40mmol) in THF (lOmL/mmol). The reaction mixture was stirred for 1 hour at 0°C and at room temperature overnight. The reaction mixture was diluted with ethyl acetate and washed with a saturated NH4C1 solution, water and brine. The organic layer was dried, filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (DCM/methanol) to afford the expected intermediate as a mixture of diastereoisomers;
Step 2: To a solution of the previous intermediate (0.52mmol) in DCM (30mL/mmol) was added trifluoroacetic acid (10.27mmol). The reaction mixture was stirred at room temperature for 3 hours and purified by flash chromatography on silica gel (DCM/methanol: 0 to 20%) followed by RP18 chromatography ((H20/CH3CN) to afford the expected compound as mixture of diastereoisomers. This mixture was purified by MS- preparative HPLC or by chiral HPLC to afford the 2 expected diastereoisomers as pure solid compounds. General procedure D
Step 1 : To a stirred solution of 4-nitrophenol (29.36mmol) and phosphorus oxychloride (14.68mmol) in diethyl ether (lOmL/mmol) at -80°C under nitrogen was added triethylamine (29.36mmol). The reaction mixture was stirred at room temperature overnight. Then, DCM (lOmL/mmol) and the appropriate L-alanine ester hydrochloride (14.68mmol) were added at 0°C under nitrogen. To the reaction mixture was added dropwise at 0°C triethylamine (29.36mmol). The reaction mixture was stirred at room temperature overnight, and then, filtrated. The filtrate was concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (PE/Et20: 0 to 50%) to afford the expected intermediate 9. Step 2: To a solution of compound la (1.55mmol) in THF (7mL/mmol) was added at room temperature under nitrogen a solution of l,8-diazabicyclo[5.4.0]undec-7-ene (3.71mmol) in acetonitrile (12mL/mmol), followed by a solution of the appropriate compound 9 (1.86mmol) in DCM (12mL/mmol). The reaction mixture was stirred at room temperature overnight, and then, quenched on tampon phosphate pH=7. The reaction mixture was extracted with DCM and the organic layer was dried, filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica gel (DCM/methanol: 0 to 10%) to afford the expected compound as a mixture of diastereoisomers. This mixture was purified by MS -Preparative HPLC to afford the pure diastereoisomer.
Compounds 10 and 11
(2 diastereomers)
Ethyl r2SV2-[[r4aR-6R.7S.7aRV6-r6-amino-2-chloro-purin-9-ylV7-fluoro-2-oxo-4a.6.7.7a- tetrahydro-4H-furo[3.2-d][1.3.2]dioxaphosphinin-2-yl]amino]propanoate
Figure imgf000118_0001
Compounds 10 and 11 were synthesized from compound la according to scheme 3 and to the general procedure D.
Compound 21
[(2 3S^S.5RV5-(6-amino-2-fluoro-purin-9-ylV3^-dihydroxy-tetrahydrofuran-2-yl]methoxy- N-benzyl-phosphonamidic acid
Figure imgf000119_0001
Compound 21 was synthesized according to scheme 6 and to the following general procedure:
To a solution of 2'OCBz fludarabine (3.38mmol) and 2-(2,2-dimethyl-3-trityloxypropanoyl)- sulfanylethoxyphosphinic acid (5.08mmol) in pyridine (12mL/mmol) at 0°C was slowly added pivaloyl chloride (6.77mmol). The reaction mixture was stirred for 1 hour at 0°C and for 2 hours at room temperature. The reaction was monitored by LC/MS. The reaction mixture was quenched with a 1M solution of NH4C1 and extracted with ethyl acetate. The organic layer was dried, filtered and concentrated under reduced pressure to afford the expected intermediate 17. To a solution of compound 17 (l. lmmol) in carbon tetrachloride (20mL/mmol) and DCM (15mL/mmol) were added benzylamine (5.5mmol) and triethylamine (6.6mmol) at room temperature. The reaction mixture was stirred overnight, and then, concentrated under reduced pressure and purified by flash chromatography on silica (DCM/methanol) to afford the expected intermediate 18.
To a solution of compound 18 (0.49mmol) in DCM (30mL/mmol) was added trifluoroacetic acid (7.4mmol) under nitrogen. The reaction mixture was stirred at room temperature overnight and purified by flash chromatography on silica (DCM/methanol: 0 to 20%) followed by MS- preparative HPLC to afford the mixture of diastereoisomers 19.
To a solution of compound 19 in EtOH (5mL/mmol) was added Palladium Black under nitrogen. After several flush N2/vaccum and vaccum/H2, the reaction mixture was stirred under H2 Atmosphere for 6hours at room temperature. The reaction mixture was filtered on Celite and concentrated under reduced pressure to afford the expected intermediate 20.
The compound 20 (0.35mmol) was dissolved in a 7N ammonia solution in methanol (50mL/mmol) and the reaction mixture was stirred at room temperature for 4 hours. The reaction mixture was concentrated under reduced pressure and purified by RP-18 chromatography (H20+TEAB/CH3CN) to afford the expected compound as solid. Compound 24a
(2 Diastereomers)
4-amino-l -[(2R,3S,4S,5R)-5-[[(benzylamino)-[(3-methyl-2-nitro-imidazol-4- yl)methoxy]phosphoryl]oxymethyl]-3.4-dihydroxy-tetrahy
Figure imgf000120_0001
Compound 24a was synthesized from compound 22a with reagent Bl and B2 according to scheme 7 and general procedure A.
Compound 24a: (Diastereoisomer 1): XH NMR (OMSO-d6, 400MHz) δ (ppm) 7.54 (d, J=7.51Hz, IH), 7.32-7.27 (m, 4H), 7.26-7.21 (m, IH), 7.20 (s, IH), 7.1 1 (brs, IH), 7.01 (brs, IH), 6.09 (d, J=3.69Hz, IH), 5.80 (dt, J=12.73Hz and 7.20Hz, IH), 5.64 (d, J=7.51Hz, IH), 5.59-5.53 (m, 2H), 5.05 (dd, J=13.26Hz and 7.79Hz, IH), 5.00 (dd, J=13.26Hz and 7.34Hz, IH), 4.14-3.88 (m, 7H), 3.86 (s, 3H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 10.27 (s, IP); MS (ESI) m/z = 550.1 (MH"). Compound 24a: (Diastereoisomer 2): ¾ NMR (OMSO-d6, 400MHz) δ (ppm) 7.59 (d, J=7.43Hz, IH), 7.323-7.29 (m, 4H), 7.26-7.20 (m, IH), 7.19 (s, IH), 7.11 (brs, IH), 7.01 (brs, IH), 6.10 (d, J=3.74Hz, IH), 5.80 (dt, J=12.55Hz and 7.14Hz, IH), 5.63 (d, J=7.43Hz, IH), 5.58-5.56 (m, 2H), 5.05 (dd, J=13.24Hz and 7.83Hz, IH), 4.99 (dd, J=13.24Hz and 7.31Hz, IH), 4.16-4.09 (m, IH), 4.06-3.91 (m, 5H), 3.90-3.88 (m, IH), 3.85 (s, 3H); 31P NMR (DMSO- d6, 162MHz) δ (ppm) 10.19 (s, IP); MS (ESI) m/z = 552.1 (MH+).
Compound 24b [(2R,3S^S,5R)-5-(4-amino-2-oxo-pyrimidin-l-vn
bis[(2-nitrothiazol-5-yl)methyl] phosphate
Figure imgf000121_0001
Compound 24b was synthesized from compound 22a with reagent B3 according to scheme 7 and general procedure B.
Compound 24b: ¾ NMR (OMSO-d6, 400MHz) δ (ppm) 8.07 (s, 1H); 8.05 (s, 1H); 7.56 (d, J=7.46Hz, 1H), 7.1 1 (brs, 1H), 7.01 (brs, 1H), 6.11 (d, J=3.48Hz, 1H), 5.64-5.58 (m, 3H), 5.47- 5.40 (m, 4H), 4.32-4.21 (m, 2H), 4.00-3.91 (m, 2H), 3.90-3.84 (m, 1H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) -1.37 (s, IP); MS (ESI) m/z = 608.0 (MH+).
Compound 24c
(Mixture of diastereomers 64/36)
[(2 3S^S.5RV5-(4-amino-2-oxo-pyrimidin-l -ylV3^-dihydroxy-tetrahydromran-2-yl]methyl (3-methyl-2-nitro-imidazol-4-yDmethyl (2-nitrothiazol-5-yDmethyl phosphate
Figure imgf000121_0002
Compound 24c was synthesized from compound 22a with reagents B2 and B3 according to scheme 7 and general procedure A.
Compound 24c: 1H NMR (OMSO-d6, 400MHz) δ (ppm) 8.06 (s, 0.64H), 8.04 (s, 0.36H), 7.57 (d, J=7.49Hz, 0.64H), 7.56 (d, J=7.49Hz, 0.36H), 7.31 -7.25 (m, 1H), 7.1 1 (brs, 1H), 7.02 (brs, 1H), 6.10 (d, J=3.48Hz, 1H), 5.65-5.57 (m, 3H), 5.44-5.37 (m, 2H), 5.28-5.21 (m, 2H), 4.28- 4.18 (m, 2H), 3.98-3.83 (m, 6H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) -1.43 (s, IP); MS (ESI) m/z = 605.2 (MH+).
Compound 24d
(2 diastereomers)
4-amino-l -[r2R.3S.4S.5RV5-[[rbenzylaminoV[r5-nitro-2- thienyl)methoxy]phosphoryl]oxymethyl]-3^-dihydroxy-tetrahydromran-2-yl]pyrimidin-2-one
Figure imgf000122_0001
Compound 24d was synthesized from compound 22b with reagents Bl and B5 according to scheme 7 and general procedure A. Compound 24d: (Diastereoisomer 1): 1H NMR (OMSO-d6, 400MHz) δ (ppm) 8.02 (d, J=4.21Hz, 1H), 7.54 (d, J=7.49Hz, 1H), 7.35-7.26 (m, 4H), 7.26-7.20 (m, 1H), 7.19 (d, J=4.21Hz, 1H), 7.10 (brs, 1H), 7.00 (brs, 1H), 6.09 (d, J=3.69Hz, 1H), 5.85 (dt, J=12.62Hz and 7.13Hz, 1H), 5.62 (d, J=7.50Hz, 1H), 5.58-5.53 (m, 2H), 5.17 (dd, J=13.81Hz and 8.51Hz, 1H), 5.11 (dd, J=13.81Hz and 8.51Hz, 1H), 4.15-3.87 (m, 7H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 10.37 (s, IP); MS (ESI) m/z = 554.2 (Mrf).
Compound 24d: (Diastereoisomer 2): XH NMR (OMSO-d6, 400MHz) δ (ppm) 8.01 (d, J=4.20Hz, 1H), 7.59 (d, J=7.49Hz, 1H), 7.33-7.28 (m, 4H), 7.25-7.21 (m, 1H), 7.18 (d, J=4.18Hz, 1H), 7.09 (brs, 1H), 7.00 (brs, 1H), 6.10 (d, J=3.66Hz, 1H), 5.89-5.82 (m, 1H), 5.61 (d, J=7.51Hz, IH), 5.57-5.55 (m, 2H), 5.17 (dd, J=13.90Hz and 8.62Hz, IH), 5.1 1 (dd, J=13.90Hz and 8.43Hz, IH), 4.17-4.1 1 (m, IH), 4.07-3.92 m, 5H), 3.89-3.88 (m, IH); 31P NMR (DMSO-i/6, 162MHz) δ (ppm) 10.28 (s, IP); MS (ESI) m/z = 554.2 (Mrf).
Compound 24e
[(2R,3S^S,5R)-5-(4-amino-2-oxo-pyrimidin-l -yl)-3^-dm^
bis[(5-nitro-2-furyl)methyll phosphate
Figure imgf000123_0001
Compound 24e was synthesized from compound 22a with reagent B4 according to scheme 7 and general procedure B.
Compound 24e: 1H NMR (OMSO-d6, 400MHz) δ (ppm) 7.65 (d, J=3.77Hz, IH), 7.64 (d, J=3.77Hz, IH), 7.54 (d, J=7.48Hz, IH), 7.1 1 (brs, IH), 7.02 (brs, IH), 6.97 (d, J=3.77Hz, IH), 6.94 (d, J=3.77Hz, IH), 6.09 (d, J=3.48Hz, IH), 5.63 (d, J=7.48Hz, IH), 5.60 (d, J=3.92Hz, IH), 5.58 (d, J=4.82Hz, IH), 5.21 -5.14 (m, 4H), 4.27-4.21 (m, 2H), 3.98-3.90 (m, 2H), 3.89- 3.84 (m, IH); 31P NMR (OMSO-d6, 162MHz) δ (ppm) -1.27 (s, IP); MS (ESI) m/z = 574.2 (MH+).
Compound 24f
(2 diastereomers)
Benzyl (2SV2-rrr(2R.3S.4S.5RV5-(4-amino-2-oxo-pynmidin-l-vn-3.4-dihvdroxy- tetrahvdrofuran-2-yllmethoxy-[(5-nitro-2-thienyl)methoxylphosphoryllaminolpropanoate
Figure imgf000124_0001
Compound 24f was synthesized from compound 22c with reagents B6 and B5 according to scheme 9 and general procedure A step 1. In this case, the addition of phosphorus oxychloride was followed by the addition of Proton sponge. Step 2 was done according to step 2 of general procedure C.
Compound 24f: (Diastereoisomer 1): 1H NMR (OMSO-d6, 400MHz) δ (ppm) 8.03 (d, J=4.21Hz, 1H), 7.53 (d, J=7.41Hz, 1H), 7.38-7.28 (m, 5H), 7.21 (d, J=4.21Hz, 1H), 7.11 (brs, 1H), 7.02 (brs, 1H), 6.08 (d, J=3.69Hz, 1H), 5.90 (dd, J=9.96Hz and 12.07Hz, 1H), 5.64 (d, J=7.41Hz, 1H), 5.58-5.54 (m, 2H), 5.20-5.15 (m, 2H), 5.15-5.07 (m, 2H), 4.13-4.03 (m, 2H), 3.99-3.80 (m, 4H), 1.31 (d, J=7.17Hz, 3H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 8.57 (s, IP); MS (ESI) m/z = 626.2 (MH+).
Compound 24f: (Diastereoisomer 2): 1H NMR (OMSO-d6, 400MHz) δ (ppm) 8.02 (d, J=4.21Hz, 1H), 7.57 (d, J=7.41Hz, 1H), 7.38-7.28 (m, 5H), 7.19 (d, J=4.21Hz, 1H), 7.10 (brs, 1H), 7.01 (brs, 1H), 6.10 (d, J=3.69Hz, 1H), 5.95 (dd, J=10.15Hz and 12.64Hz, 1H), 5.61 (d, J=7.41Hz, 1H), 5.59-5.55 (m, 2H), 5.20-5.06 (m, 4H), 4.19-4.03 (m, 2H), 3.99-3.91 (m, 2H), 3.90-3.78 (m, 2H), 1.31 (d, J=7.17Hz, 3H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 8.16 (s, IP); MS (ESI) m/z = 626.2 (MH+).
Compound 25
(2 diastereomers)
4-amino-l-[(2R.3S.4S.5RV5-[[(benzylaminoV[(5-nitro-2- mryl)methoxylphosphoi lloxymethyll-3 -dihvdroxy-tetrahvdrofuran-2-yllpyrimidin-2-one
Figure imgf000125_0001
Compound 25 was synthesized with reagent CI according to scheme 8 and general procedure C.
Compound 25: (Diastereoisomer 1): 1H NMR (OMSO-d6, 400MHz) δ (ppm) 7.66 (d, J=3.74Hz, IH), 7.54 (d, J=7.40Hz, IH), 7.33-7.28 (m, 4H), 7.25-7.21 (m, IH), 7.1 1 (brs, IH), 7.03 (brs, IH), 6.88 (d, J=3.74Hz, IH), 6.09 (d, J=3.69Hz, IH), 5.81 (dt, J=12.52Hz and 7.15Hz, IH), 5.63 (d, J=7.40Hz, IH), 5.58-5.56 (m, 2H), 5.04-4.94 (m, 2H), 4.10-4.05 (m, 2H), 4.02-3.88 (m, 5H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 10.49 (s, IP); MS (ESI) m/z = 538 (MH+). Compound 25: (Diastereoisomer 2): 1H NMR (OMSO-d6, 400MHz) δ (ppm) 7.65 (d, J=3.78Hz, IH), 7.58 (d, J=7.43Hz, IH), 7.33-7.28 (m, 4H), 7.25-7.21 (m, IH), 7.10 (brs, IH), 7.02 (brs, IH), 6.87 (d, J=3.78Hz, IH), 6.09 (d, J=3.65Hz, IH), 5.82(dt, J=12.45Hz and 7.17Hz, IH), 5.63 (d, J=7.43Hz, IH), 5.58-5.56 (m, 2H), 5.04-4.93 (m, 2H), 4.15-4.09 (m, IH), 4.06-3.91 (m, 5H), 3.89-3.87 (m, IH); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 10.39 (s, IP);
Figure imgf000125_0002
Compound 26:
(2 diastereomers)
Benzyl (2SV2-rr(4aR.6R.7S.7aRV6-(6-amino-2-chloro-purin-9-ylV7-fluoro-2-oxo-4a.6.7.7a- tetrahydro-4H-furo[3.2-d][1.3.2]dioxaphosphinin-2-yl]amino]propanoate
Figure imgf000126_0001
Compound 26 was synthesized from compound 1 a according to scheme 3 and to the following procedure D.
Compound 26a (Diastereoisomer 1): ¾ NMR (DMSO-i/6, 400MHz) δ (ppm) 8.35 (d, J=1.43Hz, 1H), 7.97 (brs, 2H), 7.41 -7.29 (m, 5H), 6.60 (dd, J=5.14Hz and 6.61Hz, 1H), 6.14 (dd, J=10.15Hz and 13.40Hz, 1H), 5.80 (td, J=55.34Hz and 7.34Hz, 1H), 5.39-5.26 (m, 1H), 5.18 (d, J=12.62Hz, 1H), 5.14 (d, J=12.62Hz, 1H), 4.63-4.52 (m, 1H), 4.52-4.42 (m, 1H), 4.28-4.18 (m, 1H), 4.00-3.87 (m, 1H), 1.37 (d, J=7.15Hz, 3H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 2.75 (s, IP); 19F NMR (OMSO-d6, 376MHz) δ (ppm) -199.64 (s, IF); MS (ESI) m/z = 527.0 (MH+). Compound 26a (Diastereoisomer 2): ¾ NMR (DMSO-i/6, 400MHz) δ (ppm) 8.55 (d, J=l .41Hz, 1H), 7.95 (brs, 2H), 7.40-7.34 (m, 5H), 6.60-6.57 (m, 1H), 6.30 (dd, J=13.69Hz and 9.94Hz, 1H), 5.75 (td, J=55.52Hz and 7.13Hz, 1H), 5.48-5.39 (m, 1H), 5.19-5.12 (m, 2H), 4.56-4.51 (m, 1H), 4.42-4.34 (m, 1H), 4.08-4.02 (m, 1H), 3.96-3.86 (m, 1H), 1.37 (d, J=7.10Hz, 3H); 31P NMR (DMSO-i/6, 162MHz) δ (ppm) 4.82 (s, IP); 19F NMR (OMSO-d6, 376MHz) δ (ppm) -199.46 (s, IF); MS (ESI) m/z = 527.0 (MR+).
Compound 28a and 28b
(2 diastereomers)
Ethyl (2SV2-rr(4aR.6R.7S.7aSV6-(6-amino-2-fluoro-punn-9-vn-7-hvdroxy-2-oxo-4a.6.7.7a- tetrahydro-4H-furo[3,2-dl[l,3,21dioxaphosphinin-2-yllaminolpropanoate
Figure imgf000127_0001
Compound 28 was synthesized from 2'OCBz-fludarabine according to scheme 3bis and to the procedure D. In this case, additional step of deprotection using ¾, Pd/C 5% in ethanol was used to afford the 2 expected diastereoisomers.
Compound 28a (Diastereoisomer 1): 31P NMR (OMSO-d6, 162MHz) δ (ppm) 3.01 (s, IP); MS (ESI) m/z = 447 (MH+).
Compound 28b (Diastereoisomer 2): 31P NMR (DMSO-i 6, 162MHz) δ (ppm) 4.97 (s, IP); MS (ESI) m/z = 447 (MH+).
Compound 29
(Mixture of diastereomers)
Methyl (2SV2-[[[(2R.3S.4S.5RV5-(6-amino-2-fluoro-purin-9-ylV3.4-dihvdroxy-tetrahvdrofuran- 2-yl]methoxy-(2-methoxy-2-oxo-ethoxy)phosphoryl]amino]-4-methyl-pentanoate
Figure imgf000127_0002
Compound 29 was synthesized according to scheme 10 and to the following procedure.
To a solution of 2'OCBz-fludarabine (1.19mmol) in THF (lOmL/mmol) at 0°C was added a solution of tert-butylmagnesium chloride, 1M in THF (3.70mmol). The reaction mixture was stirred at room temperature for lhour, and then cooled down to 0°C. The appropriate reagent Dl (1.31mmol) in THF (lOmL/mmol) was added, and the reaction mixture was stirred at 0°C for lhour and for 3days at room temperature. The reaction mixture was diluted with ethyl acetate and washed with a saturated solution of NH4C1, water and brine. The combined organic layers were dried, filtered and concentrated under reduced pressure. The crude residue was purified by flash chromatography on silica (DCM/ethanol or DCM/methanol) to afford the expected intermediate;
To a solution of this intermediate (0.07mmol) in EtOH (5mL/mmol) was added Palladium Black (0.28mmol) under nitrogen. After several flush N2/vaccum and vaccum/H2, the reaction mixture was stirred under H2 Atmosphere for lh at room temperature. The reaction mixture was filtered on Celite and concentrated under reduced pressure. The crude residue was triturated in a DCM/pentane mixture, and then purified by flash chromatography on silica gel (DCM/methanol: 0 to 5%) to afford the expected compound as solid as mixture of diastereoisomers.
Compound 29: 1H NMR (OMSO-d6, 400MHz) δ (ppm) 8.13 (s, 0.4H), 8.1 1 (s, 0.6H), 7.83 (brs, 2H), 6.18-6.16 (m, 1H), 5.78-5.65 (m, 2H), 4.52-4.44 (m, 2H), 4.26-4.09 (m, 4H), 3.99-3.93 (m, 1H), 3.76-3.69 (m, 1H), 3.68 (s, 1.8H), 3.66 (s, 1.2H), 3.62 (s, 1.2H), 3.60 (s, 1.8H), 1.73-1.65 (m, 1H), 1.52-1.22 (m, 2H), 0.88-0.82 (m, 6H); 31P NMR (OMSO-d6, 162MHz) δ (ppm) 8.68 (s, 0.6P), 8.51 (s, 0.4P); 19F NMR (OMSO-d6, 376MHz) δ (ppm) -52.38 (m, IF); MS (ESI) m/z = 565 (MH+).
Characterization data of the compounds provided herein are described in Table 1 :
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0001
Figure imgf000135_0001
Figure imgf000136_0001
Figure imgf000137_0001
[00391] Using similar procedures, the following additional compounds were also prepared.
Figure imgf000137_0002
Figure imgf000138_0001
Figure imgf000139_0001
Using similar procedures, the following additional compounds were also prepared:
Figure imgf000139_0002
Figure imgf000140_0001
Pharmaceutical Compositions and Methods of Administration
Derivatives of a variety of therapeutic agents can be formulated into pharmaceutical compositions using methods available in the art and those disclosed herein. Therapeutic agents that can be derivatized to phosphate derivatives include any anti-cancer agent that includes, or has been derivatized to include a reactive group for attachment of the phosphate moiety, including but not limited to nucleosides and nucleoside analogues including acyclic nucleosides. Any of the compounds disclosed herein can be provided in the appropriate pharmaceutical composition and be administered by a suitable route of administration.
The methods provided herein encompass administering pharmaceutical compositions containing at least one compound as described herein, including a compound of general Formulas I, Ila, lib, or lie, if appropriate in the salt form, either used alone or in the form of a combination with one or more compatible and pharmaceutically acceptable carriers, such as excipients, diluents or adjuvants, or with another anti-cancer agent.
In certain embodiments, the second agent can be formulated or packaged with the compound provided herein. Of course, the second agent will only be formulated with the compound provided herein when, according to the judgment of those of skill in the art, such co-formulation should not interfere with the activity of either agent or the method of administration. In certain embodiments, the compound provided herein and the second agent are formulated separately. They can be packaged together, or packaged separately, for the convenience of the practitioner of skill in the art.
In clinical practice the active agents provided herein may be administered by any conventional route, in particular orally, parenterally, rectally or by inhalation (e.g. in the form of aerosols). In certain embodiments, the compound provided herein is administered orally.
Use may be made, as solid compositions for oral administration, of tablets, pills, hard gelatin capsules, powders or granules. In these compositions, the active product is mixed with one or more inert diluents or adjuvants, such as sucrose, lactose, or starch.
These compositions can comprise substances other than diluents, for example a lubricant, such as magnesium stearate, or a coating intended for controlled release.
Use may be made, as liquid compositions for oral administration, of solutions which are pharmaceutically acceptable, suspensions, emulsions, syrups, and elixirs containing inert diluents, such as water or liquid paraffin. These compositions can also comprise substances other than diluents, for example wetting, sweetening or flavoring products.
The compositions for parenteral administration can be emulsions or sterile solutions. Use may be made, as solvent or vehicle, of propylene glycol, a polyethylene glycol, vegetable oils, in particular olive oil, or injectable organic esters, for example ethyl oleate. These compositions can also contain adjuvants, in particular wetting, isotonizing, emulsifying, dispersing, and stabilizing agents. Sterilization can be carried out in several ways, for example using a bacteriological filter, by radiation or by heating. They can also be prepared in the form of sterile solid compositions which can be dissolved at the time of use in sterile water or any other injectable sterile medium.
The compositions for rectal administration are suppositories or rectal capsules which contain, in addition to the active principle, excipients such as cocoa butter, semi-synthetic glycerides or polyethylene glycols. The compositions can also be aerosols. For use in the form of liquid aerosols, the compositions can be stable sterile solutions or solid compositions dissolved at the time of use in apyrogenic sterile water, in saline or any other pharmaceutically acceptable vehicle. For use in the form of dry aerosols intended to be directly inhaled, the active principle is finely divided and combined with a water-soluble solid diluent or vehicle, for example dextran, mannitol, or lactose.
In one embodiment, a composition provided herein is a pharmaceutical composition or a single unit dosage form. Pharmaceutical compositions and single unit dosage forms provided herein comprise a prophylactically or therapeutically effective amount of one or more prophylactic or therapeutic agents (e.g., a compound provided herein, or other prophylactic or therapeutic agent), and a typically one or more pharmaceutically acceptable carriers or excipients. In a specific embodiment and in this context, the term "pharmaceutically acceptable" means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term "carrier" includes a diluent, adjuvant (e.g., Freund's adjuvant (complete and incomplete)), excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water can be used as a carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Examples of suitable pharmaceutical carriers are described in "Remington 's Pharmaceutical Sciences" by E.W. Martin.
Typical pharmaceutical compositions and dosage forms comprise one or more excipients. Suitable excipients are well-known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol, and the like. Whether a particular excipient is suitable for incorporation into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a subject and the specific active ingredients in the dosage form. The composition or single unit dosage form, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
Lactose free compositions provided herein can comprise excipients that are well known in the art and are listed, for example, in the U.S. Pharmocopia (USP) SP (XXI) NF (XVI). In general, lactose free compositions comprise an active ingredient, a binder/filler, and a lubricant in pharmaceutically compatible and pharmaceutically acceptable amounts. Exemplary lactose free dosage forms comprise an active ingredient, microcrystalline cellulose, pre gelatinized starch, and magnesium stearate.
Further encompassed herein are anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds. For example, the addition of water {e.g., 5%) is widely accepted 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. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80. In effect, water and heat accelerate the decomposition of some compounds. Thus, the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
Anhydrous pharmaceutical compositions and dosage forms provided herein can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions. Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine can be anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained. Accordingly, anhydrous compositions can 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, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
Further provided are pharmaceutical compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose. Such compounds, which are referred to herein as "stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
The pharmaceutical compositions and single unit dosage forms can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations, and the like. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Such compositions and dosage forms will contain a prophylactically or therapeutically effective amount of a prophylactic or therapeutic agent, in certain embodiments, in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the subject. The formulation should suit the mode of administration. In a certain embodiment, the pharmaceutical compositions or single unit dosage forms are sterile and in suitable form for administration to a subject, for example, an animal subject, such as a mammalian subject, for example, a human subject.
A pharmaceutical composition is formulated to be compatible with its intended route of administration. Examples of routes of administration include, but are not limited to, parenteral, e.g., intravenous, intradermal, subcutaneous, intramuscular, subcutaneous, oral, buccal, sublingual, inhalation, intranasal, transdermal, topical, transmucosal, intra-tumoral, intra- synovial, and rectal administration. In a specific embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous, subcutaneous, intramuscular, oral, intranasal, or topical administration to human beings. In an embodiment, a pharmaceutical composition is formulated in accordance with routine procedures for subcutaneous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocamne to ease pain at the site of the injection.
Examples of dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal administration to a subject, including suspensions (e.g., aqueous or non aqueous liquid suspensions, oil in water emulsions, or a water in oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a subject; and sterile solids (e.g., crystalline or amorphous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a subject.
The composition, shape, and type of dosage forms provided herein will typically vary depending on their use. For example, a dosage form used in the initial treatment of viral infection may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the maintenance treatment of the same infection. Similarly, a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder. These and other ways in which specific dosage forms encompassed herein will vary from one another will be readily apparent to those skilled in the art. See, e.g., Remington 's Pharmaceutical Sciences, 20th ed., Mack Publishing, Easton PA (2000).
Generally, the ingredients of compositions are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration. Typical dosage forms comprise a compound provided herein, or a pharmaceutically acceptable salt, solvate or hydrate thereof lie within the range of from about 0.1 mg to about 1000 mg per day, given as a single once-a-day dose in the morning or as divided doses throughout the day taken with food. Particular dosage forms can have about 0.1, 0.2, 0.3, 0.4, 0.5, 1.0, 2.0, 2.5, 5.0, 10.0, 15.0, 20.0, 25.0, 50.0, 100, 200, 250, 500, or 1000 mg of the active compound.
Oral Dosage Forms
Pharmaceutical compositions that are suitable for oral administration can be presented as discrete dosage forms, such as, but are not limited to, tablets {e.g., chewable tablets), cap lets, capsules, and liquids {e.g., flavored syrups). Such dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington 's Pharmaceutical Sciences, 20th ed., Mack Publishing, Easton PA (2000).
In certain embodiments, the oral dosage forms are solid and prepared under anhydrous conditions with anhydrous ingredients, as described in detail in the sections above. However, the scope of the compositions provided herein extends beyond anhydrous, solid oral dosage forms. As such, further forms are described herein.
Typical oral dosage forms are prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques. Excipients can take a wide variety of forms depending on the form of preparation desired for administration. For example, excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents. Examples of excipients suitable for use in solid oral dosage forms {e.g., powders, tablets, capsules, and caplets) include, but are not limited to, starches, sugars, micro crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
Because of their ease of administration, tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
For example, a tablet can be prepared by compression or molding. Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free flowing form such as powder or granules, optionally mixed with an excipient. Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
Examples of excipients that can be used in oral dosage forms include, but are not limited to, binders, fillers, disintegrants, and lubricants. 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, (e.g., Nos. 2208, 2906, 2910), microcrystalline cellulose, and mixtures thereof.
Examples of fillers suitable 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. The binder or filler in pharmaceutical compositions is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL PH 101, AVICEL PH 103 AVICEL RC 581 , AVICEL PH 105 (available from FMC Corporation, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof. A specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC 581. Suitable anhydrous or low moisture excipients or additives include AVICEL PH 103™ and Starch 1500 LM.
Disintegrants are used in the compositions to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms. The amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art. Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
Disintegrants that can be used in pharmaceutical compositions and dosage forms 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, pre gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
Lubricants that can be used in pharmaceutical compositions and dosage forms 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, ethyl laureate, agar, and mixtures thereof. Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB O SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are incorporated.
Delayed Release Dosage Forms
Active ingredients such as the compounds provided herein can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art.
Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770;
3,916,899; 3,536,809; 3,598,123; 4,008,719; 5,674,533; 5,059,595; 5,591,767; 5,120,548;
5,073,543; 5,639,476; 5,354,556; 5,639,480; 5,733,566; 5,739,108; 5,891,474; 5,922,356;
5,972,891; 5,980,945; 5,993,855; 6,045,830; 6,087,324; 6,113,943; 6,197,350; 6,248,363; 6,264,970; 6,267,981; 6,376,461; 6,419,961 ; 6,589,548; 6,613,358; and 6,699,500; each of which is incorporated herein by reference in its entirety. Such dosage forms can be used to provide slow or controlled release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions. Suitable controlled release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients provided herein. Thus encompasseed herein are single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled release.
All controlled release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non controlled counterparts. Ideally, the use of an optimally designed controlled release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time. Advantages of controlled release formulations include extended activity of the drug, reduced dosage frequency, and increased subject compliance. In addition, controlled release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
Most controlled release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
In certain embodiments, the drug may be administered using intravenous infusion, an implantable osmotic pump, a transdermal patch, liposomes, or other modes of administration. In one embodiment, a pump may be used (see, Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al, Surgery 55:507 (1980); Saudek et al, N. Engl. J. Med. 321:574 (1989)). In another embodiment, polymeric materials can be used. In yet another embodiment, a controlled release system can be placed in a subject at an appropriate site determined by a practitioner of skill, i.e., thus requiring only a fraction of the systemic dose {see, e.g., Goodson, Medical Applications of Controlled Release, vol. 2, pp. 115-138 (1984)). Other controlled release systems are discussed in the review by Langer {Science 2 9: 1527-1533 (1990)). The active ingredient can be dispersed in a solid inner matrix, e.g., polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethyleneterephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinylacetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinylalcohol and cross- linked partially hydrolyzed polyvinyl acetate, that is surrounded by an outer polymeric membrane, e.g., polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinylacetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinylchloride copolymers with vinyl acetate, vinylidene chloride, ethylene and propylene, ionomer polyethylene terephthalate, butyl rubber epichlorohydrin rubbers, ethylene/vinyl alcohol copolymer, ethylene/vinyl acetate/vinyl alcohol terpolymer, and ethylene/vinyloxyethanol copolymer, that is insoluble in body fluids. The active ingredient then diffuses through the outer polymeric membrane in a release rate controlling step. The percentage of active ingredient in such parenteral compositions is highly dependent on the specific nature thereof, as well as the needs of the subject.
Parenteral Dosage Forms
In one embodiment, provided are parenteral dosage forms. Parenteral dosage forms can be administered to subjects by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses subjects' natural defenses against contaminants, parenteral dosage forms are typically sterile or capable of being sterilized prior to administration to a subject. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions. Suitable vehicles that can be used to provide parenteral dosage forms are well known to those skilled in the art. In certain embodiments, examples of a suitable vehicle include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
Compounds that increase the solubility of one or more of the active ingredients disclosed herein can also be incorporated into the parenteral dosage forms.
Transdermal, Topical & Mucosal Dosage Forms
Also provided are transdermal, topical, and mucosal dosage forms. Transdermal, topical, and mucosal dosage forms include, but are not limited to, ophthalmic solutions, sprays, aerosols, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington 's Pharmaceutical Sciences, 16th, 18th and 20th eds., Mack Publishing, Easton PA (1980, 1990 & 2000); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels. Further, transdermal dosage forms include "reservoir type" or "matrix type" patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
Suitable excipients {e.g., carriers and diluents) and other materials that can be used to provide transdermal, topical, and mucosal dosage forms encompassed herein are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied. With that fact in mind, typical excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane -1,3- diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are non toxic and pharmaceutically acceptable. Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington 's Pharmaceutical Sciences, 16 , 18th and 20 eds., Mack Publishing, Easton PA (1980, 1990 & 2000).
Depending on the specific tissue to be treated, additional components may be used prior to, in conjunction with, or subsequent to treatment with active ingredients provided. For example, penetration enhancers can be used to assist in delivering the active ingredients to the tissue. Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
The pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied, may also be adjusted to improve delivery of one or more active ingredients. Similarly, the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery. Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery. In this regard, stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery enhancing or penetration enhancing agent. Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
Method of Use
In one embodiment, provided herein is a method for treating a proliferative disease in a subject, which comprises administering to the subject a therapeutically effective amount of a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or a hydrate thereof.
In certain embodiments, the therapeutically effective amount is ranging from about 0.1 to about 100 mg/kg/day, from about 0.1 to about 50 mg/kg/day, from about 0.1 to about 40 mg/kg/day, from about 0.1 to about 30 mg/kg/day, from about 0.1 to about 25 mg/kg/day, from about 0.1 to about 20 mg/kg/day, from about 0.1 to about 15 mg/kg/day, from about 0.1 to about 10 mg/kg/day, or from about 0.1 to about 5 mg/kg/day. In one embodiment, the therapeutically effective amount is ranging from about 0.1 to about 100 mg/kg/day. In another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 50 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 40 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 30 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 25 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 20 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 15 mg/kg/day. In yet another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 10 mg/kg/day. In still another embodiment, the therapeutically effective amount is ranging from about 0.1 to about 5 mg/kg/day.
It is understood that the administered dose can also be expressed in units other than mg/kg/day. For example, doses for parenteral administration can be expressed as mg/m2/day. One of ordinary skill in the art would readily know how to convert doses from mg/kg/day to mg/m2/day to given either the height or weight of a subject or both (see, www.fda.gov/cder/cancer/animalframe.htm). For example, a dose of 1 mg/m2/day for a 65 kg human is approximately equal to 38 mg/kg/day.
In certain embodiments, the subject is a mammal. In certain embodiments, the subject is a human.
In one embodiment is a compound or a pharmaceutically acceptable salt, or solvate thereof, or stereoisomeric, tautomeric, or polymorphic form thereof for use in therapy.
In certain embodiments, the proliferative disease is a carcinoma, including, but not limited to, Kit-mediated carcinomas, adenocarcinoma, squamous cell carcinoma, adenosquamous carcinoma, teratocarcinoma, head and neck cancer, brain cancer, intracranial carcinoma, glioblastoma (including PDGFR-mediated glioblastoma), glioblastoma multiforme (including PDGFR-mediated glioblastoma multiforme), neuroblastoma, cancer of the larynx, multiple endocrine neoplasias 2A and 2B (MENS 2A and MENS 2B) (including RET-mediated MENS), thyroid cancer (including sporadic and familial medullary thyroid carcinoma), papillary thyroid carcinoma, parathyroid carcinoma (including any RET-mediated thyroid carcinoma), follicular thyroid cancer, anaplastic thyroid cancer, bronchial carcinoid, oat cell carcinoma, lung cancer, small-cell lung cancer (including FLT3 and/or Kit-mediated small cell lung cancer), non-small- cell lung cancer, stomach/gastric cancer, gastrointestinal cancer, gastrointestinal stromal tumors (GIST) (including Kit-mediated GIST and PDGFR a-mediated GIST), colon cancer, colorectal cancer, pancreatic cancer, islet cell carcinoma, hepatic/liver cancer, metastases to the liver, bladder cancer, renal cell cancer (including PDGFR-mediated renal cell cancer), cancers of the genitourinary tract, ovarian cancer (including Kit-mediated and/or PDGFR-mediated ovarian cancer), endometrial cancer (including CSF-lR-mediated endometrial cancer), cervical cancer, breast cancer (including FLT3 -mediated and/or PDGFR-mediated breast cancer), prostate cancer (including Kit-mediated prostate cancer), germ cell tumors (including Kit-mediated germ cell tumors), seminomas (including Kit-mediated seminomas), dysgerminomas (including Kit- mediated dysgerminomas), melanoma (including PDGFR-mediated melanoma), metastases to the bone (including CSF-lR-mediated bone metastases), metastatic tumors (including VEGFR- mediated tumors), stromal tumors, neuroendocrine tumors, tumor angiogenesis (including VEGFR-mediated tumor angiogenesis), and mixed mesodermal tumors.
In certain embodiments, the proliferative disease is sarcomas, including, but not limited to, PDGFR-mediated sarcomas, osteosarcoma, osteogenic sarcoma, bone cancer, glioma (including PDGFR-mediated and/or CSF-lR-mediated glioma), astrocytoma, vascular tumors (including VEGFR-mediated vascular tumors), Kaposi's sarcoma, carcinosarcoma, hemangiosar comas (including VEGFR3 -mediated hemangiosarcomas), and lymphangiosarcoma (including VEGFR3 -mediated lymphangiosarcoma).
In certain embodiments, the proliferative disease is a hematologic malignancy. In certain embodiments, the proliferative disease is a relapsed hematologic malignancy. In certain embodiments, the proliferative disease is a refractory hematologic malignancy. In certain embodiments, the proliferative disease is a drug-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a multidrug-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a Bcr-Abl kinase inhibitor-resistant hematologic malignancy. In certain embodiments, the proliferative disease is an imatinib-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a dasatinib- resistant hematologic malignancy. In certain embodiments, the proliferative disease is a nilatinib-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a bosutinib-resistant hematologic malignancy. In certain embodiments, the proliferative disease is a cytarabine-resistant hematologic malignancy.
In certain embodiments, the hematologic malignancy is myeloma, leukemia, myeloproliferative diseases, acute myeloid leukemia (AML) (including FLT3 mediated and/or KIT-mediated and/or CSFlR-mediated acute myeloid leukemia), chronic myeloid leukemias (CML) (including FLT3- mediated and/or PDGFR-mediated chronic myeloid leukemia), myelodysplastic leukemias (including FLT3 -mediated myelodysplastic leukemia), myelodysplastic syndrome (including FLT3 mediated and/or Kit-mediated myelodysplastic syndrome), idiopathic hypereosinophilic syndrome (HES) (including PDGFR-mediated HES), chronic eosinophilic leukemia (CEL) (including PDGFR-mediated CEL), chronic myelomonocytic leukemia (CMML), mast cell leukemia (including Kit-mediated mast cell leukemia), or systemic mastocytosis (including Kit- mediated systemic mastocytosis).
In certain embodiments, the hematologic malignancy is lymphoma, lymphoproliferative diseases, acute lymphoblastic leukemia (ALL), B-cell acute lymphoblastic leukemias, T-cell acute lymphoblastic leukemias, chronic lymphocytic leukemia (CLL), natural killer (NK) cell leukemia, B-cell lymphoma, T-cell lymphoma, or natural killer (NK) cell lymphoma.
In an embodiment, the hematologic malignancy is myelodysplastic syndrome (MDS).
In certain embodiments, the hematologic malignancy is Langerhans cell histiocytosis (including CSF-lR-mediated and/or FLT3-mediated Langerhans cell histiocytosis), mast cell tumors, or mastocytosis.
In certain embodiments, the hematologic malignancy is leukemia. In certain embodiments, the hematologic malignancy is relapsed leukemia. In certain embodiments, the hematologic malignancy is refractory leukemia. In certain embodiments, the hematologic malignancy is drug-resistant leukemia. In certain embodiments, the hematologic malignancy is multidrug- resistant leukemia. In certain embodiments, the hematologic malignancy is a Bcr-Abl kinase inhibitor-resistant leukemia. In certain embodiments, the hematologic malignancy is imatinib- resistant leukemia. In certain embodiments, the hematologic malignancy is dasatinib-resistant leukemia. In certain embodiments, the hematologic malignancy is nilatinib-resistant leukemia. In certain embodiments, the hematologic malignancy is bosutinib-resistant leukemia. In certain embodiments, the hematologic malignancy is cytarabine-resistant leukemia. In certain embodiments, the leukemia is acute leukemia. In certain embodiments, the leukemia is relapsed acute leukemia. In certain embodiments, the leukemia is refractory acute leukemia. In certain embodiments, the leukemia is drug-resistant acute leukemia. In certain embodiments, the leukemia is multidrug-resistant acute leukemia. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant acute leukemia. In certain embodiments, the leukemia is imatinib-resistant acute leukemia. In certain embodiments, the leukemia is dasatinib-resistant acute leukemia. In certain embodiments, the leukemia is nilatinib-resistant acute leukemia. In certain embodiments, the leukemia is bosutinib-resistant acute leukemia. In certain embodiments, the leukemia is cytarabine-resistant acute leukemia. In certain embodiments, the leukemia is a hereditary leukemia. In certain embodiments, the hereditary leukemia is severe congenital neutropenia (SCN). In certain embodiments, the hereditary leukemia is familial platelet disorder with acute myelogenous leukemia (FDP/AML). In certain embodiments, the leukemia is caused by LEF1. In certain embodiments, the leukemia is mediated by LEF1. In certain embodiments, the leukemia is caused by GSK3.
In certain embodiments, the leukemia is ALL. In certain embodiments, the leukemia is relapsed ALL. In certain embodiments, the leukemia is refractory ALL. In certain embodiments, the leukemia is drug-resistant ALL. In certain embodiments, the leukemia is multidrug-resistant ALL. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant ALL. In certain embodiments, the leukemia is imatinib-resistant ALL. In certain embodiments, the leukemia is dasatinib-resistant ALL. In certain embodiments, the leukemia is nilatinib-resistant ALL. In certain embodiments, the leukemia is bosutinib-resistant ALL. In certain embodiments, the leukemia is cytarabine-resistant ALL.
In one embodiment, ALL is leukemia that originates in the blast cells of the bone marrow (B- cells), thymus (T-cells), or lymph nodes. ALL is categorized according to the French- American- British (FAB) Morphological Classification Scheme as LI - mature-appearing lymphoblasts (T- cells or pre-B-cells), L2 - immature and pleomorphic (variously shaped) lymphoblasts (T-cells or pre-B-cells), and L3 - lymphoblasts (B-cells; Burkitt's cells). In another embodiment, ALL originates in the blast cells of the bone marrow (B-cells). In yet another embodiment, ALL originates in the thymus (T-cells). In yet another embodiment, ALL originates in the lymph nodes. In yet another embodiment, ALL is LI type characterized by mature-appearing lymphoblasts (T-cells or pre-B-cells). In yet another embodiment, ALL is L2 type characterized by immature and pleomorphic (variously shaped) lymphoblasts (T-cells or pre-B-cells). In still another embodiment, ALL is L3 type characterized by lymphoblasts (B-cells; Burkitt's cells).
In certain embodiments, the leukemia is AML. In certain embodiments, the leukemia is relapsed AML. In certain embodiments, the leukemia is refractory AML. In certain embodiments, the leukemia is drug-resistant AML. In certain embodiments, the leukemia is multidrug-resistant AML. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant AML. In certain embodiments, the leukemia is imatinib-resistant AML. In certain embodiments, the leukemia is dasatinib-resistant AML. In certain embodiments, the leukemia is nilatinib-resistant AML. In certain embodiments, the leukemia is bosutinib-resistant AML. In certain embodiments, the leukemia is cytarabine-resistant AML. In certain embodiments, AML has a RAS mutation. In certain embodiments, the RAS mutation is NRAS, KRAS, or HRAS. In certain embodiments, the RAS mutation is NRAS. In certain embodiments, the RAS mutation is KRAS. In certain embodiments, the RAS mutation is HRAS.
In certain embodiments, AML is undifferentiated AML (MO), myeloblastic leukemia (Ml), myeloblastic leukemia (M2), promyelocytic leukemia (M3 or M3 variant [M3V]), myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]), monocytic leukemia (M5), erythro leukemia (M6), or megakaryoblastic leukemia (M7). In one embodiment, AML is undifferentiated AML (MO). In another embodiment, AML is myeloblastic leukemia (Ml). In yet another embodiment, AML is myeloblastic leukemia (M2). In yet another embodiment, AML is promyelocytic leukemia (M3 or M3 variant [M3V]). In yet another embodiment, AML is myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]). In yet another embodiment, AML is monocytic leukemia (M5). In yet another embodiment, AML is erythroleukemia (M6). In still another embodiment, AML is megakaryoblastic leukemia (M7). In certain embodiments, the leukemia is chronic leukemia. In certain embodiments, the leukemia is relapsed chronic leukemia. In certain embodiments, the leukemia is refractory chronic leukemia. In certain embodiments, the leukemia is drug-resistant chronic leukemia. In certain embodiments, the leukemia is multidrug-resistant chronic leukemia. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant chronic leukemia. In certain embodiments, the leukemia is imatinib-resistant chronic leukemia. In certain embodiments, the leukemia is dasatinib-resistant chronic leukemia. In certain embodiments, the leukemia is nilatinib-resistant chronic leukemia. In certain embodiments, the leukemia is bosutinib-resistant chronic leukemia. In certain embodiments, the leukemia is cytarabine-resistant chronic leukemia.
In certain embodiments, the leukemia is CLL. In certain embodiments, the leukemia is relapsed CLL. In certain embodiments, the leukemia is refractory CLL. In certain embodiments, the leukemia is drug-resistant CLL. In certain embodiments, the leukemia is multidrug-resistant CLL. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant CLL. In certain embodiments, the leukemia is imatinib-resistant CLL. In certain embodiments, the leukemia is dasatinib-resistant CLL. In certain embodiments, the leukemia is nilatinib-resistant CLL. In certain embodiments, the leukemia is bosutinib-resistant CLL. In certain embodiments, the leukemia is cytarabine-resistant CLL.
In certain embodiments, the leukemia is CML. In certain embodiments, the leukemia is relapsed CML. In certain embodiments, the leukemia is refractory CML. In certain embodiments, the leukemia is drug-resistant CML. In certain embodiments, the leukemia is multidrug-resistant CML. In certain embodiments, the leukemia is a Bcr-Abl kinase inhibitor-resistant CML. In certain embodiments, the leukemia is imatinib-resistant CML. In certain embodiments, the leukemia is dasatinib-resistant CML. In certain embodiments, the leukemia is nilatinib-resistant CML. In certain embodiments, the leukemia is bosutinib-resistant CML. In certain embodiments, the leukemia is cytarabine-resistant CML. In certain embodiments, the leukemia is juvenile CML. In certain embodiments, the leukemia is juvenile CML with one or more NF-1 mutations.
In certain embodiments, the leukemia is T-cell leukemia. In one embodiment, the T-cell leukemia is peripheral T-cell leukemia, T-cell lymphoblastic leukemia, cutaneous T-cell leukemia, and adult T-cell leukemia. In another embodiment, the T-cell leukemia is peripheral T-cell leukemia. In yet another embodiment, the T-cell leukemia is T-cell lymphoblastic leukemia. In yet another embodiment, the T-cell leukemia is cutaneous T-cell leukemia. In still another embodiment, the T-cell leukemia is adult T-cell leukemia.
In certain embodiments, the leukemia is Philadelphia positive. In one embodiment, the Philadelphia positive leukemia is Philadelphia positive AML, including, but not limited to, undifferentiated AML (MO), myeloblastic leukemia (Ml), myeloblastic leukemia (M2), promyelocytic leukemia (M3 or M3 variant [M3V]), myelomonocytic leukemia (M4 or M4 variant with eosinophilia [M4E]), monocytic leukemia (M5), erythroleukemia (M6), or megakaryoblastic leukemia (Ml). In another embodiment, the Philadelphia positive leukemia is Philadelphia positive ALL.
In certain embodiments, the proliferative disease is cancer, including, but not limited to, head and neck cancer (originating lip, oral cavity, oropharynx, hypopharynx, larynx, nasopharynx, nasal cavity, paranasal sinuses, or salivary glands), lung cancer (including small cell lung cancer and non-small cell lung cancer), gastrointestinal tract cancers (including esophageal cancer), gastric cancer, colorectal cancer, anal cancer, pancreatic cancer, liver cancer, gallbladder cancer, extrahepatic bile duct cancer, cancer of the ampulla of vater, breast cancer, gynecologic cancers (including cancer of uterine cervix), cancer of the uterine body, vaginal cancer, vulvar cancer, ovarian cancer, gestational trophoblastic cancer neoplasia, testicular cancer, urinary tract cancers (including renal cancer), urinary blader cancer, prostate cancer, penile cancer, urethral cancer, neurologic tumors, endocrine neoplasms (including carcinoid and islet cell tumors), pheochromocytoma, adrenal cortical carcinoma, parathyroid carcinoma, and metastases to endocrine glands.
Further examples of cancers are basal cell carcinoma, squamous cell carcinoma, chondrosarcoma (a cancer arising in cartilage cells), mesenchymal-chondrosarcoma, soft tissue sarcomas (including malignant tumors that may arise in any of the mesodermal tissues (muscles, tendons, vessels that carry blood or lymph, joints and fat)), soft tissue sarcomas (include alveolar soft-part sarcoma), angiosarcoma, fibrosarcoma, leiomyosarcoma, liposarcoma, malignant fibrous histiocytoma, hemangiopericytoma, mesenchymoma, schwannoma, peripheral neuroectodermal tumours, rhabdomyosarcoma, synovial sarcoma, gestational trophoblastic tumor (malignancy in which the tissues formed in the uterus following conception become cancerous), Hodgkin's lymphoma, and laryngeal cancer.
In certain embodiments, the proliferative disease is a nonmalignant proliferation disease, including, but not limited to, atherosclerosis (including PDGFR-mediated atherosclerosis), restenosis following vascular angioplasty (including PDGFR-mediated restenosis), and fibroproliferative disorders (including obliterative bronchiolitis and idiopathic myelofibrosis).
In certain embodiments, the proliferative disease is an inflammatory disease or disorder related to immune dysfunction, immunodeficiency, or immunomodulation, including, but not limited to, autoimmune diseases, tissue transplant rejection, graft-versus-host disease, wound healing, kidney disease, multiple sclerosis, thyroiditis, type 1 diabetes, sarcoidosis, allergic rhinitis, inflammatory bowel diseases (including Crohn's disease and ulcerative colitis (UC)), systemic lupus erythematosis (SLE), arthritis, osteoarthritis, rheumatoid arthritis, osteoporosis, asthma, and chronic obstructive pulmonary disease (COPD).
In certain embodiments, the proliferative disease is an infectious disease. In certain embodiments, the infectious disease is fungal infection. In certain embodiments, the infectious disease is a superficial mycose (e.g., Tinea versicolor). In certain embodiments, the infectious disease is a cutaneous mycose (e.g., epidermis). In certain embodiments, the infectious disease is a subcutaneous mycose. In certain embodiments, the infectious disease is a systemic mycose.
In certain embodiments, the proliferative disease is leukemia, adult T-cell leukemia, promyelocytic leukemia, pre-B cell leukemia, lymphoma, Mantle cell lymphoma, breast cancer, pancreatic cancer, prostate cancer, head and neck cancer, ovarian cancer, melanoma, glioma, liver cancer, renal cancer, colorectal cancer, rhabdomyosarcoma, tongue cancer, stomach cancer, multiple myeloma, bladder cancer, thyroid cancer, epidermoid carcinoma, lung cancer, NSC lung cancer, or large cell lung cancer.
In certain embodiments, the proliferative disease is adult T-cell leukemia, promyelocytic leukemia, pre-B cell leukemia, lymphoma, mantle cell lymphoma, pancreatic cancer, prostate cancer, head and neck cancer, ovarian cancer, melanoma, glioma, liver cancer, renal cancer, colorectal cancer, rhabdomyosarcoma, tongue cancer, stomach cancer, multiple myeloma, bladder cancer, thyroid cancer, epidermoid carcinoma, NSC lung cancer, or large cell lung cancer.
In certain embodiments, the proliferative disease is leukemia, adult T-cell leukemia, promyelocytic leukemia, pre-B cell leukemia, lymphoma, mantle cell lymphoma, breast cancer, head and neck cancer, ovarian cancer, colorectal cancer, tongue cancer, multiple myeloma, or large cell lung cancer.
In an embodiment, the cancers which can be treated by the compounds described herein include, but are not limited to, Acute Lymphoblastic Leukemia; Acute Myeloid Leukemia; Adrenocortical Carcinoma; AIDS-Related Lymphoma; AIDS-Related Malignancies; Anal Cancer; Astrocytoma; Bile Duct Cancer; Bladder Cancer; Bone Cancer, Osteosarcoma/Malignant Fibrous Histiocytoma; Brain Stem Glioma; Brain Tumor, Cerebellar Astrocytoma; Brain Tumor, Cerebral Astrocytoma/Malignant Glioma; Brain Tumor, Ependymoma; Brain Tumor, Medulloblastoma; Brain Tumor, Supratentorial Primitive Neuroectodermal Tumors; Brain Tumor, Visual Pathway and Hypothalamic Glioma; Breast Cancer; Bronchial Adenomas/Carcinoids; Carcinoid Tumor; Carcinoid Tumor, Gastrointestinal; Carcinoma, Adrenocortical; Carcinoma, Islet Cell; Central Nervous System Lymphoma, Primary; Cerebral Astrocytoma/Malignant Glioma; Cervical Cancer; Chronic Lymphocytic Leukemia; Chronic Myelogenous Leukemia; Chronic Myeloproliferative Disorders; Clear Cell Sarcoma of Tendon Sheaths; Colon Cancer; Colorectal Cancer; Cutaneous T-Cell Lymphoma; Endometrial Cancer; Ependymoma; Epithelial Cancer, Ovarian; Esophageal Cancer; Esophageal Cancer; Ewing's Family of Tumors; Extracranial Germ Cell Tumor; Extrahepatic Bile Duct Cancer; Eye Cancer, Intraocular Melanoma; Eye Cancer, Retinoblastoma; Gallbladder Cancer; Gastric (Stomach) Cancer; Gastrointestinal Carcinoid Tumor; Germ Cell Tumor, Extracranial, Childhood; Germ Cell Tumor, Extragonadal; Germ Cell Tumor, Ovarian; Gestational Trophoblastic Tumor; Glioma, Childhood Brain Stem; Glioma, Childhood Visual Pathway and Hypothalamic; Hairy Cell Leukemia; Head and Neck Cancer; Hepatocellular (Liver) Cancer; Hodgkin's Lymphoma; Hypopharyngeal Cancer; Hypothalamic and Visual Pathway Glioma; Intraocular Melanoma; Islet Cell Carcinoma (Endocrine Pancreas); Kaposi's Sarcoma; Kidney Cancer; Laryngeal Cancer; Leukemia, Acute Lymphoblastic; Leukemia, Acute Myeloid; Leukemia, Chronic Lymphocytic; Leukemia, Chronic Myelogenous; Leukemia, Hairy Cell; Lip and Oral Cavity Cancer; Liver Cancer; Lung Cancer, Non-Small Cell; Lung Cancer, Small Cell; Lymphoblastic Leukemia; Lymphoma, AIDS- Related; Lymphoma, Central Nervous System (Primary); Lymphoma, Cutaneous T-Cell; Lymphoma, Hodgkin's; Lymphoma, Hodgkin's During Pregnancy; Lymphoma, Non-Hodgkin's; Lymphoma, Primary Central Nervous System; Macroglobulinemia, Waldenstrom's; Male Breast Cancer; Malignant Mesothelioma; Malignant Thymoma; Medulloblastoma, Childhood; Melanoma; Melanoma, Intraocular; Merkel Cell Carcinoma; Mesothelioma, Malignant; Metastatic Squamous Neck Cancer with Occult Primary; Multiple Endocrine Neoplasia Syndrome, Childhood; Multiple Myeloma/Plasma Cell Neoplasm; Mycosis Fungoides; Myelodysplastic Syndromes; Myelogenous Leukemia, Chronic; Myeloid Leukemia; Myeloma, Multiple; Myeloproliferative Disorders, Chronic; Nasal Cavity and Paranasal Sinus Cancer; Nasopharyngeal Cancer; Neuroblastoma; Non-Hodgkin's Lymphoma; Non-Small Cell Lung Cancer; Oral Cancer; Oral Cavity and Lip Cancer; Oropharyngeal Cancer; steosarcoma/Malignant Fibrous Histiocytoma of Bone; Ovarian Epithelial Cancer; Ovarian Germ Cell Tumor; Ovarian Low Malignant Potential Tumor; Pancreatic Cancer; Paranasal Sinus and Nasal Cavity Cancer; Parathyroid Cancer; Penile Cancer; Pheochromocytoma; Pineal and Supratentorial Primitive Neuroectodermal Tumors; Pituitary Tumor; Plasma Cell Neoplasm/Multiple Myeloma; Pleuropulmonary Blastoma; Pregnancy and Breast Cancer; Pregnancy and Hodgkin's Lymphoma; Pregnancy and Non-Hodgkin's Lymphoma; Primary Central Nervous System Lymphoma; Primary Liver Cancer; Prostate Cancer; Rectal Cancer; Renal Cell (Kidney) Cancer; Renal Pelvis and Ureter, Transitional Cell Cancer; Retinoblastoma; Rhabdomyosarcoma; Salivary Gland Cancer; Sarcoma, Ewing's Family of Tumors; Sarcoma, Kaposi's; Sarcoma (Osteosarcoma)/Malignant Fibrous Histiocytoma of Bone; Sarcoma, Soft Tissue; Sezary Syndrome; Skin Cancer; Skin Cancer (Melanoma); Skin Carcinoma, Merkel Cell; Small Cell Lung Cancer; Small Intestine Cancer; Soft Tissue Sarcoma; Squamous Neck Cancer with Occult Primary, Metastatic; Stomach (Gastric) Cancer; Supratentorial Primitive Neuroectodermal Tumors; T- Cell Lymphoma, Cutaneous; Testicular Cancer; Thymoma, Malignant; Thyroid Cancer; Transitional Cell Cancer of the Renal Pelvis and Ureter; Trophoblastic Tumor, Gestational; Ureter and Renal Pelvis, Transitional Cell Cancer; Urethral Cancer; Uterine Sarcoma; Vaginal Cancer; Visual Pathway and Hypothalamic Glioma; Vulvar Cancer; Waldenstrom's Macro globulinemia; and Wilms' Tumor.
In one embodiment, a cancer potentially associated with mutant IDH enzyme activity is brain cancer, such as an astrocytic tumor (e.g., pilocytic astrocytoma, subependymal giant-cell astrocytoma, diffuse astrocytoma, pleomorphic xanthoastrocytoma, anaplastic astrocytoma, astrocytoma, giant cell glioblastoma, glioblastoma, secondary glioblastoma, primary adult glioblastoma, and primary pediatric glioblastoma); oligodendroglial tumor (e.g., oligodendroglioma, and anaplastic oligodendroglioma); oligoastrocytic tumor (e.g., oligoastrocytoma, and anaplastic oligoastrocytoma); ependymoma (e.g., myxopapillary ependymoma, and anaplastic ependymoma); medulloblastoma; primitive neuroectodermal tumor, schwannoma, meningioma, meatypical meningioma, anaplastic meningioma; and pituitary adenoma. In another embodiment, the brain cancer is glioma, glioblastoma multiforme, paraganglioma, or suprantentorial primordial neuroectodermal tumors (sPNET). In another embodiment, a cancer potentially associated with mutant IDH enzyme activity is leukemia, such as acute myeloid leukemia (AML), myelodysplastic syndrome (MDS), chronic myelogenous leukemia (CML), myeloproliferative neoplasm (MPN), post-MPN AML, post-MDS AML, del(5q)-associated high risk MDS or AML, blast-phase chronic myelogenous leukemia, angioimmunoblastic lymphoma and acute lymphoblastic leukemia.
In one embodiment, a cancer potentially associated with mutant IDH enzyme activity is skin cancer, including melanoma. In another embodiment, a cancer potentially associated with mutant IDH enzyme activity is prostate cancer, breast cancer, thyroid cancer, colon cancer, or lung cancer. In another embodiment, a cancer potentially associated with mutant IDH enzyme activity is sarcoma, including central chondrosarcoma, central and periosteal chondroma, and fibrosarcoma. In another embodiment, a cancer potentially associated with mutant IDH enzyme activity is cholangiocarcinoma.
Also provided are compounds described herein for use in the treatments described herein. Also provided are uses of compounds described herein for the manufacture of medicaments for the treatments described herein.
In certain embodiments, the subject to be treated with one of the methods provided herein has not been treated with anticancer therapy for the proliferative disease to be treated prior to the administration of a compound provided herein, e.g., a compound of Formulas I, II, or Ila, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof
In certain embodiments, the subject to be treated with one of the methods provided herein has been treated with anticancer therapy for the proliferative disease to be treated prior to the administration of a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof
In certain embodiments, the subject to be treated with one of the methods provided herein has developed drug resistance to the anticancer therapy. The methods provided herein encompass treating a subject regardless of patient's age, although some diseases or disorders are more common in certain age groups. Further provided herein is a method for treating a subject who has undergone surgery in an attempt to treat the disease or condition at issue, as well as the one who have not. Because the subjects with cancer have heterogeneous clinical manifestations and varying clinical outcomes, the treatment given to a particular subject may vary, depending on his/her prognosis. The skilled clinician will be able to readily determine without undue experimentation, specific secondary agents, types of surgery, and types of non-drug based standard therapy that can be effectively used to treat an individual subject with cancer.
Depending on the disease to be treated and the subject's condition, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, CIV, intracistemal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration. A compound provided herein, e.g., an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, may be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable excipients, carriers, adjuvants and vehicles, appropriate for each route of administration.
In one embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered orally. In another embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered parenterally. In yet another embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered intravenously. In yet another embodiment, a compound provided herein, e.g., a compound of a diastereomer, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered intramuscularly. In yet another embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered subcutaneously. In still another embodiment, a compound provided herein, e.g., a compound of Formulas I, Ha, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administered topically.
A compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, can be delivered as a single dose such as, e.g., a single bolus injection, or oral tablets or pills; or over time such as, e.g., continuous infusion over time or divided bolus doses over time. The compound provided herein can be administered repetitively if necessary, for example, until the patient experiences stable disease or regression, or until the patient experiences disease progression or unacceptable toxicity. For example, stable disease for solid tumors generally means that the perpendicular diameter of measurable lesions has not increased by 25% or more from the last measurement. Response Evaluation Criteria in Solid Tumors (RECIST) Guidelines, Journal of the National Cancer Institute 92(3): 205-216 (2000). Stable disease or lack thereof is determined by methods known in the art such as evaluation of patient symptoms, physical examination, visualization of the tumor that has been imaged using X-ray, CAT, PET, or MRI scan and other commonly accepted evaluation modalities. A compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, can be administered once daily (QD), or divided into multiple daily doses such as twice daily (BID), and three times daily (TID). In addition, the administration can be continuous, i.e., every day, or intermittently. The term "intermittent" or "intermittently" as used herein is intended to mean stopping and starting at either regular or irregular intervals. For example, intermittent administration of a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is administration for one to six days per week, administration in cycles {e.g., daily administration for two to eight consecutive weeks, then a rest period with no administration for up to one week), or administration on alternate days.
In certain embodiments, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, is cyclically administered to a patient. Cycling therapy involves the administration of an active agent for a period of time, followed by a rest for a period of time, and repeating this sequential administration. Cycling therapy can reduce the development of resistance to one or more of the therapies, avoid or reduce the side effects of one of the therapies, and/or improves the efficacy of the treatment.
In one embodiment is a combination comprising an effective amount of a compound as provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, or a tautomeric, or polymorphic form thereof and one, two, three or more other therapeutic agents, e.g. anti-cancer agents.
A compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof, can also be combined or used in combination with other therapeutic agents useful in the treatment and/or prevention of a disease described herein. As used herein, the term "in combination" includes the use of more than one therapy (e.g., one or more prophylactic and/or therapeutic agents). However, the use of the term "in combination" does not restrict the order in which therapies (e.g., prophylactic and/or therapeutic agents) are administered to a subject with a disease or disorder. A first therapy (e.g., a prophylactic or therapeutic agent such as a compound provided herein) can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapy (e.g., a prophylactic or therapeutic agent) to the subject. Triple therapy is also contemplated herein.
The route of administration of a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is independent of the route of administration of a second therapy. In one embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered orally. In another embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered intravenously. Thus, in accordance with these embodiments, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered orally or intravenously, and the second therapy can be administered orally, parenterally, intraperitoneally, intravenously, intraarterially, transdermally, sublingually, intramuscularly, rectally, transbuccally, intranasally, liposomally, via inhalation, vaginally, intraoccularly, via local delivery by catheter or stent, subcutaneously, intraadiposally, intraarticularly, intrathecally, or in a slow release dosage form. In one embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, and a second therapy are administered by the same mode of administration, orally or by IV. In another embodiment, a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, is administered by one mode of administration, e.g., by IV, whereas the second agent (an anticancer agent) is administered by another mode of administration, e.g., orally.
In certain embodiments, each method provided herein may independently, further comprise the step of administering a second therapeutic agent. In one embodiment, the second therapeutic agent is an anticancer agent. In another embodiment, the anticancer agent is an antimetabolite, including, but not limited to, 5-fluoro uracil, methotrexate, cytarabine (also known as cytosine arabinoside or Ara-C), and HDAC (high dose cytarabine) and fludarabine. In yet another embodiment, the anticancer agent is an antimicrotubule agent, including, but not limited to, vinca alkaloids (e.g., vincristine and vinblastine) taxanes (e.g., paclitaxel and docetaxel), and epothilones and their derivatives (e.g., ixabepilone). In yet another embodiment, the anticancer agent is an alkylating agent, including, but not limited to, cyclophosphamide, melphalan, carmustine, and nitrosoureas (e.g., bischloroethylnitrosurea and hydroxyurea). In yet another embodiment, the anticancer agent is a platinum agent, including, but not limited to, cisplatin, carbop latin, oxalip latin, satraplatin (JM-216), and CI-973. In yet another embodiment, the anticancer agent is an anthracycline, including, but not limited to, doxrubicin and daunorubicin. In yet another embodiment, the anticancer agent is an antitumor antibiotic, including, but not limited to, mitomycin, idarubicin, adriamycin, and daunomycin (also known as daunorubicin). In yet another embodiment, the anticancer agent is a topoisomerase inhibitor, e.g., etoposide and camptothecins. In yet another embodiment, the anticancer agent is selected from the group consisting of adriamycin, busulfan, cytarabine, cyclophosphamide, dexamethasone, fludarabine, fluorouracil, hydroxyurea, interferons, oblimersen, platinum derivatives, taxol, topotecan, and vincristine.
In another embodiment, the anticancer agent is a Bcr-Abl kinase inhibitor. In one embodiment, the Bcr-Abl kinase inhibitor is imatinib, BMS354825 (dasatinib), AMN107 (nilotinib), AP23464, AZD0530, CGP76030, ON012380, INN-0406 (NS-187), SKI-606 (bosutimb), VX- 680, or pyrrolo[2,3-d]pyrimidines including PD166326, PD173955 and PD180970. In another embodiment, the Bcr-Abl kinase inhibitor is imatinib. In yet another embodiment, the Bcr-Abl kinase inhibitor is dasatinib. In yet another embodiment, the Bcr-Abl kinase inhibitor is nilotinib. In yet another embodiment, the Bcr-Abl kinase inhibitor is AP23464. In yet another embodiment, the Bcr-Abl kinase inhibitor is AZD0530. In yet another embodiment, the Bcr-Abl kinase inhibitor is CGP76030. In yet another embodiment, the Bcr-Abl kinase inhibitor is SKI- 606. In yet another embodiment, the Bcr-Abl kinase inhibitor is ON012380. In yet another embodiment, the Bcr-Abl kinase inhibitor is INN-0406 (NS-187). In yet another embodiment, the Bcr-Abl kinase inhibitor is a pyrrolo[2,3-d]pyrimidine. In another embodiment, the Bcr-Abl kinase inhibitor is VX-680. In another embodiment, the Bcr-Abl kinase inhibitor is PD166326. In yet another embodiment, the Bcr-Abl kinase inhibitor is PD173955. In still another embodiment, the Bcr-Abl kinase inhibitor is PD 180970.
In still another embodiment, the anticancer agent is a FLT3 kinase inhibitor. In one embodiment, the FLT3 kinase inhibitor is PKC 412, MLN 578, CEP-701, CT 53518, CT-53608, CT-52923, D-64406, D-65476, AGL-2033, AG1295, AG1296, KN-1022, PKC-412, SU5416, SU5614, SU11248, L-00021649, or CHIR-258. In another embodiment, the FLT3 kinase inhibitor is PKC 412. In yet another embodiment, the FLT3 kinase inhibitor is MLN 578. In yet another embodiment, the FLT3 kinase inhibitor is CEP-701. In yet another embodiment, the FLT3 kinase inhibitor is CT 53518. In yet another embodiment, the FLT3 kinase inhibitor is CT- 53608. In yet another embodiment, the FLT3 kinase inhibitor is CT-52923. In yet another embodiment, the FLT3 kinase inhibitor is D-64406. In yet another embodiment, the FLT3 kinase inhibitor is D-65476. In yet another embodiment, the FLT3 kinase inhibitor is AGL-2033. In yet another embodiment, the FLT3 kinase inhibitor is AG1295. In yet another embodiment, the FLT3 kinase inhibitor is AG1296. In yet another embodiment, the FLT3 kinase inhibitor is KN-1022. In yet another embodiment, the FLT3 kinase inhibitor is KN-1022. In yet another embodiment, the FLT3 kinase inhibitor is SU5416. In yet another embodiment, the FLT3 kinase inhibitor is SU5614. In yet another embodiment, the FLT3 kinase inhibitor is SU1 1248. In yet another embodiment, the FLT3 kinase inhibitor is L-00021649. In still another embodiment, the FLT3 kinase inhibitor is CFIIR-258.
Other therapies or anticancer agents that may be used in combination with a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or He, including an enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, include surgery, radiotherapy (e.g., gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes), endocrine therapy, biologic response modifiers (e.g., interferons, interleukins, and tumor necrosis factor (TNF)), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, alkylating drugs (mechlorethamine, chlorambucil, cyclophosphamide, melphalan, and ifosfamide), antimetabolites (cytarabine (also known as cytosine arabinoside or Ara-C), HDAC (high dose cytarabine), and methotrexate), purine antagonists and pyrimidine antagonists (6-mercaptopurine, 5-fluorouracil, cytarbine, and gemcitabine), spindle poisons (vinblastine, vincristine, vinorelbine, and paclitaxel), podophyllotoxins (etoposide, irinotecan, and topotecan), antibiotics (daunorubicin, doxorubicin, bleomycin, and mitomycin), nitrosoureas (carmustine and lomustine), inorganic ions (cisplatin and carboplatin), enzymes (asparaginase), and hormones (tamoxifen, leuprolide, flutamide, and megestrol), imatinib, adriamycin, dexamethasone, and cyclophosphamide. For a more comprehensive discussion of updated cancer therapies see, http://www.nci.nih.gov/, a list of the FDA approved oncology drugs at http://www.fda.gov, and The Merck Manual, Seventeenth Ed. 1999, the entire contents of which are hereby incorporated by reference.
In one embodiment, the other anticancer agent is selected from the group consisting of vascular endothelial growth factor (VEGF) receptor inhibitors, topoisomerase II inhibitors, smoothen inhibitors, alkylating agents, anti-tumor antibiotics, anti-metabolites, retinoids, immunomodulatory agents including but not limited to anti-cancer vaccines, CTLA-4, LAG-3, PD-1 antagonists and BET bromodomain inhibitors.
Examples of vascular endothelial growth factor (VEGF) receptor inhibitors include, but are not limited to, bevacizumab (sold under the trademark AVASTIN by Genentech/Roche), axitinib, (N-methyl-2-[[3-[([pound])-2-pyridin-2-ylethenyl]-l H-indazol-6- yl]sulfanyl]benzamide, also known as AG013736, and described in PCT Publication No. WO 01 /002369), Brivanib Alaninate ((S)-((R)-l-(4-(4-Fluoro-2-methyl-lH-indol-5-yloxy)-5- methylpyrrolo[2,l-f [l,2,4]triazin-6-yloxy)propan-2-yl)2-aminopropanoate, also known as BMS- 582664), motesanib (N-(2,3-dihydro-3,3-dimethyl-l H-indoi-6-y!)-2-[(4- pyridinyimethyj)amino]-3-pyfidinecarboxamide. and described in PCT Publication No. WO 02/068470), pasireotide (also known as SO 230, and described in PCT Publication No. WO 02/010192), and sorafenib (sold under the tradename NEXAVAR).
Examples of topoisomerase II inhibitors, include but are not limited to, etoposide (also known as VP-16 and Etoposide phosphate, sold under the tradenames TOPOSAR, VEPESID and ETOPOPHOS), and teniposide (also known as VM-26, sold under the tradename VUMON).
Examples of alkylating agents, include but are not limited to, 5-azacytidine (sold under the trade name VIDAZA), decitabine (sold under the trade name of DECOGEN), temozolomide (sold under the trade names TEMODAR and TEMODAL by Schering- Plough/Merck), dactinomycin (also known as actinomycin-D and sold under the tradename COSMEGEN), melphalan (also known as L-PAM, L-sarcolysin, and phenylalanine mustard, sold under the tradename ALKERAN), altretamine (also known as hexamethylmelamine (HMM), sold under the tradename HEXALEN), carmustine (sold under the tradename BCNU), bendamustine (sold under the tradename TREANDA), busulfan (sold under the tradenames Busulfex(R) and Myleran(R)), carboplatin (sold under the tradename Paraplatin(R)), lomustine (also known as CCNU, sold under the tradename CeeNU(R)), cisplatin (also known as CDDP, sold under the tradenames Platinol(R) and Platinol(R)-AQ), chlorambucil (sold under the tradename Leukeran(R)), cyclophosphamide (sold under the tradenames Cytoxan(R) and Neosar(R)), dacarbazine (also known as DTIC, DIC and imidazole carboxamide, sold under the tradename DTIC-Dome(R)), altretamine (also known as hexamethylmelamine (HMM) sold under the tradename Hexalen(R)), ifosfamide (sold under the tradename Ifex(R)), procarbazine (sold under the tradename Matulane(R)), mechlorethamine (also known as nitrogen mustard, mustine and mechloroethamine hydrochloride, sold under the tradename Mustargen(R)), streptozocin (sold under the tradename Zanosar(R)), thiotepa (also known as thiophosphoamide, TESPA and TSPA, and sold under the tradename Thioplex(R). Examples of anti-tumor antibiotics include, but are not limited to, doxorubicin (sold under the tradenames Adriamycin(R) and Rubex(R)), bleomycin (sold under the tradename lenoxane(R)), daunorubicin (also known as dauorubicin hydrochloride, daunomycin, and rubidomycin hydrochloride, sold under the tradename Cerubidine(R)), daunorubicin liposomal (daunorubicin citrate liposome, sold under the tradename DaunoXome(R)), mitoxantrone (also known as DHAD, sold under the tradename Novantrone(R)), epirubicin (sold under the tradename Ellence(TM)), idarubicin (sold under the tradenames Idamycin(R), Idamycin PFS(R)), and mitomycin C (sold under the tradename Mutamycin(R)).
Examples of anti-metabolites include, but are not limited to, claribine (2- chlorodeoxyadenosine, sold under the tradename leustatin(R)), 5-fluorouracil (sold under the tradename Adrucil(R)), 6-thioguanine (sold under the tradename Purinethol(R)), pemetrexed (sold under the tradename Alimta(R)), cytarabine (also known as arabinosylcytosine (Ara-C), sold under the tradename Cytosar-U(R)), cytarabine liposomal (also known as Liposomal Ara-C, sold under the tradename DepoCyt(TM)), decitabine (sold under the tradename Dacogen(R)), hydroxyurea (sold under the tradenames Hydrea(R), Droxia(TM) and Mylocel(TM)), fludarabine (sold under the tradename Fludara(R)), floxuridine (sold under the tradename FUDR(R)), cladribine (also known as 2-chlorodeoxyadenosine (2-CdA) sold under the tradename Leustatin(TM)), methotrexate (also known as amethopterin, methotrexate sodium (MTX), sold under the tradenames Rheumatrex(R) and Trexall(TM)), and pentostatin (sold under the tradename Nipent(R)).
Examples of retinoids include, but are not limited to, alitretinoin (sold under the tradename Panretin(R)), tretinoin (all-trans retinoic acid, also known as ATRA, sold under the tradename Vesanoid(R)), Isotretinoin (13-c/s-retinoic acid, sold under the tradenames Accutane(R), Amnesteem(R), Claravis(R), Clarus(R), Decutan(R), Isotane(R), Izotech(R), Oratane(R), Isotret(R), and Sotret(R)), and bexarotene (sold under the tradename Targretin(R)).
"PD-1 antagonist" means any chemical compound or biological molecule that blocks binding of PD-L1 expressed on a cancer cell to PD-1 expressed on an immune cell (T cell, B cell or NKT cell) and preferably also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1. Alternative names or synonyms for PD-1 and its ligands include: PDCD1, PD1, CD279 and SLEB2 for PD-1 ; PDCD1L1, PDL1, B7H1, B7-4, CD274 and B7-H for PD-L1 ; and PDCD1L2, PDL2, B7-DC, Btdc and CD273 for PD-L2. In any of the treatment method, medicaments and uses of the present invention in which a human individual is being treated, the PD-1 antagonist blocks binding of human PD-Ll to human PD-1, and preferably blocks binding of both human PD-Ll and PD-L2 to human PD-1. Human PD-1 amino acid sequences can be found in NCBI Locus No. : NP 005009. Human PD-Ll and PD-L2 amino acid sequences can be found in NCBI Locus No.: NP_054862 and NP_079515, respectively.
PD-1 antagonists useful in any of the treatment method, medicaments and uses of the present invention include a monoclonal antibody (mAb), or antigen binding fragment thereof, which specifically binds to PD-1 or PD-Ll, and preferably specifically binds to human PD-1 or human PD-Ll . The mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region. In some embodiments the human constant region is selected from the group consisting of IgGl, IgG2, IgG3 and IgG4 constant regions, and in preferred embodiments, the human constant region is an IgGl or IgG4 constant region. In some embodiments, the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab')2, scFv and Fv fragments.
Examples of mAbs that bind to human PD-1, and useful in the treatment method, medicaments and uses of the present invention, are described in US7488802, US7521051, US8008449, US8354509, US8168757, WO2004/004771, WO2004/072286, WO2004/056875, and US2011/0271358.
Examples of mAbs that bind to human PD-Ll , and useful in the treatment method, medicaments and uses of the present invention, are described in WO2013/019906, W02010/077634 Al and US8383796. Specific anti-human PD-Ll mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include MPDL3280A, BMS-936559, MEDI4736, MSB0010718C and an antibody which comprises the heavy chain and light chain variable regions of SEQ ID NO: 24 and SEQ ID NO: 21, respectively, of WO2013/019906.
Other PD-1 antagonists useful in any of the treatment method, medicaments and uses of the present invention include an immunoadhesin that specifically binds to PD-1 or PD- Ll, and preferably specifically binds to human PD-1 or human PD-Ll, e.g., a fusion protein containing the extracellular or PD-1 binding portion of PD-Ll or PD-L2 fused to a constant region such as an Fc region of an immunoglobulin molecule. Examples of immunoadhesion molecules that specifically bind to PD-1 are described in WO2010/027827 and WO2011/066342. Specific fusion proteins useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include AMP -224 (also known as B7- DCIg), which is a PD-L2-FC fusion protein and binds to human PD-1.
Examples of other cytotoxic agents include, but are not limited to, arsenic trioxide
(sold under the tradename Trisenox(R)), asparaginase (also known as L-asparaginase, and Erwinia L-asparaginase, sold under the tradenames Elspar(R) and Kidrolase(R)).
In an embodiment, the other anticancer agent is a BET bromodomain inhibitor. Examples of BET bromodomain inhibitor include the compounds described in U.S. Patent No. 5712274, WO1994006802, U.S. Patent No. 8476260 and WO2009/084693.
The compounds provided herein can also be provided as an article of manufacture using packaging materials well known to those of skill in the art. See, e.g., U.S. Pat. Nos. 5,323,907; 5,052,558; and 5,033,252. Examples of pharmaceutical packaging materials include, but are not limited to, blister packs, bottles, tubes, inhalers, pumps, bags, vials, containers, syringes, and any packaging material suitable for a selected formulation and intended mode of administration and treatment.
Kits
In certain embodiments, provided herein also are kits which, when used by the medical practitioner, can simplify the administration of appropriate amounts of active ingredients to a subject. In certain embodiments, the kit provided herein includes a container and a dosage form of a compound or composition provided herein, including a single enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof.
In certain embodiments, the kit includes a container comprising a dosage form of the compound provided herein, including a single enantiomer, a mixture of enantiomers, a diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, or solvate thereof, in a container comprising one or more other therapeutic agent(s) described herein.
Kits provided herein can further include devices that are used to administer the active ingredients. Examples of such devices include, but are not limited to, syringes, needle-less injectors drip bags, patches, and inhalers. The kits provided herein can also include condoms for administration of the active ingredients.
Kits provided herein can further include pharmaceutically acceptable vehicles that can be used to administer one or more active ingredients. For example, if an active ingredient is provided in a solid form that must be reconstituted for parenteral administration, the kit can comprise a sealed container of a suitable vehicle in which the active ingredient can be dissolved to form a particulate-free sterile solution that is suitable for parenteral administration. Examples of pharmaceutically acceptable vehicles include, but are not limited to: aqueous vehicles, including, but not limited to, Water for Injection USP, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles, including, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles, including, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate.
In certain embodiments, provided herein is a method of inhibiting the growth of a cell, comprising the step of contacting the cell with a compound provided herein, e.g., a compound of Formulas I, Ila, lib, or lie, including an enantiomer, a mixture of enantiomers, a diastereomer, a mixture of two or more diastereomers, a tautomer, a mixture of two or more tautomers, or an isotopic variant thereof; or a pharmaceutically acceptable salt, solvate, or hydrate thereof.
In certain embodiment, the effective amount of the compound provided herein ranges from about 1 pM to about 1 mM, from about 10 pM to about 10 μΜ, from about 100 pM to about 2 μΜ, or from about 1 nM to about 1 μΜ.
In certain embodiments, the cell is a mammalian cell. In certain embodiments, the mammal cell is a human cell. In certain embodiment, the cell is a tumor cell. In certain embodiment, the cell is a mammalian tumor cell. In certain embodiment, the cell is a human tumor cell. In certain embodiment, the cell is a cancerous cell. In certain embodiment, the cell is a mammalian cancerous cell. In certain embodiment, the cell is a human cancerous cell. In certain embodiment, the cancerous cell is a metastatic cancerous cell. In certain embodiment, the cancerous cell is a metastatic microbial cell. In certain embodiment, the cancerous cell is a metastatic bacterial cell. In certain embodiment, the cancerous cell is a metastatic fungal cell. In certain embodiment, the cell is a hematologic malignancy cell. In certain embodiment, the cell is a leukemia cell. In certain embodiments, the cell is a relapsed leukemia cell. In certain embodiments, the cell is a refractory leukemia cell. In certain embodiments, the cell is a drug- resistant leukemia cell. In certain embodiments, the cell is a multidrug-resistant leukemia cell. In certain embodiments, the cell is a Bcr-Abl kinase inhibitor-resistant leukemia cell. In certain embodiments, the cell is an imatinib-resistant leukemia cell. In certain embodiments, the cell is a dasatinib-resistant leukemia cell. In certain embodiments, the cell is a nilatinib-resistant leukemia cell. In certain embodiments, the cell is a bosutinib-resistant leukemia cell. In certain embodiments, the cell is a cytarabine-resistant leukemia cell.
In certain embodiment, the cell is a leukemia stem cell. In certain embodiments, the cell is a relapsed leukemia stem cell. In certain embodiments, the cell is a refractory leukemia stem cell. In certain embodiments, the cell is a drug-resistant leukemia stem cell. In certain embodiments, the cell is a multidrug-resistant leukemia stem cell. In certain embodiments, the cell is a Bcr-Abl kinase inhibitor-resistant leukemia stem cell. In certain embodiments, the cell is an imatinib- resistant leukemia stem cell. In certain embodiments, the cell is a dasatinib-resistant leukemia stem cell. In certain embodiments, the cell is a nilatinib-resistant leukemia stem cell. In certain embodiments, the cell is a bosutinib-resistant leukemia stem cell. In certain embodiments, the cell is a cytarabine-resistant leukemia stem cell.
In certain embodiment, the cell is an acute leukemia cell. In certain embodiments, the cell is a relapsed acute leukemia cell. In certain embodiments, the cell is a refractory acute leukemia cell. In certain embodiments, the cell is a drug-resistant acute leukemia cell. In certain embodiments, the cell is a multidrug-resistant acute leukemia cell. In certain embodiments, the cell is a Bcr- Abl kinase inhibitor-resistant acute leukemia cell. In certain embodiments, the cell is an imatinib-resistant acute leukemia cell. In certain embodiments, the cell is a dasatinib-resistant acute leukemia cell. In certain embodiments, the cell is a nilatinib-resistant acute leukemia cell. In certain embodiments, the cell is a bosutinib-resistant acute leukemia cell. In certain embodiments, the cell is a cytarabine-resistant acute leukemia cell.
In certain embodiments, the cell is an ALL cell. In certain embodiments, the cell is a relapsed ALL cell. In certain embodiments, the cell is a refractory ALL cell. In certain embodiments, the cell is a drug-resistant ALL cell. In certain embodiments, the cell is a multidrug-resistant ALL cell. In certain embodiments, the cell is a Bcr-Abl kinase inhibitor-resistant ALL cell. In certain embodiments, the cell is an imatinib-resistant ALL cell. In certain embodiments, the cell is a dasatinib-resistant ALL cell. In certain embodiments, the cell is a nilatinib-resistant ALL cell. In certain embodiments, the cell is a bosutinib-resistant ALL cell. In certain embodiments, the cell is a cytarabine-resistant ALL cell.
In certain embodiments, the cell is an AML cell. In certain embodiments, the cell is a relapsed AML cell. In certain embodiments, the cell is a refractory AML cell. In certain embodiments, the cell is a drug-resistant AML cell. In certain embodiments, the cell is a multidrug-resistant AML cell. In certain embodiments, the cell is a Bcr-Abl kinase inhibitor-resistant AML cell. In certain embodiments, the cell is an imatinib-resistant AML cell. In certain embodiments, the cell is a dasatinib-resistant AML cell. In certain embodiments, the cell is a nilatinib-resistant AML cell. In certain embodiments, the cell is a bosutinib-resistant AML cell. In certain embodiments, the cell is a cytarabine-resistant AML cell.
In certain embodiments, the cell is a chronic leukemia cell. In certain embodiments, the cell is a relapsed chronic leukemia cell. In certain embodiments, the cell is a refractory chronic leukemia cell. In certain embodiments, the cell is a drug-resistant chronic leukemia cell. In certain embodiments, the cell is a multidrug-resistant chronic leukemia cell. In certain embodiments, the cell is a Bcr-Abl kinase inhibitor-resistant chronic leukemia cell. In certain embodiments, the cell is an imatinib-resistant chronic leukemia cell. In certain embodiments, the cell is a dasatinib-resistant chronic leukemia cell. In certain embodiments, the cell is a nilatinib-resistant chronic leukemia cell. In certain embodiments, the cell is a bosutinib-resistant chronic leukemia cell. In certain embodiments, the cell is a cytarabine-resistant chronic leukemia cell.
In certain embodiments, the cell is a CLL cell. In certain embodiments, the cell is a relapsed CLL cell. In certain embodiments, the cell is a refractory CLL cell. In certain embodiments, the cell is a drug-resistant CLL cell. In certain embodiments, the cell is a multidrug-resistant CLL cell. In certain embodiments, the cell is a Bcr-Abl kinase inhibitor-resistant CLL cell. In certain embodiments, the cell is an imatinib-resistant CLL cell. In certain embodiments, the cell is a dasatinib-resistant CLL cell. In certain embodiments, the cell is a nilatinib-resistant CLL cell. In certain embodiments, the cell is a bosutinib-resistant CLL cell. In certain embodiments, the cell is a cytarabine-resistant CLL cell. In certain embodiments, the cell is a CML cell. In certain embodiments, the cell is a relapsed CML cell. In certain embodiments, the cell is a refractory CML cell. In certain embodiments, the cell is a drug-resistant CML cell. In certain embodiments, the cell is a multidrug-resistant CML cell. In certain embodiments, the cell is a Bcr-Abl kinase inhibitor-resistant CML cell. In certain embodiments, the cell is an imatinib-resistant CML cell. In certain embodiments, the cell is a dasatinib-resistant CML cell. In certain embodiments, the cell is a nilatinib-resistant CML cell. In certain embodiments, the cell is a bosutinib-resistant CML cell. In certain embodiments, the cell is a cytarabine-resistant CML cell.
In certain embodiments, the cell is Philadelphia positive leukemia cell. In one embodiment, the cell is a Philadelphia positive ALL cell. In another embodiment, the cell is a Philadelphia positive AML cell. In yet another embodiment, the cell is a Philadelphia positive CLL cell. In still another embodiment, the cell is a Philadelphia positive CML cell.
The inhibition of cell growth can be gauged by, e.g., counting the number of cells contacted with a compound of interest, comparing the cell proliferation with otherwise identical cells not contacted with the compound, or determining the size of the tumor that encompasses the cells. The number of cells, as well as the size of the cells, can be readily assessed using any method known in the art (e.g., trypan blue exclusion and cell counting, measuring incorporation of H- thymidine into nascent DNA in a cell).
Assay Methods Patient samples were obtained from a non-interventional and prospective study. The study included samples from adult patients over 18 years of age who were diagnosed with Acute Myeloid Leukemia (AML).
In one embodiment, the method of data acquisition was performed as follows: on day 1 bone marrow (BM) or peripheral blood (PB) patient sample was received. A small part was separated from the rest of the sample for validation, while the majority of the sample was diluted with culture media and plated into 96-well plates previously prepared with the desired compounds and compound combinations, e.g., a compound or compound combination described herein. The number of live leukemic cells seeded in each well was fixed between 8000 and 32000, depending on the percentage of leukemic cells for each sample. These plates were incubated for 72 hours and analyzed on day 4. Antibodies were added to identify leukemic cells using a gating strategy based on FSC/SSC and expression or lack of expression of different surface markers. The monoclonal antibodies selection was performed to optimize the identification of leukemic cell in each sample.
In one embodiment, non-limiting examples of biomarkers such as the biomarkers CD34, CD45, CD 117, and HLADR known as "backbone markers" for AML (van Dongen, J.J. and A. Orfao, EuroFlow: Resetting leukemia and lymphoma immunophenotyping. Basis for companion diagnostics and personalized medicine, Leukemia, 2012, 26, 1899-907) were included in the combination.
In one embodiment, antibody combinations such as CD117/CD45, CD34/CD45, and HLADR CD45 were used. Live leukemic cells were identified by their light scatter properties (FSCint/hV SSCint) in the absence of Annexin-V-FITC staining. FSC/SSC selection was performed to exclude debris. The average percentage of cell viability upon receipt of the sample was 80% and samples were only processed if the viability was greater than 50%.
In one embodiment, sample validation was performed as follows: BM and PB samples were extracted under sterile conditions and received in the laboratory within 24 hours of extraction. Initial analysis evaluated the number of pathological cells and their viability. Different volumes of sample (1 μL, 3 μL, 5 μL and 7 μL) were aliquoted in duplicate into a 96-well plate. To lyse red blood cells, 180 μL of ammonium chloride lysis solution was added to each well (2g KHC03, 16.58g NH4CI, 0.074g Na2EDTA 2H20, H20 to 1L). Following a 10 mm incubation period at 4°C, plate was centrifuged for 5 min at 1200 rpm and the supernatant removed. The lysis step was performed twice. To analyze, 20 iL of a combination of Annexin V-FITC (Immunostep, Salamanca, Spain), binding buffer (BB, 2.4g HEPES, 8.19g NaCl, 0.37g Cl2Ca, H20 to 1L), and the following monoclonal antibodies (mAb) were added to each well: CD1 17 (clone 104D2)-PE (Becton Dickinson, San Jose, CA, US), CD34 (clone 581)-PerCP (BioLegend, San Diego, CA, US), HLADR (clone L243)-PB (BioLegend) and CD45 (ΓΠ30)- PO (Life Technologies, Carlsbad, CA, US) (van Dongen, J. J., et al., EuroFlow antibody panels or standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes, Leukemia, 2012, 26, 1908-75). After 15 min of incubation at room temperature in the dark, a wash step was performed using binding buffer solution. The pellet was resuspended in 30 μΕ BB for analysis in Vivia's ExviTech© platform. Cell count and viability upon arrival were computed and the optimal volume of sample to use per well was determined.
In one embodiment, the assay was performed according to following method: the whole sample was diluted with RPMI 1640, supplemented with 20% (v/v) FBS (Thermo Scientific, Waltham, MA, US), 2% HEPES, 1% antibiotic (Zell Shield, Labclimcs, Barcelona, Spain) and 1% L- glutamine 200mM (Lonza, Hopkinton, MA, US) to a final volume of 60 μL· per well. The mixture was dispensed into 96-well plates containing a compound described herein with a Multidrop Combi Smart (Thermo Scientific, Waltham, MA, US). Drug plates were previously prepared using an Echo 550 Liquid Handler (LabCyte, Sunnyvale, CA, US). Five or eight concentrations were used for each compound tested, adjusted to cover the range of activities across patients. The compounds tested were also tested against the corresponding parent as a control. The plates were incubated for 48 hours or 72 hours at 37°C in humidified air containing 5% C02.
In one embodiment, data analysis was conducted with Summit software (Beckman Coulter). Identification of pathological cells was performed using a gating strategy based on FSC/SSC and expression or lack of expression of the different mAb markers. Depletion was measured as the difference in the number of live cells in a well with the compounds described herein vs the control wells without the compounds. Annexin V was then used to exclude dying cells and measure only the number of live cells in the drug wells and in the control wells. Those cells without Annexin V staining and appropriate FSC/SSC were considered as live cells (Koopman, G., et ah, Annexin V for flow cytometric detection of phosphatidylserine expression on B cells undergoing apoptosis, Blood, 1994, 84, 1415-20). Using the above parameters, FCS Analyzer was used to determine the effect of each of the individual compound. Data were transfered to ActivityBase (IDBS, Guildford, UK) for final analysis.
The disclosure will be further understood by the following non-limiting examples.
The ex-vivo effect of the compounds on AML samples when the plates were incubated for 72 hours is illustrated in Table 2:
Compound EC50 abs. Emax (%)
(μΜ)
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
The ex-vivo effect of the compounds on AML samples when the plates were incubated for 48 hours is illustrated in Table 3 :
Compound EC50 abs. (μΜ) Emax (%)
D A
C C
Diastereomer 1
D A
Diastereomer 2
Diastereomer 1
Figure imgf000187_0001
Diastereomer 2 o >
Figure imgf000188_0001
EC50 values are provided as follows:
A < 1 μΜ, 1 < B < 10 μΜ, 10 < C < 20 μΜ, D > 20 μΜ;
Emax values are provided as follows:
A = 0-10%; B = 10-20%; C > 20%
In Vitro Inhibition
Materials
Cells were grown in RPMI-1640 supplemented with L-Glutamine and 10% FBS of the following cancer cell lines:
CCRF-CEM; CCRF-CEM-cytarabine resistant; HL-60;
Method
18, 96 well plates of each cell line were seeded with the optimized number of cells per well in a total volume of 50 μΕ per well. The plates were left overnight. Plate wells were seeded with 100 μΕ media for media control. The following day, cells were exposed to test compounds as described below. At the same time as drug exposure, a CTG assay was conducted on the 18th plate for the 0 hr count.
The compounds were added to cells and medium already on the plate to give desired final concentrations. 50 μΕ media were added to cell control wells, and 50 μΕ of mix added to vehicle control wells. 10 μΜ Doxorubicin was added to appropriate wells as control. Cells exposed to test compound were incubated at 37°C for 72 hr followed by a CTG assay.
CellTiter-Glo (CTG)
At the end of the 72 hr exposure period, plates were removed for a CellTiter-Glo (CTG) assay from a 37°C, 5% C02 incubator and placed on the bench at room temperature for 30 mins. 100 μΕ of CellTiter-Glo reagent was added and mixed for 2 mins, followed by a further 10 min incubation at room temperature. Luminescence was recorded using Synergy 4.0. All the exemplified compounds were tested in accordance with this method and were found to have IC50 values of less than ΙΟΟμΜ at each cell line. The results for compounds are provided in Table 4.
Table 4 -In vitro inhibition of CCRF-CEM; CCRF-CEM-cvtarabine resistant; and HL-60.
Figure imgf000190_0001
T e IC50 vaues n Ta es 1 are as o ows:
A = < 1 μΜ
B = > 1 and 10<μΜ C = > 10 and < 25 μΜ
D = > 25 and < 100 μΜ
The embodiments described above are intended to be merely exemplary, and those skilled in the art will recognize, or will be able to ascertain using no more than routine experimentation, numerous equivalents of specific compounds, materials, and procedures. All such equivalents are considered to be within the scope of the claimed subject matter and are encompassed by the appended claims.
Since modifications will be apparent to those of skill in the art, it is intended that the claimed subject matter be limited only by the scope of the appended claims.

Claims

What is claimed is:
1. A compound of Formula I :
Figure imgf000192_0001
p-N02PhCH20- — NHCH2Ph, -N(C¾)CH2Ph, -NHR2, -NH(CH2)5N(CH3)2, -BH3,
Figure imgf000192_0002
Figure imgf000193_0001
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
W is NH2, halo, OMe or OH;
T is NH2, F, CI or halogen;
R1 is hydrogen, halo, OH, protected OH, cyano or alkynyl; Q is OR3; E is CR4R5; L is H, p-Me, p-OMe, p-Cl, or 3,4-Cl; n is 1 ; m is 1 or 2; p is absent or O;
Ry is alkyl, alkenyl, alkynyl, alkoxycarbonyl, or hydroxyalkyl, each independently optionally substituted;
R2 is hydrogen or alkyl;
R is hydrogen, alkyl, alkenyl, alkynyl, alkylcarbonyl or aralkyl; R4 and R5 are selected from: i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl, or alkylheteroaryl, wherein the alkyl is optionally substituted by alkoxy; or ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring;
R is hydrogen,
Figure imgf000194_0001
, or
R6 with one of X and Y, together with the atoms to which they are attached, combine to form a six-membered heterocyclic ring where R6 and the one of X and Y together represent a single divalent -0-; or
R6 with one of X and Y, together with the atoms to which they are attached, combine to form a 7-12-membered heterocyclic ring; wherein, when R is
Figure imgf000195_0001
s Ϋ then at least one Ry is other than alkyl.
2. The compound of claim 1 , wherein R1 is OH or F.
3. The compound of claim 1 or claim 2, wherein:
O
U
X is -NHCH2PJ , Y is selected from OH v
Figure imgf000195_0002
4. The compound of any one of claims 1 or 2 having the Formula lid:
Figure imgf000195_0003
(lid)
or a pharmaceutically acceptable salt thereof
5. The compound of claim 1 or claim 2 having the Formula He:
Figure imgf000195_0004
(He) or a pharmaceutically acceptable salt thereof, wherein:
R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl, or cycloalkyl; or R4 is alkyl or cycloalkyl, and R5 is hydrogen.
6. The compound of claim 1 or claim 2 having the Formula Ilf:
Figure imgf000196_0001
or a pharmaceutically acceptable salt thereof, wherein:
R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl, or cycloalkyl; or R4 is alkyl or cycloalkyl, and R5 is hydrogen.
7. The compound of claim 1 or claim 2 having the Formula Ilg:
Figure imgf000196_0002
pharmaceutically acceptable salt thereof
The compound of claim 1 or claim 2 having the Formula Ilh:
Figure imgf000197_0001
(Eh)
or a pharmaceutically acceptable salt thereof, wherein:
R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl, or R4 is alkyl and R5 hydrogen.
9. The compound of claim 1 or claim 2 having one of the following Formulas:
Figure imgf000197_0002
10. The compound of claim 1 or claim 2 having one of the following Formulas:
Figure imgf000198_0001
Figure imgf000198_0002
The compound of claim 1 or claim 2 having one of the following Formulas: or
Figure imgf000199_0001
13. The compound of claim 1 or claim 2 having one of the following Formulas:
Figure imgf000199_0002
14. The compound of claim 1 or claim 2 having the Formula:
Figure imgf000200_0001
15. A compound of Formula Ila:
Figure imgf000200_0002
(Ila) or a pharmaceutically acceptable salt thereof, wherein
Figure imgf000200_0003
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid:
NH2, CI, or OMe;
T is NH2, F, or CI;
R1 is hydrogen, OH, or F; R2 is hydrogen or alkyl; Q is OR3; E is CR ; n is 1 ;
R3 is hydrogen, alkyl, alkenyl, or alkynyl; R4 and R5 are selected from: i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl, or alkylheteroaryl, wherein the alkyl is optionally substituted by alkoxy; or ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring;
R is alkyl, alkenyl, or alkynyl, each independently optionally substituted.
16. The compound of claim 15 having the Formula:
Figure imgf000201_0001
or a pharmaceutically acceptable salt thereof, wherein:
R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl, or cycloalkyl; or R4 is alkyl or cycloalkyl, and R5 is hydrogen.
17. The compound of any one of claims 15 or 16 having one of the following Formulas:
Figure imgf000202_0001
18. The compound of any one of claims 15 or 16 having one of the following Formulas:
Figure imgf000202_0002
19. A compound of Formula lib:
Figure imgf000202_0003
(lib) or a pharmaceutically acceptable salt thereof, wherein
YisOH,
Figure imgf000202_0004
,0rOR7;
Figure imgf000203_0001
Rz is H or C(0)Z where Z is a fatty acid chain selected from palmitoleic, oleic, linoleic, or arachidonic acid;
R2 is hydrogen or alkyl;
Q is OR3;
E is CR ; n is 1 ;
R is hydrogen, alkyl, alkenyl, alkynyl or aralkyl; R4 and R5 are selected from: i) R4 and R5 are each independently hydrogen, alkyl, alkenyl, alkynyl, aryl, aralkyl, cycloalkyl, cycloalkenyl, alkylheterocyclyl or alkylheteroaryl, wherein the alkyl is optionally substituted by alkoxy; or ii) R4 and R5 together with the carbon atom to which they are attached form a 3-7 membered cycloalkyl ring;
R7 is alkyl, alkenyl, or alkynyl, each independently optionally substituted.
20. The compound of claim 19 having the Formula:
Figure imgf000203_0002
or a pharmaceutically acceptable salt thereof, wherein:
R2 is hydrogen; R3 is alkyl; R4 is hydrogen and R5 is alkyl, or cycloalkyl; or R4 is alkyl or cycloalkyl, and R5 is hydrogen.
21. The compound of any one of claims 19 or 20 having the Formula:
Figure imgf000204_0001
22. A compound selected from:
ethyl ((4aR,6R,7S,7aR)-6-(6-amino-2-chloro-9H-purin-9-yl)-7-fluoro-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-alaninate, ethyl ((4aR,6R,7S,7aR)-6-(6-amino-2-chloro-9H-purin-9-yl)-7-fluoro-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-alaninate, ((2R,3R,4S,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-4-fluoro-3- hydroxytetrahydrofuran-2-yl)methyl hydrogen benzylphosphoramidate,
((2R,3R,4S,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-4-fluoro-3- hydroxytetrahydrofuran-2-yl)methyl di(((3-hydroxy-2,2- dimethylpropanoyl)thio)ethyl) phosphate,
S-(2-(((((2R,3R,4S,5R)-5-(6-amino-2-chloro-9H-punn-9-yl)-4-fluoro-3- hydroxytetrahydrofuran-2-yl)methoxy)(benzylamino)phosphoryl)oxy)ethyl) 3- hydroxy-2,2-dimethylpropanethioate,
S-(2-(((((2R,3R,4S,5R)-5-(6-amino-2-chloro-9H-punn-9-yl)-4-fluoro-3- hydroxytetrahydrofuran-2-yl)methoxy)(benzylamino)phosphoryl)oxy)ethyl) 3- hydroxy-2,2-dimethylpropanethioate,
methyl ((((2R,3R,4S,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-4-fluoro-3- hydroxytetrahydrofuran-2-yl)methoxy)(2-methoxy-2-oxoethoxy)phosphoryl)-L- leucinate,
methyl ((((2R,3R,4S,5R)-5-(6-amino-2-chloro-9H-purin-9-yl)-4-fluoro-3- hydroxytetrahydrofuran-2-yl)methoxy)(2-methoxy-2-oxoethoxy)phosphoryl)-L- leucinate,
((2R,3 S,4S, 5R)-5 -(4-amino-2-oxopynmidin-l (2H)-yl)-3 ,4- dihydroxytetrahydrofuran-2-yl)methyl hydrogen benzylphosphoramidate, ((2R,3S,4S,5R)-5-(4-amino-2-oxopyrimidin-l(2H)-yl)-3,4- dihydroxytetrahy drofuran-2-y l)methyl (( 1 -methyl-2-nitro- 1 H-imidazol-5 - yl)methyl) benzylphosphoramidate,
((2R,3S,4S,5R)-5-(4-amino-2-oxopynmidin-l(2H)-yl)-3,4- dihydroxytetrahy drofuran-2-y l)methyl (( 1 -methyl-2-nitro- 1 H-imidazol-5 - yl)methyl) benzylphosphoramidate,
methyl ((((2R,3S,4S,5R)-5-(4-amino-2-oxopynmidin-l(2H)-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(2-methoxy-2-oxoethoxy)phosphoryl)-L- leucinate,
ethyl ((((2R,3S,4S,5R)-5-(4-amino-2-oxopynmidin-l (2H)-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(2-ethoxy-2-oxoethoxy)phosphoryl)-L- leucinate,
ethyl ((((2R,3S,4S,5R)-5-(4-amino-2-oxopynmidin-l (2H)-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(2-ethoxy-2-oxoethoxy)phosphoryl)-L- leucinate,
S-(2-(((((2R,3S,4S,5R)-5-(4-amino-2-oxopynmidin-l(2H)-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(benzylamino)phosphoryl)oxy)ethyl) 3- hydroxy-2,2-dimethylpropanethioate,
methyl (2S)-((((2R,3S,4S,5R)-3,4-dihydroxy-5-(4-((E)-octadec-9-enamido)-2- oxopyrimidin-l (2H)-yl)tetrahydrofuran-2-yl)methoxy)(((S)-l-methoxy-4-methyl- l-oxopentan-2-yl)amino)phosphoryl)-L-leucinate,
((2R,3 S,4S, 5R)-5 -(6-amino-2-fluoro-9H-purin-9-yl)-3 ,4- dihydroxytetrahydrofuran-2-yl)methyl hydrogen benzylphosphoramidate,
((2R,3S,4S,5R)-5-(4-amino-2-oxopynmidin-l(2H)-yl)-3,4- dihydroxytetrahy drofuran-2-y l)methyl (( 1 -methyl-2-nitro- 1 H-imidazol-5 - yl)methyl) benzylphosphoramidate,
ethyl ((4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-leucinate, ethyl ((4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-leucinate, isopropyl ((((2R,3S,4S,5R)-5-(6-amino-2-fluoro-9H-purin-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(phenoxy)phosphoryl)-L-alaninate, S-(2-(((((2R,3S,4S,5R)-5-(6-amino-2-fluoro-9H-purin-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(benzylamino)phosphoryl)oxy)ethyl) 3- hydroxy-2,2-dimethylpropanethioate,
ethyl ((((2R,3S,4S,5R)-5-(6-amino-2-fluoro-9H-punn-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(2-((3-hydroxy-2,2- dimethylpropanoyl)thio)ethoxy)phosphoryl)-L-leucinate,
(4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-punn-9-yl)-7-hydroxy-2- isopropoxytetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinine 2-oxide,
(4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-punn-9-yl)-7-hydroxy-2- isopropoxytetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinine 2-oxide, ethyl ((((2R,3S,4S,5R)-5-(6-amino-2-fluoro-9H-punn-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(hydroxy)phosphoryl)-L-leucinate, ethyl ((((2R,3S,4S,5R)-5-(6-amino-2-fluoro-9H-punn-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(2-ethoxy-2-oxoethoxy)phosphoryl)-L- leucinate,
methyl ((((2R,3S,4S,5R)-5-(6-amino-2-fluoro-9H-purin-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(2S- 1 -methoxy- 1 -oxo-4-methylpent-2- ylamino)phosphoryl)-L-leucinate,
methyl ((((2R,3R,4S,5R)-5-(6-amino-2-fluoro-9H-purin-9-yl)-4-
(((benzyloxy)carbonyl)oxy)-3-hydroxytetrahydrofuran-2-yl)methoxy)(2-methoxy-
2-oxoethoxy)phosphoryl)-L-leucinate,
methyl ((((2R,3S,4S,5R)-5-(6-amino-2-fluoro-9H-purin-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(2-methoxy-2-oxoethoxy)phosphoryl)-L- leucinate, (2S)-2-(((4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)amino)propyl propionate,
(2S)-2-(((4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)amino)propyl propionate,
ethyl ((4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-alaninate, ethyl ((4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-alaninate, ethyl ((4aR,6R,7S,7aS)-6-(2-amino-6-methoxy-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-alaninate, ethyl ((4aR,6R,7S,7aS)-6-(2-amino-6-methoxy-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)-L-alaninate,
(2S)-2-(((4aR,6R,7S,7aS)-6-(2-amino-6-methoxy-9H-purin-9-yl)-7-hydroxy-2- oxidotetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2-yl)amino)propyl propionate,
S-(2-(((((2R,3S,4S,5R)-5-(2-amino-6-methoxy-9H-punn-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methoxy)(benzylamino)phosphoryl)oxy)ethyl) 3- hydroxy-2,2-dimethylpropanethioate, or
((2R,3S,4S,5R)-5-(2-amino-6-methoxy-9H-purin-9-yl)-3,4- dihydroxytetrahydrofuran-2-yl)methyl hydrogen benzylphosphoramidate, or a pharmaceutically acceptable salt thereof
A compound selected from:
ethyl (2S)-2-[[(4aR,6R,7S,7aR)-6-(6-amino-2-chloro-purin-9-yl)-7-fluoro-2-oxo- 4a,6,7,7a-tetrahydro-4H-furo [3 ,2-d] [ 1 ,3,2]dioxaphosphinin-2- yl]amino]propanoate (diastereomer 1 and 2);
[(2R,3S,4S,5R)-5-(6-amino-2-fluoro-purin-9-yl)-3,4-dihydroxy-tetrahydrofuran- 2-yl]methoxy-N-benzyl-phosphonamidic acid; 4-amino-l -[(2R,3S,4S,5R)-5-[[(benzylamino)-[(3 -methyl -2-nitro-imidazol-4- yl)methoxy]phosphoryl]oxymethyl]-3,4-dihydroxy-tetrahydrofuran-2- yl]pyrimidin-2-one (diastereomer 1 and 2);
[(2R,3S,4S,5R)-5-(4-amino-2-oxo-pyrimidin-l-yl)-3,4-dihydroxy- tetrahydrofuran-2-yl]methyl bis[(2-nitrothiazol-5-yl)methyl] phosphate;
[(2R,3S,4S,5R)-5-(4-amino-2-oxo-pyrimidin-l-yl)-3,4-dihydroxy- tetrahydrofuran-2-yl]methyl (3-methyl-2-nitro-imidazol-4-yl)methyl (2- nitrothiazol-5-yl)methyl phosphate;
4-amino-l -[(2R,3S,4S,5R)-5-[[(benzylamino)-[(5-nitro-2- thienyl)methoxy]phosphoryl]oxymethyl]-3,4-dihydroxy-tetrahydrofuran-2- yl]pyrimidin-2-one (diastereomer 1 and 2);
[(2R,3S,4S,5R)-5-(4-amino-2-oxo-pyrimidin-l-yl)-3,4-dihydroxy- tetrahydrofuran-2-yl]methyl bis[(5-nitro-2-furyl)methyl] phosphate;
benzyl (2S)-2-[[[(2R,3S,4S,5R)-5-(4-amino-2-oxo-pyrimidin-l-yl)-3,4-dihydroxy tetrahydrofuran-2-yl]methoxy-[(5-nitro-2- thienyl)methoxy]phosphoryl]amino]propanoate (diastereomer 1 and 2);
4-amino-l -[(2R,3S,4S,5R)-5-[[(benzylamino)-[(5-nitro-2- furyl)methoxy]phosphoryl]oxymethyl]-3,4-dihydroxy-tetrahydrofuran-2- yl]pyrimidin-2-one(diastereomer 1 and 2);
benzyl (2S)-2-[[(4aR,6R,7S,7aR)-6-(6-amino-2-chloro-purin-9-yl)-7-fluoro-2- oxo-4a,6,7,7a-tetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2- yl]amino]propanoate (diastereomer 1 and 2); and
ethyl (2S)-2-[[(4aR,6R,7S,7aS)-6-(6-amino-2-fluoro-punn-9-yl)-7-hydroxy-2- oxo-4a,6,7,7a-tetrahydro-4H-furo[3,2-d][l,3,2]dioxaphosphinin-2- yl]amino]propanoate (diastereomer 1 and 2);
Methyl (2S)-2-[[[(2R,3S,4S,5R)-5-(6-amino-2-fluoro-purin-9-yl)-3,4-dihydroxy- tetrahydrofuran-2-yl]methoxy-(2-methoxy-2-oxo-ethoxy)phosphoryl]amino]-4- methyl-pentanoate;
or a pharmaceutically acceptable salt thereof
24. A pharmaceutical composition comprising a compound of any one of claims 1-23, or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable excipient, carrier, or diluents.
25. The pharmaceutical composition of claim 24, wherein the composition is an oral formulation.
26. A method for the treatment of a blood cancer comprising administering an effective treatment amount of a compound of any of claims 1-23, or a pharmaceutically acceptable salt thereof or a composition of claim 24 or 25.
27. The method of claim 26, wherein the blood cancer is a leukemia cancer.
28. The method of claim 27, wherein the leukemia is acute myelogenous leukemia
(AML).
29. The method of claim 26, wherein said compound or composition is administered in combination or alternation with a second chemotherapeutic agent.
30. A combination comprising a compound of any one of claims 1-23, or a pharmaceutically acceptable salt thereof and one, two, three or more other therapeutic agents.
PCT/IB2015/000957 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer WO2015181624A2 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
JP2016569379A JP2017516779A (en) 2014-05-28 2015-05-27 Nucleoside derivatives for cancer treatment
US15/313,646 US20170101431A1 (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer
CN201580028114.XA CN106459127A (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer
RU2016149761A RU2016149761A (en) 2014-05-28 2015-05-27 NUCLEOSIDE DERIVATIVES FOR THE TREATMENT OF CANCER DISEASE
CA2947939A CA2947939A1 (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer
AU2015265607A AU2015265607A1 (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer
EP15770630.0A EP3149017B1 (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer
MX2016015568A MX2016015568A (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer.
KR1020167036090A KR20170005492A (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462004006P 2014-05-28 2014-05-28
US62/004,006 2014-05-28

Publications (2)

Publication Number Publication Date
WO2015181624A2 true WO2015181624A2 (en) 2015-12-03
WO2015181624A3 WO2015181624A3 (en) 2016-03-31

Family

ID=54196997

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2015/000957 WO2015181624A2 (en) 2014-05-28 2015-05-27 Nucleoside derivatives for the treatment of cancer

Country Status (5)

Country Link
JP (1) JP2017516779A (en)
KR (1) KR20170005492A (en)
AU (1) AU2015265607A1 (en)
CA (1) CA2947939A1 (en)
WO (1) WO2015181624A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2017207986A1 (en) * 2016-06-01 2017-12-07 Nucana Biomed Limited Phosphoramidate nucleoside derivatives as anticancer agents
US10766917B2 (en) 2015-05-27 2020-09-08 Idenix Pharmaceuticals Llc Nucleotides for the treatment of cancer
EP3610020A4 (en) * 2017-12-01 2021-01-27 Aptabio Therapeutics Inc. Therapeutic agent for blood cancer
US11400107B2 (en) 2016-06-01 2022-08-02 NuCana plc Cancer treatments
WO2022245584A1 (en) * 2021-05-17 2022-11-24 Ligand Pharmaceuticals Incorporated Unnatural configuration nucleotide prodrug compounds
WO2023081105A1 (en) * 2021-11-02 2023-05-11 Ligand Pharmaceuticals Incorporated Cyclic phosphoramidate compounds

Citations (50)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
WO1994006802A1 (en) 1992-09-18 1994-03-31 Yoshitomi Pharmaceutical Industries, Ltd. Thienodiazepine compound and medicinal use thereof
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5639480A (en) 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5739108A (en) 1984-10-04 1998-04-14 Monsanto Company Prolonged release of biologically active polypeptides
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US5922356A (en) 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
US5972891A (en) 1992-12-07 1999-10-26 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US5993855A (en) 1995-09-18 1999-11-30 Shiseido Company, Ltd. Delayed drug-releasing microspheres
US6045830A (en) 1995-09-04 2000-04-04 Takeda Chemical Industries, Ltd. Method of production of sustained-release preparation
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6113943A (en) 1996-10-31 2000-09-05 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
WO2001002369A2 (en) 1999-07-02 2001-01-11 Agouron Pharmaceuticals, Inc. Indazole compounds and pharmaceutical compositions for inhibiting protein kinases, and methods for their use
US6197350B1 (en) 1996-12-20 2001-03-06 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6267981B1 (en) 1995-06-27 2001-07-31 Takeda Chemical Industries, Ltd. Method of producing sustained-release preparation
WO2002010192A2 (en) 2000-08-01 2002-02-07 Novartis Ag Somatostatin analogues
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
WO2002068470A2 (en) 2001-02-26 2002-09-06 Pharma Pacific Pty Ltd Interferon-alpha induced gene
US6589548B1 (en) 1998-05-16 2003-07-08 Mogam Biotechnology Research Institute Controlled drug delivery system using the conjugation of drug to biodegradable polyester
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
WO2009084693A1 (en) 2007-12-28 2009-07-09 Mitsubishi Tanabe Pharma Corporation Antitumor agent
WO2010027827A2 (en) 2008-08-25 2010-03-11 Amplimmune, Inc. Targeted costimulatory polypeptides and methods of use to treat cancer
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3804827A (en) * 1972-02-28 1974-04-16 Icn Pharmaceuticals 1-beta-d-arabinofuranosylcytosine-3',5'-cyclic phosphate and derivatives thereof
EP1069903A1 (en) * 1998-03-11 2001-01-24 Lipitek International, Inc. Novel nucleoside analogs and uses in treating disease
GB0505781D0 (en) * 2005-03-21 2005-04-27 Univ Cardiff Chemical compounds
WO2007056596A2 (en) * 2005-11-09 2007-05-18 Wayne State University Phosphoramidate derivatives of fau
US20080261913A1 (en) * 2006-12-28 2008-10-23 Idenix Pharmaceuticals, Inc. Compounds and pharmaceutical compositions for the treatment of liver disorders
US9156874B2 (en) * 2011-01-03 2015-10-13 Nanjing Molecular Research, Inc. Double-liver-targeting phosphoramidate and phosphonoamidate prodrugs
SG192841A1 (en) * 2011-03-01 2013-09-30 Nucana Biomed Ltd Phosphoramidate derivatives of 5 - fluoro - 2 ' - deoxyuridine for use in the treatment of cancer
EP2852604B1 (en) * 2012-05-22 2017-04-12 Idenix Pharmaceuticals LLC 3',5'-cyclic phosphoramidate prodrugs for hcv infection

Patent Citations (56)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3536809A (en) 1969-02-17 1970-10-27 Alza Corp Medication method
US3598123A (en) 1969-04-01 1971-08-10 Alza Corp Bandage for administering drugs
US3845770A (en) 1972-06-05 1974-11-05 Alza Corp Osmatic dispensing device for releasing beneficial agent
US3916899A (en) 1973-04-25 1975-11-04 Alza Corp Osmotic dispensing device with maximum and minimum sizes for the passageway
US4008719A (en) 1976-02-02 1977-02-22 Alza Corporation Osmotic system having laminar arrangement for programming delivery of active agent
US5739108A (en) 1984-10-04 1998-04-14 Monsanto Company Prolonged release of biologically active polypeptides
US5354556A (en) 1984-10-30 1994-10-11 Elan Corporation, Plc Controlled release powder and process for its preparation
US5033252A (en) 1987-12-23 1991-07-23 Entravision, Inc. Method of packaging and sterilizing a pharmaceutical product
US5052558A (en) 1987-12-23 1991-10-01 Entravision, Inc. Packaged pharmaceutical product
US5073543A (en) 1988-07-21 1991-12-17 G. D. Searle & Co. Controlled release formulations of trophic factors in ganglioside-lipsome vehicle
US5059595A (en) 1989-03-22 1991-10-22 Bioresearch, S.P.A. Pharmaceutical compositions containing 5-methyltetrahydrofolic acid, 5-formyltetrahydrofolic acid and their pharmaceutically acceptable salts in controlled-release form active in the therapy of organic mental disturbances
US5639480A (en) 1989-07-07 1997-06-17 Sandoz Ltd. Sustained release formulations of water soluble peptides
US5120548A (en) 1989-11-07 1992-06-09 Merck & Co., Inc. Swelling modulated polymeric drug delivery device
US5733566A (en) 1990-05-15 1998-03-31 Alkermes Controlled Therapeutics Inc. Ii Controlled release of antiparasitic agents in animals
US5639476A (en) 1992-01-27 1997-06-17 Euro-Celtique, S.A. Controlled release formulations coated with aqueous dispersions of acrylic polymers
US5323907A (en) 1992-06-23 1994-06-28 Multi-Comp, Inc. Child resistant package assembly for dispensing pharmaceutical medications
WO1994006802A1 (en) 1992-09-18 1994-03-31 Yoshitomi Pharmaceutical Industries, Ltd. Thienodiazepine compound and medicinal use thereof
US5972891A (en) 1992-12-07 1999-10-26 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5591767A (en) 1993-01-25 1997-01-07 Pharmetrix Corporation Liquid reservoir transdermal patch for the administration of ketorolac
US6376461B1 (en) 1993-06-24 2002-04-23 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6087324A (en) 1993-06-24 2000-07-11 Takeda Chemical Industries, Ltd. Sustained-release preparation
US5712274A (en) 1993-09-16 1998-01-27 Yoshitomi Pharmaceutical Industries, Ltd. Thienotriazolodiazepine compounds and their pharmaceutical use
US5674533A (en) 1994-07-07 1997-10-07 Recordati, S.A., Chemical And Pharmaceutical Company Pharmaceutical composition for the controlled release of moguisteine in a liquid suspension
US6267981B1 (en) 1995-06-27 2001-07-31 Takeda Chemical Industries, Ltd. Method of producing sustained-release preparation
US6045830A (en) 1995-09-04 2000-04-04 Takeda Chemical Industries, Ltd. Method of production of sustained-release preparation
US5993855A (en) 1995-09-18 1999-11-30 Shiseido Company, Ltd. Delayed drug-releasing microspheres
US5980945A (en) 1996-01-16 1999-11-09 Societe De Conseils De Recherches Et D'applications Scientifique S.A. Sustained release drug formulations
US6264970B1 (en) 1996-06-26 2001-07-24 Takeda Chemical Industries, Ltd. Sustained-release preparation
US6419961B1 (en) 1996-08-29 2002-07-16 Takeda Chemical Industries, Ltd. Sustained release microcapsules of a bioactive substance and a biodegradable polymer
US5922356A (en) 1996-10-09 1999-07-13 Sumitomo Pharmaceuticals Company, Limited Sustained release formulation
US6113943A (en) 1996-10-31 2000-09-05 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
US6699500B2 (en) 1996-10-31 2004-03-02 Takeda Chemical Industries, Ltd. Sustained-release preparation capable of releasing a physiologically active substance
US6197350B1 (en) 1996-12-20 2001-03-06 Takeda Chemical Industries, Ltd. Method of producing a sustained-release preparation
US5891474A (en) 1997-01-29 1999-04-06 Poli Industria Chimica, S.P.A. Time-specific controlled release dosage formulations and method of preparing same
US6613358B2 (en) 1998-03-18 2003-09-02 Theodore W. Randolph Sustained-release composition including amorphous polymer
US6589548B1 (en) 1998-05-16 2003-07-08 Mogam Biotechnology Research Institute Controlled drug delivery system using the conjugation of drug to biodegradable polyester
WO2001002369A2 (en) 1999-07-02 2001-01-11 Agouron Pharmaceuticals, Inc. Indazole compounds and pharmaceutical compositions for inhibiting protein kinases, and methods for their use
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
WO2002010192A2 (en) 2000-08-01 2002-02-07 Novartis Ag Somatostatin analogues
WO2002068470A2 (en) 2001-02-26 2002-09-06 Pharma Pacific Pty Ltd Interferon-alpha induced gene
WO2004004771A1 (en) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Immunopotentiating compositions
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
WO2004056875A1 (en) 2002-12-23 2004-07-08 Wyeth Antibodies against pd-1 and uses therefor
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2009084693A1 (en) 2007-12-28 2009-07-09 Mitsubishi Tanabe Pharma Corporation Antitumor agent
US8476260B2 (en) 2007-12-28 2013-07-02 Mitsubishi Tanabe Pharma Corporation Antitumor agent
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
WO2010027827A2 (en) 2008-08-25 2010-03-11 Amplimmune, Inc. Targeted costimulatory polypeptides and methods of use to treat cancer
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
WO2010077634A1 (en) 2008-12-09 2010-07-08 Genentech, Inc. Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2011066342A2 (en) 2009-11-24 2011-06-03 Amplimmune, Inc. Simultaneous inhibition of pd-l1/pd-l2
WO2013019906A1 (en) 2011-08-01 2013-02-07 Genentech, Inc. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors

Non-Patent Citations (26)

* Cited by examiner, † Cited by third party
Title
"Annexin V for flow cytometric detection ofphosphatidylserine expression on B cells undergoing apoptosis", BLOOD, vol. 84, 1994, pages 1415 - 20
"Introduction to Pharmaceutical Dosage Forms", 1985, LEA & FEBIGER
"Remington's Pharmaceutical Sciences", 1980, MACK PUBLISHING
"Remington's Pharmaceutical Sciences", 2000, MACK PUBLISHING
"Response Evaluation Criteria in Solid Tumors (RECIST) Guidelines", JOURNAL OF THE NATIONAL CANCER INSTITUTE, vol. 92, no. 3, 2000, pages 205 - 216
"The Merck Manual", 1999
BUCHWALD ET AL., SURGERY, vol. 88, 1980, pages 507
FACTS 2013, August 2013 (2013-08-01)
FOSTER ET AL., ADV. DRUG RES., vol. 14, 1985, pages 1 - 36
GATELY, J. NUCL. MED., vol. 27, 1986, pages 388
GOODSON, MEDICAL APPLICATIONS OF CONTROLLED RELEASE, vol. 2, 1984, pages 115 - 138
GORDON, DRUG METAB. DISPOS., vol. 15, 1987, pages 589
GREENE ET AL.: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS
GREENE: "Protective Groups in Organic Synthesis", 1991, JOHN WILEY AND SONS
JENS T. CARSTENSEN: "Drug Stability: Principles & Practice", 1995, MARCEL DEKKER, pages: 379 - 80
KUSHNER ET AL., CAN. J. PHYSIOL. PHARMACOL., vol. 77, 1999, pages 79 - 88
LANGER, SCIENCE, vol. 249, 1990, pages 1527 - 1533
LIJINSKY, FOOD COSMET. TOXICOL., vol. 20, 1982, pages 393
LIJINSKY, J. NAT. CANCER INST., vol. 69, 1982, pages 1127
MANGOLD, MUTATION RES., vol. 308, 1994, pages 33
SAUDEK ET AL., N. ENGL. J. MED., vol. 321, 1989, pages 574
SEFTON, CRC CRIT. REF. BIOMED. ENG., vol. 14, 1987, pages 201
VAN DONGEN, J.J. ET AL.: "EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes", LEUKEMIA, vol. 26, 2012, pages 1908 - 75
VAN DONGEN, J.J.; A. ORFAO: "EuroFlow: Resetting leukemia and lymphoma immunophenotyping. Basis for companion diagnostics and personalized medicine", LEUKEMIA, vol. 26, 2012, pages 1899 - 907
WADE D, CHEM. BIOL. INTERACT., vol. 117, 1999, pages 191
ZELLO, METABOLISM, vol. 43, 1994, pages 487

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10766917B2 (en) 2015-05-27 2020-09-08 Idenix Pharmaceuticals Llc Nucleotides for the treatment of cancer
WO2017207986A1 (en) * 2016-06-01 2017-12-07 Nucana Biomed Limited Phosphoramidate nucleoside derivatives as anticancer agents
CN109195981A (en) * 2016-06-01 2019-01-11 努卡那有限公司 Phosphoramidate nucleoside derivative as anticancer agent
JP2019517510A (en) * 2016-06-01 2019-06-24 ニューカナ パブリック リミテッド カンパニー Phosphoramidate nucleoside derivatives as anticancer agents
EA036409B1 (en) * 2016-06-01 2020-11-06 НУКАНА ПиЭлСи Phosphoramidate nucleoside derivatives as anticancer agents
US10906929B2 (en) 2016-06-01 2021-02-02 NuCana plc Phosphoramidate nucleoside derivatives as anticancer agents
JP7025351B2 (en) 2016-06-01 2022-02-24 ニューカナ パブリック リミテッド カンパニー Phosphoramidate nucleoside derivative as an anticancer agent
US11400107B2 (en) 2016-06-01 2022-08-02 NuCana plc Cancer treatments
EP3610020A4 (en) * 2017-12-01 2021-01-27 Aptabio Therapeutics Inc. Therapeutic agent for blood cancer
WO2022245584A1 (en) * 2021-05-17 2022-11-24 Ligand Pharmaceuticals Incorporated Unnatural configuration nucleotide prodrug compounds
WO2023081105A1 (en) * 2021-11-02 2023-05-11 Ligand Pharmaceuticals Incorporated Cyclic phosphoramidate compounds

Also Published As

Publication number Publication date
KR20170005492A (en) 2017-01-13
CA2947939A1 (en) 2015-12-03
JP2017516779A (en) 2017-06-22
WO2015181624A3 (en) 2016-03-31
AU2015265607A1 (en) 2016-11-17

Similar Documents

Publication Publication Date Title
EP3149017B1 (en) Nucleoside derivatives for the treatment of cancer
WO2015181624A2 (en) Nucleoside derivatives for the treatment of cancer
US10766917B2 (en) Nucleotides for the treatment of cancer
US10723754B2 (en) 2′,4′-bridged nucleosides for HCV infection
US9211300B2 (en) 4′-fluoro nucleosides for the treatment of HCV
US10238680B2 (en) D-amino acid phosphoramidate pronucleotides of halogeno pyrimidine compounds for liver disease
US9187515B2 (en) 2′,4′-fluoro nucleosides for the treatment of HCV
US10202411B2 (en) 3′-substituted methyl or alkynyl nucleosides nucleotides for the treatment of HCV
JP2017504572A (en) Nucleotides for liver cancer treatment
US20170198005A1 (en) 2&#39;-dichloro and 2&#39;-fluoro-2&#39;-chloro nucleoside analogues for hcv infection
US20160113956A1 (en) Amino acid phosphoramidate pronucleotides of 2&#39;-cyano, azido and amino nucleosides for the treatment of hcv
EP3004130A1 (en) 1&#39;,4&#39;-thio nucleosides for the treatment of hcv
US20140099283A1 (en) 2&#39;-chloro nucleoside analogs for hcv infection
US10815264B2 (en) Nucleotides for the treatment of cancer
EP3083654A1 (en) 4&#39;-or nucleosides for the treatment of hcv
WO2015061683A1 (en) D-amino acid phosphoramidate and d-alanine thiophosphoramidate pronucleotides of nucleoside compounds useful for the treatment of hcv

Legal Events

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

Ref document number: 15770630

Country of ref document: EP

Kind code of ref document: A2

ENP Entry into the national phase

Ref document number: 2947939

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2015265607

Country of ref document: AU

Date of ref document: 20150527

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 15313646

Country of ref document: US

ENP Entry into the national phase

Ref document number: 2016569379

Country of ref document: JP

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: MX/A/2016/015568

Country of ref document: MX

NENP Non-entry into the national phase

Ref country code: DE

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112016027574

Country of ref document: BR

REEP Request for entry into the european phase

Ref document number: 2015770630

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 2015770630

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 20167036090

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2016149761

Country of ref document: RU

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112016027574

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20161124