PHOSPHADIAZINE HCV POLYMERASE INHIBITORS I AND II

Provided herein are phosphadiazine polymerase inhibitor, for example, of any of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, pharmaceutical compositions comprising the compounds, and processes of preparation thereof. Also provided are methods of their use for the treatment of an HCV infection in a host in need thereof.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims priority to U.S. Provisional Application No. 60/967,237, filed Aug. 31, 2007, the content of which is incorporated herein by reference in its entirety.

FIELD

Provided herein are phosphadiazine polymerase inhibitor compounds, pharmaceutical compositions comprising the compounds, and processes of preparation thereof. Also provided are methods of their use for the treatment of an HCV infection in a host in need thereof.

BACKGROUND

Hepatitis C virus (HCV) is known to cause at least 80% of posttransfusion hepatitis and a substantial proportion of sporadic acute hepatitis (Houghton et al., Science 1989, 244, 362-364; Thomas, Curr. Top. Microbiol. Immunol. 2000, 25-41). Preliminary evidence also implicates HCV in many cases of “idiopathic” chronic hepatitis, “cryptogenic” cirrhosis, and probably hepatocellular carcinoma unrelated to other hepatitis viruses, such as hepatitis B virus (Di Besceglie et al., Scientfic American, 1999, October, 80-85; Boyer et al., J. Hepatol. 2000, 32, 98-112).

HCV is an enveloped virus containing a positive-sense single-stranded RNA genome of approximately 9.4 kb (Kato et al., Proc. Natl. Acad. Sci. USA 1990, 87, 9524-9528; Kato, Acta Medica Okayama, 2001, 55, 133-159). The viral genome consists of a 5′ untranslated region (UTR), a long open reading frame encoding a polyprotein precursor of approximately 3011 amino acids, and a short 3′ UTR. The 5′ UTR is the most highly conserved part of the HCV genome and is important for the initiation and control of polyprotein translation. Translation of the HCV genome is initiated by a cap-independent mechanism known as internal ribosome entry. This mechanism involves the binding of ribosomes to an RNA sequence known as the internal ribosome entry site (IRES). An RNA pseudoknot structure has recently been determined to be an essential structural element of the HCV TRES. Viral structural proteins include a nucleocapsid core protein (C) and two envelope glycoproteins, E1 and E2. HCV also encodes two proteinases, a zinc-dependent metalloproteinase encoded by the NS2-NS3 region and a serine proteinase encoded in the NS3 region. These proteinases are required for cleavage of specific regions of the precursor polyprotein into mature peptides. The carboxyl half of nonstructural protein 5, NS5B, contains the RNA-dependent RNA polymerase. The function of the remaining nonstructural proteins, NS4A and NS4B, and that of NS5A (the amino-terminal half of nonstructural protein 5) remain unknown.

Presently, the most effective HCV therapy employs a combination of alpha-interferon and ribavirin, leading to sustained efficacy in about 40% of patients (Poynard et al., Lancet 1998, 352, 1426-1432). Recent clinical results demonstrate that pegylated alpha-interferon is superior to unmodified alpha-interferon as monotherapy. However, even with experimental therapeutic regimens involving combinations of pegylated alpha-interferon and ribavirin, a substantial fraction of patients do not have a sustained reduction in viral load (Manns et al, Lancet 2001, 358, 958-965; Fried et al., N. Engl. J. Med. 2002, 347, 975-982; Hadziyannis et al., Ann. Intern. Med. 2004, 140, 346-355). Furthermore, research shows that using pegylated interferon and ribavirin to treat patients with HCV can cause significant side effects, such as alopecia, anorexia, depression, fatigue, myalgia, nausea and prunitus (Ward et al., American Family Physician. 2005, Vol. 72, No. 4; Al-Huthail, The Saudi Journal of Gastroenterology. 2006, Vol. 12, No. 2, 59-67). Severe weight loss is also reported as a side effect in the interferon-based therapy in combination with ribavirin (Bani-Sadr et al., Journal of Viral Hepatitis. 2008, 15(4): 255-260). Thus, there is a clear and unmet need to develop effective therapeutics for treatment of HCV infection.

SUMMARY OF THE DISCLOSURE

Provided herein are phosphadiazine polymerase inhibitor compounds, pharmaceutical compositions comprising the compounds, and processes of preparation thereof. Also provided are methods of the use of the compounds for the treatment of an HCV infection in a host in need thereof.

In one aspect, provided herein is a compound of Formula I′ or II′:

or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein:

    • each R1 is independently H, alkyl, arylalkyl, heteroarylalkyl, halogen, —NR10SO2R8, —OR8, NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl;
    • each R2 is independently H, alkyl, aryl-CH2—, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl, or R1 and R2 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each of R4 is independently H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
    • each R4′ is independently H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
    • each R5 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R4 and R5 together form a part of a 3-8 membered heterocycloalkyl ring;
    • each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5 and R6 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R5′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, heteroaryl, —NR8R10, alkenyl, or alkynl;
    • each R6′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl; R5′ and R6′ together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl, or heteroaryl ring;
    • each R12 is independently F, —OR8, —SR8, —NR8R9, alkyl, or aryl;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9; and
    • each Y is independently O or S,
    • wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

In another aspect, provided herein is a compound of Formula I, II or III:

or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein

    • each R1 is independently H, alkyl, arylalkyl, heteroarylalkyl, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl;
    • each R2 is independently H, alkyl, aryl-CH2—, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl, or R1 and R2 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R5 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5 and R6 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5 and R6 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R12 is independently F, —OR8, —SR8, —NR8R9, alkyl, or aryl;
    • each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9;
    • each n is independently an integer from 1 to 4;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; and
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9,
    • wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

Also provided herein are pharmaceutical compositions comprising a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof; in combination with one or more pharmaceutically acceptable excipients or carriers.

Also provided herein is a method for treating or preventing an HCV infection, which comprises administering to a subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, Ia, or IIIa, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

Also provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a liver disease or disorder associated with an HCV infection, comprising administering to a subject a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

Also provided herein is a method for inhibiting replication of a virus in a host, which comprises contacting the host with a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

Also provided herein is a method for inhibiting replication of a virus, which comprises contacting the virus with a therapeutically effective amount of a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereoft or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

Also provided herein is a method for inhibiting the activity of a polymerase, which comprises contacting the polymerase with a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

Provided is a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, or a pharmaceutical composition thereof, for use in therapy. Also provided is a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, or a pharmaceutical composition thereof, for use in treating or preventing an HCV infection. Also provided is a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, or a pharmaceutical composition thereof, for use in treating, preventing, or ameliorating one or more symptoms of a liver disease or disorder associated with an HCV infection. Also provided is a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, or a pharmaceutical composition thereof, for use in inhibiting replication of a virus in a host. Also provided is the use of a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, or a pharmaceutical composition thereof, for manufacture of a medeicament for treating or preventing an HCV infection. Also provided is the use of a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, or a pharmaceutical composition thereof, for manufacture of a medeicament for treating, preventing, or ameliorating one or more symptoms of a liver disease or disorder associated with an HCV infection. Also provided is the use of a compound disclosed herein, e.g., a compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, or a pharmaceutical composition thereof, for manufacture of a medcicament for inhibiting replication of a virus in a host.

DETAILED DESCRIPTION

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. In the event that there is a plurality of definitions for a term used herein, those in this section prevail unless stated otherwise.

The term “subject” refers to an animal, including, but not limited to, a primate (e.g., human), cow, sheep, goat, horse, dog, cat, rabbit, rat, or mouse. The terms “subject” and “patient” are used interchangeably herein in reference, for example, to a mammalian subject, such as a human subject.

The term “host” refers to a unicellular or multicellular organism in which a virus can replicate, including, but not limited to, a cell, cell line, and animal, such as human.

The terms “treat,” “treating,” and “treatment” are meant to include alleviating or abrogating a disorder, disease, or condition, or one or more of the symptoms associated with the disorder, disease, or condition; or alleviating or eradicating the cause(s) of the disorder, disease, or condition itself.

The terms “prevent,” “preventing,” and “prevention” are meant to include a method of delaying and/or precluding the onset of a disorder, disease, or condition, and/or its attendant symptoms; barring a subject from acquiring a disease; or reducing a subject's risk of acquiring a disorder, disease, or condition.

The term “therapeutically effective amount” are meant to include the amount of a compound that, when administered, is sufficient to prevent development of, or alleviate to some extent, one or more of the symptoms of the disorder, disease, or condition being treated. The term “therapeutically effective amount” also refers to the amount of a compound that is sufficient to elicit the biological or medical response of a cell, tissue, system, animal, or human, which is being sought by a researcher, veterinarian, medical doctor, or clinician.

The term “IC50” refers an amount, concentration, or dosage of a compound that is required for 50% inhibition of a maximal response in an assay that measures such response.

The term “pharmaceutically acceptable carrier,” “pharmaceutically acceptable excipient,” “physiologically acceptable carrier,” or “physiologically acceptable excipient” refers to a pharmaceutically-acceptable material, composition, or vehicle, such as a liquid or solid filler, diluent, excipient, solvent, or encapsulating material. In one embodiment, each component is “pharmaceutically acceptable” in the sense of being compatible with the other ingredients of a pharmaceutical formulation, and suitable for use in contact with the tissue or organ of humans and animals without excessive toxicity, irritation, allergic response, immunogenicity, or other problems or complications, commensurate with a reasonable benefit/risk ratio. See, Remington: The Science and Practice of Pharmacy, 21st Edition; Lippincott Williams & Wilkins: Philadelphia, Pa., 2005; Handbook of Pharmaceutical Excipients, 5th Edition; Rowe et al., Eds., The Pharmaceutical Press and the American Pharmaceutical Association: 2005; and Handbook of Pharmaceutical Additives, 3rd Edition; Ash and Ash Eds., Gower Publishing Company: 2007; Pharmaceutical Preformulation and Formulation, Gibson Ed., CRC Press LLC: Boca Raton, Fla., 2004).

The term “about” or “approximately” means an acceptable error for a particular value as determined by one of ordinary skill in the art, which depends in part on how the value is measured or determined. In certain embodiments, the term “about” or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain embodiments, the term “about” or “approximately” means within 50%, 20%, 15%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, or 0.05% of a given value or range.

The terms “active ingredient” and “active substance” refer to a compound, which is administered, alone or in combination with one or more pharmaceutically acceptable excipients, to a subject for treating, preventing, or ameliorating one or more symptoms of a disorder or disease. As used herein, “active ingredient” and “active substance” may be an optically active isomer of a compound described herein.

The terms “drug,” “therapeutic agent,” and “chemotherapeutic agent” refer to a compound, or a pharmaceutical composition thereof, which is administered to a subject for treating, preventing, or ameliorating one or more symptoms of a condition, disorder, or disease.

The term “release controlling excipient” refers to an excipient whose primary function is to modify the duration or place of release of an active substance from a dosage form as compared with a conventional immediate release dosage form.

The term “nonrelease controlling excipient” refers to an excipient whose primary function do not include modifying the duration or place of release of an active substance from a dosage form as compared with a conventional immediate release dosage form.

The term “alkyl” refers to a linear or branched saturated monovalent hydrocarbon radical. The term “alkyl” also encompasses both linear and branched alkyl, unless otherwise specified. In certain embodiments, the alkyl is a linear saturated monovalent hydrocarbon radical that has 1 to 20 (C1-20), 1 to 15 (C1-15), 1 to 10 (C1-10), or 1 to 6 (C1-6) carbon atoms, or branched saturated 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. In certain embodiments, the alkyl is a linear or branched saturated monovalent hydrocarbon radical that has at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms. As used herein, linear C1-6 and branched C3-6 alkyl groups are also referred as “lower alkyl.” Examples of alkyl groups include, but are not limited to, methyl, ethyl, propyl (including all isomeric forms), n-propyl, isopropyl, butyl (including all isomeric forms), n-butyl, isobutyl, t-butyl, pentyl (including all isomeric forms), and hexyl (including all isomeric forms). For example, C1-6 alkyl refers to a linear saturated monovalent hydrocarbon radical of 1 to 6 carbon atoms or a branched saturated monovalent hydrocarbon radical of 3 to 6 carbon atoms. In certain embodiments, the alkyl may be substituted.

The term “alkylene” refers to a linear or branched saturated divalent hydrocarbon radical, wherein the alkylene may optionally be substituted. The term “alkylene” encompasses both linear and branched alkylene, unless otherwise specified. In certain embodiments, the alkylene is a linear saturated divalent hydrocarbon radical that has 1 to 20 (C1-20), 1 to 15 (C1-15), 1 to 10 (C1-10), or 1 to 6 (C1-6) carbon atoms, or branched saturated 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. In certain embodiments, the alkylene is a linear or branched saturated divalent hydrocarbon radical that has at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 carbon atoms. As used herein, linear C1-6 and branched C3-6 alkylene groups are also referred as “lower alkylene.” Examples of alkylene groups include, but are not limited to, methylene, ethylene, propylene (including all isomeric forms), n-propylene, isopropylene, butylene (including all isomeric forms), n-butylene, isobutylene, t-butylene, pentylene (including all isomeric forms), and hexylene (including all isomeric forms). For example, C2-6 alkylene refers to a linear saturated divalent hydrocarbon radical of 2 to 6 carbon atoms or a branched saturated divalent hydrocarbon radical of 3 to 6 carbon atoms.

The term “alkenyl” refers to a linear or branched monovalent hydrocarbon radical, which contains one or more carbon-carbon double bonds. The alkenyl may be optionally substituted, e.g., as described herein. The term “alkenyl” also embraces radicals having “cis” and “trans” configurations, or alternatively, “E” and “Z” configurations, as appreciated by those of ordinary skill in the art. As used herein, the term “alkenyl” encompasses both linear and branched alkenyl, unless otherwise specified. 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, propenyl, allyl, propenyl, butenyl, and 4-methylbutenyl.

The term “alkenylene” refers to a linear or branched divalent hydrocarbon radical, which contains one or more carbon-carbon double bonds. The alkenylene may be optionally substituted, e.g., as described herein. Similarly, the term “alkenylene” also embraces radicals having “cis” and “trans” configurations, or alternatively, “E” and “Z” configurations. As used herein, the term “alkenylene” encompasses both linear and branched alkenylene, unless otherwise specified. 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 (C2-20), 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, propenylene, allylene, propenylene, butenylene, and 4-methylbutenylene.

The term “alkynyl” refers to a linear or branched monovalent hydrocarbon radical, which contains one or more carbon-carbon triple bonds. The alkynyl may be optionally substituted, e.g., as described herein. The term “alkynyl” also encompasses both linear and branched alkynyl, unless otherwise specified. In certain embodiments, the alkynyl 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 alkynyl groups include, but are not limited to, ethynyl (—C≡CH) and propargyl (—CH2C≡CH). For example, C2-6 alkynyl 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 term “alkynylene” refers to a linear or branched divalent hydrocarbon radical, which contains one or more carbon-carbon triple bonds. The alkynylene may be optionally substituted, e.g., as described herein. The term “alkynylene” also encompasses both linear and branched alkynylene, unless otherwise specified. In certain embodiments, the alkynylene is a linear divalent 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 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 alkynylene groups include, but are not limited to, ethynylene (—C≡C—) and propargylene (—CH2C≡C—). For example, C2-6 alkynyl 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 term “cycloalkyl” refers to a cyclic saturated bridged or non-bridged monovalent hydrocarbon radical, which may be optionally substituted, e.g., as described herein. In certain embodiments, the cycloalkyl has from 3 to 20 (C3-20), from 3 to 15 (C3-15), from 3 to 10 (C3-10), or from 3 to 7 (C3-7) carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, decalinyl, and adamantyl.

The term “cycloalkylene” refers to a cyclic saturated bridged or non-bridged divalent hydrocarbon radical, which may be optionally substituted, e.g., as described herein. In certain embodiments, the cycloalkylene has from 3 to 20 (C3-20), from 3 to 15 (C3-15), from 3 to 10 (C3-10), or from 3 to 7 (C3-7) carbon atoms. Examples of cycloalkylene groups include, but are not limited to, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cycloheptylene, decalinylene, and adamantylene.

The term “aryl” refers to a monocyclic or multicyclic monovalent aromatic group. In certain embodiments, the aryl has from 6 to 20 (C6-20), from 6 to 15 (C6-15), or from 6 to 10 (C6-10) ring atoms. Examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, azulenyl, anthryl, phenanthryl, pyrenyl, biphenyl, and terphenyl. Aryl also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthyl, indenyl, indanyl, or tetrahydronaphthyl (tetralinyl). All such aryl groups may also be optionally substituted, e.g., as described herein.

The term “arylene” refers to a monocyclic or multicyclic divalent aromatic group. In certain embodiments, the arylene has from 6 to 20 (C6-20), from 6 to 15 (C6-15), or from 6 to 10 (C6-10) ring atoms. Examples of arylene groups include, but are not limited to, phenylene, naphthylene, fluorenylene, azulenylene, anthrylene, phenanthrylene, pyrenylene, biphenylene, and terphenylene. Arylene also refers to bicyclic or tricyclic carbon rings, where one of the rings is aromatic and the others of which may be saturated, partially unsaturated, or aromatic, for example, dihydronaphthylene, indenylene, indanylene, or tetrahydro-naphthylene (tetralinyl). All such aryl groups may also be optionally substituted, e.g., as described herein.

The term “heteroaryl” refers to a monocyclic or multicyclic aromatic group, wherein at least one ring contains one or more heteroatoms independently selected from O, S, and N. 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, pyrrolyl, pyrazolyl, pyrazolinyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, thienyl, oxadiazolyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, and triazinyl. Examples of bicyclic heteroaryl groups include, but are not limited to, indolyl, benzothiazolyl, benzoxazolyl, benzothienyl, quinolinyl, tetrahydroisoquinolinyl, isoquinolinyl, benzimidazolyl, benzopyranyl, indolizinyl, benzofuiranyl, isobenzofuranyl, chromonyl, coumarinyl, cinnolinyl, quinoxalinyl, indazolyl, purinyl, pyrrolopyridinyl, furopyridinyl, thienopyridinyl, dihydroisoindolyl, and tetrahydroquinolinyl. Examples of tricyclic heteroaryl groups include, but are not limited to, carbazolyl, benzindolyl, phenanthrollinyl, acridinyl, phenanthridinyl, and xanthenyl. All such heteroaryl groups may also be optionally substituted, e.g., as described herein.

The term “heterocyclyl” or “heterocyclic” refers to a monocyclic or multicyclic non-aromatic ring system, wherein one or more of the 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. Examples of heterocyclyl groups include, but are not limited to, pyrrolidinyl, piperidinyl, 2-oxopyrrolidinyl, 2-oxopiperidinyl, morpholinyl, piperazinyl, tetrahydropyranyl, and thiomorpholinyl. All such heterocyclic groups may also be optionally substituted, e.g., as described herein.

The term “alkoxy” refers to an —OR radical, wherein R is, for example, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, or heterocyclyl, each as defined herein. Examples of alkoxy groups include, but are not limited to, methoxy, ethoxy, propoxy, n-propoxy, 2-propoxy, n-butoxy, isobutoxy, tert-butoxy, cyclohexyloxy, phenoxy, benzoxy, and 2-naphthyloxy.

The term “acyl” refers to a —C(O)R radical, wherein R is, for example, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, or heterocyclyl, each as defined herein. Examples of acyl groups include, but are not limited to, acetyl, propionyl, butanoyl, isobutanoyl, pentanoyl, hexanoyl, heptanoyl, octanoyl, nonanoyl, decanoyl, dodecanoyl, tetradecanoyl, hexadecanoyl, octadecanoyl, eicosanoyl, docosanoyl, myristoleoyl, palmitoleoyl, oleoyl, linolcoyl, arachidonoyl, benzoyl, pyridinylcarbonyl, and furoyl.

The term “halogen”, “halide” or “halo” refers to fluorine, chlorine, bromine, or iodine.

The term “arylaklyl” refers to an aryl group appended to an alkyl radical, such as aryl-(CH2)—, aryl-CH2—CH2—, and aryl-CH2—CH2—CH2—.

The term “heteroarylalkyl” refers to an heteroaryl group appended to an alikyl radical, such as heteroaryl-(CH2)—, heteroaryl-CH2—CH2—, and heteroaryl-CH2—CH2—CH2—.

The term “optionally substituted” is intended to mean that a group, such as an alkyl, alkylene, alkenyl, alkenylene, alkynyl, alkynylene, alkoxyl, cycloalkyl, cycloalkylene, aryl, arylene, heteroaryl, or heterocyclyl group, may be substituted with one or more substituents independently selected from, e.g., halo, cyano (—CN), nitro (—NO2), —SRa, —S(O)Ra, —S(O)2Ra, —Ra, —C(O)Ra, —C(O)ORa, —C(O)NRbRc, —OCH2C(O)NRbRc, —C(NRa)NRbRc, —ORa, —OC(O)Ra, —OC(O)ORa, —OC(O)NRbRc, —OC(═NRa)NRbRc, —OS(O)Ra, —OS(O)2Ra, —OS(O)NRbRc, —OS(O)2NRbRc, —NRbRc, —NRaC(O)Rb, —NRaC(O)ORb, —NRaC(O)NRbRc, —NRaC(═NRd)NRbRc, —NRaS(O)Rb, —NRaS(O)2Rb, —NRaS(O)RbRc, —NRaS(O)2RbRc, or —OSi—RaRbRc; wherein Ra, Rb, Rc and Rd are each independently, e.g., hydrogen, alkyl, alkenyl, alkynyl, cycloalkyl, aryl, heteroaryl, or heterocyclyl, each optionally substituted, e.g., as described herein; or Rb and Rc together with the N atom to which they are attached form heterocyclyl or heteroaryl, each optionally substituted, e.g., as described herein. The group can be substituted with any described moiety, including, but not limited to, one or more moieties selected from the group consisting of halogen (fluoro, chloro, bromo, or iodo), hydroxyl, amino, alkylamino (e.g., monoalkylamino, dialkylamino, or trialkylamino), arylamino (e.g., monoarylamino, diarylamino, or triarylamino), alkoxy, aryloxy, 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. As used herein, all groups that can be substituted in one embodiment are “optionally substituted,” unless otherwise specified.

In certain embodiments, “optically active” and “enantiomerically active” refer to a collection of molecules, which has an enantiomeric excess of no less than about 50%, no less than about 70%, no less than about 80%, no less than about 90%, no less than about 91%, no less than about 92%, no less than about 93%, or no less than about 94% no less than about 95%, no less than about 96%, no less than about 97%, no less than about 98%, no less than about 99%, or no less than about 99.5%, no less than about 99.8%. In certain embodiments, the compound comprises about 95% or more of the desired enantiomer and about 5% or less of the less preferred enantiomer based on the total weight of the racemate in question.

In describing an optically active compound, the prefixes R and S are used to denote the absolute configuration of the molecule about its chiral center(s). The (+) and (−) are used to denote the optical rotation of the compound, that is, the direction in which a plane of polarized light is rotated by the optically active compound. The (−) prefix indicates that the compound is levorotatory, that is, the compound rotates the plane of polarized light to the left or counterclockwise. The (+) prefix indicates that the compound is dextrorotatory, that is, the compound rotates the plane of polarized light to the right or clockwise. However, the sign of optical rotation, (+) and (−), is not related to the absolute configuration of the molecule, R and S.

The term “solvate” refers to a compound provided herein or a salt thereof, which 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.

Compounds

Provided herein are compounds which are usefull for the treatment of HCV infection, which, in one embodiment, can have activity as HCV polymerase inhibitors. Also provided herein are pharmaceutical compositions that comprise the compounds, methods of manufacture of the compounds, and methods of use of the compounds for the treatment of HCV infection in a host in need of treatment.

In one aspect, provided herein is a compound of Formula I′ or II′:

or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein:

    • each R1 is independently H, alkyl, arylalkyl, heteroarylalkyl, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyt, heterocyclylalkyl, sulfonyl, or heteroaryl;
    • each R2 is independently H, alkyl, aryl-CH2—, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl, or R1 and R2 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R4 is independently H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
    • each R4′ is independently H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
    • each R5 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R4 and R5 together form a part of a 3-8 membered heterocycloalkyl ring or heteroaryl ring;
    • each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5 and R6 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R5′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, heteroaryl, —NR8R10, alkenyl, or alkynl;
    • each R6′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5′ and R6′ together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R12 is independently F, —OR8, —SR8, —NR8R9, alkyl, or aryl;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl,
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9; and
    • each Y is independently O or S,
    • wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

In certain embodiments, each pair of R5′ and R6′ together independently form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring. In some embodiments, R5′ and R6′ together independently form a part of a ring having formula O or P:

where

    • each * is a bond;
    • each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl;
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9;
    • each n is independently an integer from 0 to 1, from 0 to 2, from 0 to 3, or from 0 to 4;
    • each m is independently an integer from 0 to 1, from 0 to 2, or from 0 to 3;
    • each A is independently CR15R16 or NR17;
    • each A′ is independently CR15R16, NR17, N, CR15, N-oxide, N—OR8—, S or O;
    • each R15 is independently a bond, H, halogen, —NR10SO2R8, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R10, —(C1-C6 alkylene)-NR9′S(O)2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
    • each R16 is independengly a bond, H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl; and
    • each R17 is independently a bond, H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl,
      wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

In some embodiments, each pair of R5 and R6 together independently form a benzo group having formula (A):

where

    • each * is a bond;
    • each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl;
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8R9; and
    • n is an integer from 1 to 4.

In some embodiments, each pair of R5′ and R6′ together independently form a part of a ring having one of formulae A, C-L:

where

    • each * is a bond;
    • each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)2R9′, —S(O)2R9′, or —S(O)2NR8R9;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl;
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8R9;
    • each n is an integer from 1 to 3; and
    • each X is independently S, O, NH, or N(C1-C6 alkyl).
      wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

In certain embodiments, each n is independently an integer from 1 to 2. In certain embodiments, each n is 1.

In other embodiments, the compound of Formula I′ or II′ has the following formula I″ or II″:

where compounds of formula I″ can exist in the following resonance structures I″-a, or I″-b:

and compounds of formula II″ can exist in the following resonance structures II″-a, II″-b, or II″-c:

where

    • each Y is independently O or S;
    • each A is independently CR18 or N;
    • each A′ is independently CR15R16, NR17, N, CR15, N-oxide, N—OR8—, S or O;
    • each R12 is independently F, —OR8, —SR8, —NR8R9, alkyl, or aryl;
    • each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1l-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9; each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl;
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8R9;
    • each R15 is independently a bond, H, halogen, —NR10SO2R8, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R10, —(C1-C6 alkylene)-NR9′S(O)2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryll;
    • R16 is a bond, H, halogen, —NR10SO2R8, —R8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
    • R17 is a bond, H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl; and
    • R18 is a bond, H, halogen, —NR10SO2R8, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R10, —(C1-C6 alkylene)-NR9′S(O)2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
    • each n is independently an integer from 1 to 4;
    • each m is independently an integer from 1 to 3; and
    • Z has the following structures:

where

    • each R1 is independently H, alkyl, arylalkyl, heteroarylalkyl, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl;
    • each R2 is independently H, alkyl, aryl-CH2—, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl, or R1 and R2 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R4 is independently H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
    • each R5 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R4 and R5 together form a part of a 3-8 membered heterocycloalkyl ring; and
    • each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5 and R6 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring,
      wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

In certain embodiments, each n is independently an integer from 1 to 3. In certain embodiments, each n is independently an integer from 1 to 2. In certain embodiments, each n is 1.

Each compound of Formula I′ or II′ may exist in various tautomeric forms. Accordingly, provided herein are tautomeric forms of compounds of Formula I′, for example, when R4 is H, when R4′ is H, or when R4 and R4′ are H. For example, compounds having formula I′ where R4 and R4′ are H may exist in, but not limited to, the following tautomeric forms I′a, I′b or I′c:

Provided herein are tautomeric forms of compounds of Formula II′, for example, when R4 is H, when R4′ is H, or when R4 and R4′ are H. For example, compounds having formula II where R4 and R4′ are H may exist in, but not limited to, the following tautomeric forms II′a, II′b or II′c:

In one aspect, provided herein is a compound of Formula I or II:

or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein

    • each R1 is independently H, alkyl, arylalkyl, heteroarylalkyl, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl;
    • each R2 is independently H, alkyl, aryl-CH2—, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl, or R1 and R2 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
    • each R12 is independently F, —OR8, —SR8, —NR8R9, alkyl, or aryl;
    • each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8 C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9;
    • each n is independently an integer from 1 to 4;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; and
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9,
    • wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

In one aspect, provided herein is a compound of Formula Ia, IIa or IIIa:

or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof;

    • wherein each R1 is independently H, alkyl, arylalkyl, heteroarylalkyl, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl;
    • each R2 is independently H, alkyl, aryl-CH2—, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl, or R1 and R2 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
    • each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
    • each R12 is independently F, —OR8, —SR8, —NR8R9, alkyl, or aryl; and
    • each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9;
    • each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
    • each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
    • each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; and
    • each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9,
      wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

In some embodiments, provided herein is a compound according to any of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa as described herein, or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or alkyl-cycloalkyl is unsubstituted.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl, arylalkyl, or heteroarylalkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is C1-6 alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 2-cyclopropylethyl. In certain embodiments according to Formula I, II, III, I′, II″, I″, II″, Ia, IIa, or IIIa, R1 is 3,3-dimethylbutyl. In further embodiments, R1 has one of the following structures:

In certain embodiments according to Formula II, R2 is independently hydrogen, alkyl or arylalkyl. In other embodiments, R2 is hydrogen. In further embodiments, R2 is methyl.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R6 is H, halo, —OR8, —NR8R9, —C(O)R8, alkyl, arylakyl, aryl, or heteroaryl. In other embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R6 is hydrogen or halogen. In some embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R6 is H, I, Cl, F, methyl, isobutyl, t-butyl, phenyl or benzyl. In certain embodiments according to Formula IV or IVa, R6 is (S)-tert-butyl. In other embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R6 is F. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R6 is heteroaryl. In further embodiments, R6 is heteroaryl having one of the following structures:

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa R32 is F, —OR8, —SR8, —NR8R9, alkyl, or aryl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R12 is C1-6 alkoxy. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R12 is methoxy. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R12 is ethoxy. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R12 is OH. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R12 is NH2. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R12 is —CH2-cyclopropyl, isopropyl, —CH2CH2CH2—C(O)NHCH3, —CH2CH2CH2CH2—C(O)NH2, or —CH2CH2OCH3.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R14 is H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl where R8, R9 and R10 are as defined herein. In other embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R14 is hydrogen. In some embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R14 is —NR10SO2R8 where R8 is methyl and R10 is H or alkyl such as methyl or ethyl. In some embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R14 is OCH2C(O)NR8R9 where each of R8 and R9 is independently H or alkyl.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R8 is C1-6 alkyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, or C1-6 alkyl-C3-7 cycloalkylene, each optionally substituted as described herein. In certain embodiments, R8 is C1-6 alkyl, optionally substituted as described herein. In certain embodiments, R8 is C3-7 cycloalkyl, optionally substituted as described herein. In certain embodiments, R8 is cyclopropyl, 1-methylcyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl. In certain embodiments, R8 is C6-14 aryl, optionally substituted as described herein. In certain embodiments, R8 is heteroaryl, optionally substituted as described herein. In certain embodiments, R8 is heterocyclyl, optionally substituted as described herein.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R8 is C1-6 alkyl. In certain embodiments according to Formula I, Ia, II, IIa, III or IIIa, R8 is methyl.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R9 is hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R8 and R9 together with the N atom to which they are attached form heterocyclyl.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is H, alkyl or halogen; R12 is —OR8; R14 is H or —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″,II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is halogen; R12 is —OR8; R14 is H or —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is F; R12 is —OR8; R14 is H or —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is F; R12 is —OR8; R14 is H or —NHSO2Me; and R8 is H, methyl or ethyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is C1-6 alkyl; R6 is C1-6 alkyl; R12 is —OR8; R14 is —NHSO2R8; and each R8 is independently methyl or ethyl. In certain embodiments according to this paragraph, each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or alkyl-cycloalkyl is unsubstituted.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 2-cyclopropylethyl; R2 is H; R6 is H, alkyl or halogen; R12 is —OR8; R14 is H or —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 2-cyclopropylethyl; R2 is H; R6 is halogen; R12 is —OR8; R14 is H or —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 2-cyclopropylethyl; R2 is H; R6 is F; R12 is —OR8; R14 is H or —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 2-cyclopropylethyl; R2 is H; R6 is F; R12 is —OR8; R14 is H or —NHSO2Me; and R8 is H, methyl or ethyl. In certain embodiments according to this paragraph, each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or alkyl-cycloalkyl is unsubstituted.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is H, alkyl or halogen; R12 is —OR8; R14 is H; and R8 is H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is halogen; R12 is —OR8; R14 is H; and R8 is H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is F; R12 is —OR8; R14 is H; and R8 is H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is alkyl; R2 is H; R6 is F; R12 is —OR8; R14 is H; and R8 is H, methyl or ethyl. In certain embodiments according to this paragraph, each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or alkyl-cycloalkyl is unsubstituted.

In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 3,3-dimethylbutyl; R2 is H; R6 is H, alkyl or halogen; R12 is —OR8; R14 is —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 3,3-dimethylbutyl; R2 is H; R6 is halogen; R12 is —OR8; R14 is —NHSO2R8; and each R8 is independently H or alkyl. In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 3,3-dimethylbutyl; R2 is H; R6 is F; R12 is —OR8; R14 is —NHSO2R8; and each R8 is independently H or alkyl In certain embodiments according to Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, R1 is 3,3-dimethylbutyl; R2 is H; R6 is F; R12 is —OR8; R14 is —NHSO2Me; and R8 is H, methyl or ethyl. In certain embodiments according to this paragraph, each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or alkyl-cycloalkyl is unsubstituted.

In one embodiment, provided herein is compound 1:

In yet another embodiment, provided herein is compound 2:

In yet another embodiment, provided herein is compound 3:

In yet another embodiment, provided herein is compound 4:

In yet another embodiment, provided herein is compound 5:

In yet another embodiment, provided herein is compound 6:

In yet another embodiment, provided herein is compound 7:

In yet another embodiment, provided herein is compound 8:

In yet another embodiment, provided herein is compound 9:

In yet another embodiment, provided herein is compound 10:

In yet another embodiment, provided herein is compound 11:

In yet another embodiment, provided herein is compound 12:

In yet another embodiment, provided herein is compound 13:

In yet another embodiment, provided herein is compound 14:

In yet another embodiment, provided herein is compound 15:

In certain embodiments, provided herein are the following compounds according to formulae I-1 to I-20:

In certain embodiments, provided herein are the following compounds according to formulae I-21 to I-40:

In certain embodiments, provided herein are the following compounds according to formulae I-41 to I-60:

In certain embodiments, provided herein are the following compounds according to formulae I-61 to I-73:

In certain embodiments, provided herein are the following compounds according to formulae II-1 to II-20:

In certain embodiments, provided herein are the following compounds according to formulae II-21 to II-40:

In certain embodiments, provided herein are the following compounds according to formulae II-41 to II-48:

In certain embodiments, provided herein are the following compounds according to formulae III-1 to III-16:

The compounds provided herein are intended to encompass all possible stereoisomers, unless a particular stereochemistry is specified. Where the compound provided herein contains an alkenyl or alkenylene group, the compound may exist as one or mixture of geometric cis/trans (or Z/E) isomers. Where structural isomers are interconvertible via a low energy barrier, the compound may exist as a single tautomer or a mixture of tautomers. This can take the form of proton tautomerism in the compound that contains, for example, an imino, keto, or oxime group; or so-called valence tautomerism in the compound that contain an aromatic moiety. It follows that a single compound may exhibit more than one type of isomerism.

The compounds provided herein may be enantiomerically pure, such as a single enantiomer or a single diastereomer, or be stereoisomeric mixtures, such as a mixture of an enantiomeric pair, a racemic mixture, or a diastereomeric mixture. As such, one of skill in the art will recognize that administration of a compound in its (R) form is equivalent, for compounds that undergo epimerization in vivo, to administration of the compound in its (S) form. Conventional techniques for the preparation/isolation of individual enantiomers include synthesis from a suitable optically pure precursor, asymmetric synthesis from achiral starting materials, or resolution of an enantiomeric mixture, for example, chiral chromatography, recrystallization, resolution, diastereomeric salt formation, or derivatization into diastereomeric adducts followed by separation.

When the compound provided herein contains an acidic or basic moiety, it may also be provided as a pharmaceutically acceptable salt (See, Berge et al., J. Pharm. Sci. 1977, 66, 1-19; and “Handbook of Pharmaceutical Salts, Properties, and Use,” Stahl and Wermuth, Ed.; Wiley-VCH and VHCA, Zurich, 2002).

Suitable acids for use in the preparation of pharmaceutically acceptable salts include, but are not limited to, acetic acid, 2,2-dichloroacetic acid, acylated amino acids, adipic acid, alginic acid, ascorbic acid, L-aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, boric acid, (+)-camphoric acid, camphorsulfonic acid, (+)-(1S)-camphor-10-sulfonic acid, capric acid, caproic acid, caprylic acid, cinnamic acid, citric acid, cyclamic acid, cyclohexanesulfamic acid, dodecylsulfuric acid, ethane-1,2-disulfonic acid, ethanesulfonic acid, 2-hydroxy-ethanesulfonic acid, formic acid, fumaric acid, galactaric acid, gentisic acid, glucoheptonic acid, D-gluconic acid, D-glucuronic acid, L-glutamic acid, α-oxoglutaric acid, glycolic acid, hippuric acid, hydrobromic acid, hydrochloric acid, hydroiodic acid, (+)-L-lactic acid, (±)-DL-lactic acid, lactobionic acid, lauric acid, maleic acid, (−)-L-malic acid, malonic acid, (±)-DL-mandelic acid, methanesulfonic acid, naphthalene-2-sulfonic acid, naphthalene-1,5-disulfonic acid, 1-hydroxy-2-naphthoic acid, nicotinic acid, nitric acid, oleic acid, orotic acid, oxalic acid, palmitic acid, pamoic acid, perchloric acid, phosphoric acid, L-pyroglutamic acid, saccharic acid, salicylic acid, 4-amino-salicylic acid, sebacic acid, stearic acid, succinic acid, sulfuric acid, tannic acid, (+)-L-tartaric acid, thiocyanic acid, p-toluenesulfonic acid, undecylenic acid, and valeric acid.

Suitable bases for use in the preparation of pharmaceutically acceptable salts, including, but not limited to, inorganic bases, such as magnesium hydroxide, calcium hydroxide, potassium hydroxide, zinc hydroxide, or sodium hydroxide; and organic bases, such as primary, secondary, tertiary, and quaternary, aliphatic and aromatic amines, including L-arginine, benethamine, benzathine, choline, deanol, diethanolamine, diethylamine, dimethylamine, dipropylamine, diisopropylamine, 2-(diethylamino)-ethanol, ethanolamine, ethylamine, ethylenediamine, isopropylamine, N-methyl-glucamine, hydrabamine, 1H-imidazole, L-lysine, morpholine, 4-(2-hydroxyethyl)-morpholine, methylamine, piperidine, piperazine, propylamine, pyrrolidine, 1-(2-hydroxyethyl)-pyrrolidine, pyridine, quinuclidine, quinoline, isoquinoline, secondary amines, triethanolamine, trimethylamine, triethylamine, N-methyl-D-glucamine, 2-amino-2-(hydroxymethyl)-1,3-propanediol, and tromethamine.

The compound provided herein may also be provided as a prodrug, which is a functional derivative of the compound, for example, of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa and is readily convertible into the parent compound in vivo. Prodrugs are often useful because, in some situations, they may be easier to administer than the parent compound. They may, for instance, be bioavailable by oral administration whereas the parent compound is not. The prodrug may also have enhanced solubility in pharmaceutical compositions over the parent compound. A prodrug may be converted into the parent drug by various mechanisms, including enzymatic processes and metabolic hydrolysis. See Harper, Progress in Drug Research 1962, 4, 221-294; Morozowich et al. in “Design of Biopharmaceutical Properties through Prodrugs and Analogs,” Roche Ed., APHA Acad. Pharm. Sci. 1977; “Bioreversible Carriers in Drug in Drug Design, Theory and Application,” Roche Ed., APHA Acad. Pharm. Sci. 1987; “Design of Prodrugs,” Bundgaard, Elsevier, 1985; Wang et al., Curr. Pharm. Design 1999, 5, 265-287; Pauletti et al., Adv. Drug. Delivery Rev. 1997, 27, 235-256; Mizen et al., Pharm. Biotech. 1998, 11, 345-365; Gaignault et al., Pract. Med. Chem. 1996, 671-696; Asgharnejad in “Transport Processes in Pharmaceutical Systems,” Amidon et al., Ed., Marcell Dekker, 185-218, 2000; Balant et al., Eur. J. Drug Metab. Pharmacokinet. 1990, 15, 143-53; Balimane and Sinko, Adv. Drug Delivery Rev. 1999, 39, 183-209; Browne, Clin. Neuropharmacol. 1997, 20, 1-12; Bundgaard, Arch. Pharm. Chem. 1979, 86, 1-39; Bundgaard, Controlled Drug Delivery 1987, 17, 179-96; Bundgaard, Adv. Drug Delivery Rev. 1992, 8, 1-38; Fleisher et al., Adv. Drug Delivery Rev. 1996, 19, 115-130; Fleisher et al., Methods Enzymol. 1985, 112, 360-381; Farquhar et al., J. Pharm. Sci. 1983, 72, 324-325; Freeman et al., J. Chem. Soc., Chem. Commun. 1991, 875-877; Friis and Bundgaard, Eur. J. Pharm. Sci. 1996, 4, 49-59; Gangwar et al., Des. Biopharm. Prop. Prodrugs Analogs, 1977, 409-421; Nathwani and Wood, Drugs 1993, 45, 866-94; Sinhababu and Thakker, Adv. Drug Delivery Rev. 1996, 19, 241-273; Stella et al., Drugs 1985, 29, 455-73; Tan et al., Adv. Drug Delivery Rev. 1999, 39, 117-151; Taylor, Adv. Drug Delivery Rev. 1996, 19, 131-148; Valentino and Borchardt, Drug Discovery Today 1997, 24 148-155; Wiebe and Knaus, Adv. Drug Delivery Rev. 1999, 39, 63-80; and Waller et al., Br. J. Clin. Pharmac. 1989, 28, 497-507.

Methods of Synthesis

The compound provided herein can be prepared, isolated, or obtained by any method known to one of skill in the art. For an example, a compound of Formula I can be prepared as shown in Scheme 1.

In certain embodiments, anthranilic acid 1A, or a derivative thereof, can be cyclized with carbonate in the presence of, for example, triphosgene, to form bicyclic compound 1B. Bicyclic compound 1B can be coupled with alcohol RBOH to form bicyclic compound 1C. Bicyclic compound 1C can be reacted with ethylcyanoacetate to form bicyclic carbonitrile 1D. Bicyclic carbonitrile 1D can be coupled with a bromoaniline to form compound 1E. Compound 1E can be coupled with triethylphosphite to form compound 1F. Compound 1F can be cyclized, for example, in dimethylacetamide with heat to form phosphadiazine 1G. The ethyl group can be removed from the phosphadiazine group of compound 1G to yield hydroxyphosphadiazine compound 1H. Hydroxyphosphadiazine compound 1H can be coupled with a variety RD compounds to form further phosphadiazine derivatives, such as aminophosphadiazine compounds. Protecting groups can be used where suitable according to the judgment of one of skill in the art.

An alternative strategy is shown in Scheme 2. In certain embodiments, compound 2D can be coupled with a diethyl (1-aminophenyl)phosphonate to yield phosphadiazine compound 2K. The ethyl group can be removed from the phosphadiazine group of compound 2K to yield hydroxyphosphadiazine compound 2L. Hydroxyphosphadiazine compound 2L can be coupled with a variety RD compounds to form further phosphadiazine derivatives, such as methoxyphosphadiazine compounds 2M. Protecting groups can be used where suitable according to the judgment of one of skill in the art.

The starting materials used in the synthesis of the compounds provided herein are either commercially available or can be readily prepared. For example, the preparation of 5-fluoroanthranilic acid has been described in U.S. Pat. No. 5,523,472, the contents of which are hereby incorporated by reference in their entirety. Diethyl (1-aminophenyl)phosphonates can be prepared from 2-iodo-4-nitroaniline, commercially available (e.g. Sigma-Aldrich, Interchim intermediates, Aurora screening library, or as described in Tetrahedron Letters, 2004, 45(46):8569-8573, the contents of which are hereby incorporated by reference in their entirety), as described in the examples below.

A compound of Formula II can be prepared as shown in Scheme 3.

In certain embodiments, compounds 3A-3F can be prepared as described, for example, in U.S. 2005/0107364 A1, WO 2005/019191, or Krueger et al., 2007, Bioorg. Med. Chem. Lett. 17:2289-2292, the contents of each of which are hereby incorporated by reference in their entirety. Compound 3F can be cyclized in the presence of, for example, acid, to form bicyclic compound 3G. Bicyclic compound 3G can be reacted with a tris(alkylthio)methyl compound to form compound 3H as described in the references above. Compound 3H can be reacted with the depicted phosphoaniline compound Diethyl (1-aminophenyl)phosphonates can be prepared from 2-iodo-4-nitroaniline, commercially available (e.g. Sigma-Aldrich, Interchim intermediates, Aurora screening library, or as described in Tetrahedron Letters, 2004, 45(46):8569-8573, the contents of which are hereby incorporated by reference in their entirety) to form compound 3I. Compound 3I can be reacted with ammonia gas to form compound 3J. Compound 3J can be cyclized with AlMe3 to form phosphadiazine compound 3J. The ethyl group can be removed from the phosphadiazine group of compound 3K to yield hydroxyphosphadiazine compound 3L. Hydroxyphosphadiazine compound 3L can be coupled with a variety RD compounds to form further phosphadiazine derivatives, such as aminophosphadiazine compounds. Protecting groups can be used where suitable according to the judgment of one of skill in the art.

A compound of Formula III can be prepared as shown in Scheme 4.

In certain embodiments, 1H-2-pyridone 4A, or a derivative thereof, can be reacted with RBI in the presence of potassium carbonate and acetonitrile to form 2-pyridone 4B. 2-Pyridone 4B, or a derivative thereof, can react with a base in the presence of, for example, dioxane, to form 4-hydroxy-2-pyridone 4C. 4-Hydroxy-2-pyridone 4C can react with sodium iodide symporter (NIS) in the presence of, for example, trifluoroacetic acid and acetonitrile, to form 5-iodo-4-hydroxy-2-pyridone 4D. 5-Iodo-4-hydroxy-2-pyridone 4D can couple with compound J in the presence of, for example, trimethylaluminum and dioxane, to form phosphadiazine 4E. Phosphadiazine 4E can react with a variety of RA—X compounds in the presence of, for example, sodium carbonate, tetrahydrofuran, and a catalyst, to form phosphadiazine 4F.

A compound of Formula Ill can also be prepared as shown in Scheme 5.

In certain embodiments, 1H-2-pyridone 5A, or a derivative thereof, can be reacted with RBI in the presence of potassium carbonate and acetonitrile to form 2-pyridone 5B. 2-Pyridone 5B, or a derivative thereof, can react with a base in the presence of, for example, dioxane, to form 4-hydroxy-2-pyridone 5C. 4-Hydroxy-2-pyridone 5C can react with sodium iodide symporter (NIS) in the presence of, for example, trifluoroacetic acid and acetonitrile, to form 5-iodo-4-hydroxy-2-pyridone 5D. 5-Iodo-4-hydroxy-2-pyridone 5D can react with RA-X in the presence of, for example, cesium carbonate, copper (I) iodide, and dimethylformamide, to form compound 5E, compound 5E can couple with compound J in the presence of, for example, trimethylaluminum and dioxane, to form compound 5F.

Pharmaceutical Compositions

Provided herein are pharmaceutical compositions comprising a compound provided herein as an active ingredient, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, in a pharmaceutically acceptable vehicle, carrier, diluent, or excipient, or a mixture thereof; in combination with one or more pharmaceutically acceptable excipients or carriers. In certain embodiments, the pharmaceutical composition comprises at least one release controlling excipient or carrier. In certain embodiments, the pharmaceutical composition comprises at least one nonrelease controlling excipient or carrier. In certain embodiments, the pharmaceutical composition comprises at least one release controlling and at least one nonrelease controlling excipients or carriers.

The compound provided herein may be administered alone, or in combination with one or more other compounds provided herein, one or more other active ingredients. The pharmaceutical compositions that comprise a compound provided herein may be formulated in various dosage forms for oral, parenteral, and topical administration. The pharmaceutical compositions may also be formulated as a modified release dosage form, including delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. These dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Modified-Release Drug Deliver Technology, Rathbone et al., Eds., Drugs and the Pharmaceutical Science, Marcel Dekker, Inc.: New York, N.Y., 2003; Vol. 126).

In one embodiment, the pharmaceutical compositions are provided in a dosage form for oral administration, which comprise a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.

In another embodiment, the pharmaceutical compositions are provided in a dosage form for parenteral administration, which comprise a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.

In yet another embodiment, the pharmaceutical compositions are provided in a dosage form for topical administration, which comprise a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof; and one or more pharmaceutically acceptable excipients or carriers.

The pharmaceutical compositions provided herein may be provided in unit-dosage forms or multiple-dosage forms. Unit-dosage forms, as used herein, refer to physically discrete units suitable for administration to human and animal subjects and packaged individually as is known in the art. Each unit-dose contains a predetermined quantity of the active ingredient(s) sufficient to produce the desired therapeutic effect, in association with the required pharmaceutical carriers or excipients. Examples of unit-dosage forms include ampoules, syringes, and individually packaged tablets and capsules. Unit-dosage forms may be administered in fractions or multiples thereof. A multiple-dosage form is a plurality of identical unit-dosage forms packaged in a single container to be administered in segregated unit-dosage form. Examples of multiple-dosage forms include vials, bottles of tablets or capsules, or bottles of pints or gallons.

The pharmaceutical compositions provided herein may be administered at once, or multiple times at intervals of time. It is understood that the precise dosage and duration of treatment may vary with the age, weight, and condition of the patient being treated, and may be determined empirically using known testing protocols or by extrapolation from in vivo or in vitro test or diagnostic data. It is further understood that for any particular individual, specific dosage regimens should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the formulations.

A. Oral Administration

The pharmaceutical compositions provided herein may be provided in solid, semisolid, or liquid dosage forms for oral administration. As used herein, oral administration also include buccal, lingual, and sublingual administration. Suitable oral dosage forms include, but are not limited to, tablets, capsules, pills, troches, lozenges, pastilles, cachets, pellets, medicated chewing gum, granules, bulk powders, effervescent or non-effervescent powders or granules, solutions, emulsions, suspensions, solutions, wafers, sprinkles, elixirs, and syrups. In addition to the active ingredient(s), the pharmaceutical compositions may contain one or more pharmaceutically acceptable carriers or excipients, including, but not limited to, binders, fillers, diluents, disintegrants, wetting agents, lubricants, glidants, coloring agents, dye-migration inhibitors, sweetening agents, and flavoring agents.

Binders or granulators impart cohesiveness to a tablet to ensure the tablet remaining intact after compression. Suitable binders or granulators include, but are not limited to, starches, such as corn starch, potato starch, and pre-gelatinized starch (e.g., STARCH 1500); gelatin; sugars, such as sucrose, glucose, dextrose, molasses, and lactose; natural and synthetic gums, such as acacia, alginic acid, alginates, extract of Irish moss, panwar gum, ghatti gum, mucilage of isabgol husks, carboxymethylcellulose, methylcellulose, polyvinylpyrrolidone (PVP), Veegum, larch arabogalactan, powdered tragacanth, and guar gum; celluloses, such as ethyl cellulose, cellulose acetate, carboxymethyl cellulose calcium, sodium carboxymethyl cellulose, methyl cellulose, hydroxyethylcellulose (HEC), hydroxypropylcellulose (HPC), hydroxypropyl methyl cellulose (HPMC); microcrystalline celluloses, such as AVICEL-PH-101, AVICEL-PH-103, AVICEL RC-581, AVICEL-PH-105 (FMC Corp., Marcus Hook, Pa.); and mixtures thereof. Suitable fillers include, but are not limited to, talc, calcium carbonate, microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof The binder or filler may be present from about 50 to about 99% by weight in the pharmaceutical compositions provided herein.

Suitable diluents include, but are not limited to, dicalcium phosphate, calcium sulfate, lactose, sorbitol, sucrose, inositol, cellulose, kaolin, mannitol, sodium chloride, dry starch, and powdered sugar. Certain diluents, such as mannitol, lactose, sorbitol, sucrose, and inositol, when present in sufficient quantity, can impart properties to some compressed tablets that permit disintegration in the mouth by chewing. Such compressed tablets can be used as chewable tablets.

Suitable disintegrants include, but are not limited to, agar; bentonite; celluloses, such as methylcellulose and carboxymethylcellulose; wood products; natural sponge; cation-exchange resins; alginic acid; gums, such as guar gum and Veegum HV; citrus pulp; cross-linked celluloses, such as croscarmellose; cross-linked polymers, such as crospovidone; cross-linked starches; calcium carbonate; microcrystalline cellulose, such as sodium starch glycolate; polacrilin potassium; starches, such as corn starch, potato starch, tapioca starch, and pre-gelatinized starch; clays; aligns; and mixtures thereof. The amount of a disintegrant in the pharmaceutical compositions provided herein varies upon the type of formulation, and is readily discernible to those of ordinary skill in the art. The pharmaceutical compositions provided herein may contain from about 0.5 to about 15% or from about 1 to about 5% by weight of a disintegrant.

Suitable lubricants include, but are not limited to, calcium stearate; magnesium stearate; mineral oil; light mineral oil; glycerin; sorbitol; mannitol; glycols, such as glycerol behenate and polyethylene glycol (PEG); stearic acid; sodium lauryl sulfate; talc; hydrogenated vegetable oil, including peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil; zinc stearate; ethyl oleate; ethyl laureate; agar; starch; lycopodium; silica or silica gels, such as AEROSIL® 200 (W.R. Grace Co., Baltimore, Md.) and CAB-O-SIL® (Cabot Co. of Boston, Mass.); and mixtures thereof. The pharmaceutical compositions provided herein may contain about 0.1 to about 5% by weight of a lubricant.

Suitable glidants include colloidal silicon dioxide, CAB-O-SIL® (Cabot Co. of Boston, Mass.), and asbestos-free talc. Coloring agents include any of the approved, certified, water soluble FD&C dyes, and water insoluble FD&C dyes suspended on alumina hydrate, and color lakes and mixtures thereof. A color lake is the combination by adsorption of a water-soluble dye to a hydrous oxide of a heavy metal, resulting in an insoluble form of the dye. Flavoring agents include natural flavors extracted from plants, such as fruits, and synthetic blends of compounds which produce a pleasant taste sensation, such as peppermint and methyl salicylate. Sweetening agents include sucrose, lactose, mannitol, syrups, glycerin, and artificial sweeteners, such as saccharin and aspartame. Suitable emulsifying agents include gelatin, acacia, tragacanth, bentonite, and surfactants, such as polyoxyethylene sorbitan monooleate (TUREEN® 20), polyoxyethylene sorbitan monooleate 80 (TWEEN® 80), and triethanolamine oleate. Suspending and dispersing agents include sodium carboxymethylcellulose, pectin, tragacanth, Veegum, acacia, sodium carbomethylcellulose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Preservatives include glycerin, methyl and propylparaben, benzoic add, sodium benzoate and alcohol. Wetting agents include propylene glycol monostearate, sorbitan monooleate, diethylene glycol monolaurate, and polyoxyethylene lauryl ether. Solvents include glycerin, sorbitol, ethyl alcohol, and syrup. Examples of non-aqueous liquids utilized in emulsions include mineral oil and cottonseed oil. Organic acids include citric and tartaric acid. Sources of carbon dioxide include sodium bicarbonate and sodium carbonate.

It should be understood that many carriers and excipients may serve several functions, even within the same formulation.

The pharmaceutical compositions provided herein may be provided as compressed tablets, tablet triturates, chewable lozenges, rapidly dissolving tablets, multiple compressed tablets, or enteric-coating tablets, sugar-coated, or film-coated tablets. Enteric-coated tablets are compressed tablets coated with substances that resist the action of stomach acid but dissolve or disintegrate in the intestine, thus protecting the active ingredients from the acidic environment of the stomach. Enteric-coatings include, but are not limited to, fatty acids, fats, phenyl salicylate, waxes, shellac, ammoniated shellac, and cellulose acetate phthalates. Sugar-coated tablets are compressed tablets surrounded by a sugar coating, which may be beneficial in covering up objectionable tastes or odors and in protecting the tablets from oxidation. Film-coated tablets are compressed tablets that are covered with a thin layer or film of a water-soluble material. Film coatings include, but are not limited to, hydroxyethylcellulose, sodium carboxymethylcellulose, polyethylene glycol 4000, and cellulose acetate phthalate. Film coating imparts the same general characteristics as sugar coating. Multiple compressed tablets are compressed tablets made by more than one compression cycle, including layered tablets, and press-coated or dry-coated tablets.

The tablet dosage forms may be prepared from the active ingredient in powdered, crystalline, or granular forms, alone or in combination with one or more carriers or excipients described herein, including binders, disintegrants, controlled-release polymers, lubricants, diluents, and/or colorants. Flavoring and sweetening agents are especially useful in the formation of chewable tablets and lozenges.

The pharmaceutical compositions provided herein may be provided as soft or hard capsules, which can be made from gelatin, methylcellulose, starch, or calcium alginate. The hard gelatin capsule, also known as the dry-filled capsule (DFC), consists of two sections, one slipping over the other, thus completely enclosing the active ingredient. The soft elastic capsule (SEC) is a soft, globular shell, such as a gelatin shell, which is plasticized by the addition of glycerin, sorbitol, or a similar polyol. The soft gelatin shells may contain a preservative to prevent the growth of microorganisms. Suitable preservatives are those as described herein, including methyl- and propyl-parabens, and sorbic acid. The liquid, semisolid, and solid dosage forms provided herein may be encapsulated in a capsule. Suitable liquid and semisolid dosage forms include solutions and suspensions in propylene carbonate, vegetable oils, or triglycerides. Capsules containing such solutions can be prepared as described in U.S. Pat. Nos. 4,328,245; 4,409,239; and 4,410,545. The capsules may also be coated as known by those of skill in the art in order to modify or sustain dissolution of the active ingredient.

The pharmaceutical compositions provided herein may be provided in liquid and semisolid dosage forms, including emulsions, solutions, suspensions, elixirs, and syrups. An emulsion is a two-phase system, in which one liquid is dispersed in the form of small globules throughout another liquid, which can be oil-in-water or water-in-oil. Emulsions may include a pharmaceutically acceptable non-aqueous liquids or solvent, emulsifying agent, and preservative. Suspensions may include a pharmaceutically acceptable suspending agent and preservative. Aqueous alcoholic solutions may include a pharmaceutically acceptable acetal, such as a di(lower alkyl) acetal of a lower alkyl aldehyde, e.g., acetaldehyde diethyl acetal; and a water-miscible solvent having one or more hydroxyl groups, such as propylene glycol and ethanol. Elixirs are clear, sweetened, and hydroalcoholic solutions. Syrups are concentrated aqueous solutions of a sugar, for example, sucrose, and may also contain a preservative. For a liquid dosage form, for example, a solution in a polyethylene glycol may be diluted with a sufficient quantity of a pharmaceutically acceptable liquid carrier, e.g., water, to be measured conveniently for administration.

Other useful liquid and semisolid dosage forms include, but are not limited to, those containing the active ingredient(s) provided herein, and a dialkylated mono- or poly-alkylene glycol, including, 1,2-dimethoxymethane, diglyme, triglyme, tetraglyme, polyethylene glycol-350-dimethyl ether, polyethylene glycol-550-dimethyl ether, polyethylene glycol-750-dimethyl ether, wherein 350, 550, and 750 refer to the approximate average molecular weight of the polyethylene glycol. These formulations may further comprise one or more antioxidants, such as butylated hydroxytoluene (BHT), butylated hydroxyanisole (BHA), propyl gallate, vitamin E, hydroquinone, hydroxycoumarins, ethanolamine, lecithin, cephalin, ascorbic acid, malic acid, sorbitol, phosphoric acid, bisulfite, sodium metabisulfite, thiodipropionic acid and its esters, and dithiocarbamates.

The pharmaceutical compositions provided herein for oral administration may be also provided in the forms of liposomes, micelles, microspheres, or nanosystems. Micellar dosage forms can be prepared as described in U.S. Pat. No. 6,350,458.

The pharmaceutical compositions provided herein may be provided as non-effervescent or effervescent, granules and powders, to be reconstituted into a liquid dosage form. Pharmaceutically acceptable carriers and excipients used in the non-effervescent granules or powders may include diluents, sweeteners, and wetting agents. Pharmaceutically acceptable carriers and excipients used in the effervescent granules or powders may include organic acids and a source of carbon dioxide.

Coloring and flavoring agents can be used in all of the above dosage forms.

The pharmaceutical compositions provided herein may be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.

The pharmaceutical compositions provided herein may be co-formulated with other active ingredients which do not impair the desired therapeutic action, or with substances that supplement the desired action, such as drotrecogin-α, and hydrocortisone.

B. Parenteral Administration

The pharmaceutical compositions provided herein may be administered parenterally by injection, infusion, or implantation, for local or systemic administration. Parenteral administration, as used herein, include intravenous, intraarterial, intraperitoneal, intrathecal, intraventricular, intraurethral, intrasternal, intracranial, intramuscular, intrasynovial, and subcutaneous administration.

The pharmaceutical compositions provided herein may be formulated in any dosage forms that are suitable for parenteral administration, including solutions, suspensions, emulsions, micelles, liposomes, microspheres, nanosystems, and solid forms suitable for solutions or suspensions in liquid prior to injection. Such dosage forms can be prepared according to conventional methods known to those skilled in the art of pharmaceutical science (see, Remington: The Science and Practice of Pharmacy, supra).

The pharmaceutical compositions intended for parenteral administration may include one or more pharmaceutically acceptable carriers and excipients, including, but not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, cryoprotectants, lyoprotectants, thickening agents, pH adjusting agents, and inert gases.

Suitable aqueous vehicles include, but are not limited to, water, saline, physiological saline or phosphate buffered saline (PBS), sodium chloride injection, Ringers injection, isotonic dextrose injection, sterile water injection, dextrose and lactated Ringers injection. Non-aqueous vehicles include, but are not limited to, fixed oils of vegetable origin, castor oil, corn oil, cottonseed oil, olive oil, peanut oil, peppermint oil, safflower oil, sesame oil, soybean oil, hydrogenated vegetable oils, hydrogenated soybean oil, and medium-chain triglycerides of coconut oil, and palm seed oil. Water-miscible vehicles include, but are not limited to, ethanol, 1,3-butanediol, liquid polyethylene glycol (e.g., polyethylene glycol 300 and polyethylene glycol 400), propylene glycol, glycerin, N-methyl-2-pyrrolidone, N,N-dimethylacetamide, and dimethyl sulfoxide.

Suitable antimicrobial agents or preservatives include, but are not limited to, phenols, cresols, mercurials, benzyl alcohol, chlorobutanol, methyl and propyl p-hydroxybenzoates, thimerosal, benzalkonium chloride (e.g., benzethonium chloride), methyl- and propyl-parabens, and sorbic acid. Suitable isotonic agents include, but are not limited to, sodium chloride, glycerin, and dextrose. Suitable buffering agents include, but are not limited to, phosphate and citrate. Suitable antioxidants are those as described herein, including bisulfite and sodium metabisulfite. Suitable local anesthetics include, but are not limited to, procaine hydrochloride. Suitable suspending and dispersing agents are those as described herein, including sodium carboxymethylcelluose, hydroxypropyl methylcellulose, and polyvinylpyrrolidone. Suitable emulsifying agents include those described herein, including polyoxyethylene sorbitan monolaurate, polyoxyethylene sorbitan monooleate 80, and triethanolamine oleate. Suitable sequestering or chelating agents include, but are not limited to EDTA. Suitable pH adjusting agents include, but are not limited to, sodium hydroxide, hydrochloric acid, citric acid, and lactic acid. Suitable complexing agents include, but are not limited to, cyclodextrins, including α-cyclodextrin, β-cyclodextrin, hydroxypropyl-β-cyclodextrin, sulfobutylether-β-cyclodextrin, and sulfobutylether 7-β-cyclodextrin (CAPTISOL®, CyDex, Lenexa, Kans.).

The pharmaceutical compositions provided herein may be formulated for single or multiple dosage administration. The single dosage formulations are packaged in an ampoule, a vial, or a syringe. The multiple dosage parenteral formulations must contain an antimicrobial agent at bacteriostatic or fungistatic concentrations. All parenteral formulations must be sterile, as known and practiced in the art.

In one embodiment, the pharmaceutical compositions are provided as ready-to-use sterile solutions. In another embodiment, the pharmaceutical compositions are provided as sterile dry soluble products, including lyophilized powders and hypodermic tablets, to be reconstituted with a vehicle prior to use. In yet another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile suspensions. In yet another embodiment, the pharmaceutical compositions are provided as sterile dry insoluble products to be reconstituted with a vehicle prior to use. In still another embodiment, the pharmaceutical compositions are provided as ready-to-use sterile emulsions.

The pharmaceutical compositions provided herein may be formulated as immediate or modified release dosage forms, including delayed-, sustained, pulsed-, controlled, targeted-, and programmed-release forms.

The pharmaceutical compositions may be formulated as a suspension, solid, semi-solid, or thixotropic liquid, for administration as an implanted depot. In one embodiment, the pharmaceutical compositions provided herein are dispersed in a solid inner matrix, which is surrounded by an outer polymeric membrane that is insoluble in body fluids but allows the active ingredient in the pharmaceutical compositions diffuse through.

Suitable inner matrixes include polymethylmethacrylate, polybutylmethacrylate, plasticized or unplasticized polyvinylchloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, polyisoprene, polyisobutylene, polybutadiene, polyethylene, ethylene-vinyl acetate copolymers, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, hydrophilic polymers, such as hydrogels of esters of acrylic and methacrylic acid, collagen, cross-linked polyvinyl alcohol, and cross-linked partially hydrolyzed polyvinyl acetate.

Suitable outer polymeric membranes include polyethylene, polypropylene, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, ethylene/vinyl acetate copolymers, silicone rubbers, polydimethyl siloxanes, neoprene rubber, chlorinated polyethylene, polyvinylchloride, vinyl chloride 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.

C. Topical Administration

The pharmaceutical compositions provided herein may be administered topically to the skin, orifices, or mucosa. The topical administration, as used herein, includes (intra)dermal, conjunctival, intracorneal, intraocular, ophthalmic, auricular, transdermal, nasal, vaginal, urethral, respiratory, and rectal administration.

The pharmaceutical compositions provided herein may be formulated in any dosage forms that are suitable for topical administration for local or systemic effect, including emulsions, solutions, suspensions, creams, gels, hydrogels, ointments, dusting powders, dressings, elixirs, lotions, suspensions, tinctures, pastes, foams, films, aerosols, irrigations, sprays, suppositories, bandages, dermal patches. The topical formulation of the pharmaceutical compositions provided herein may also comprise liposomes, micelles, microspheres, nanosystems, and mixtures thereof.

Pharmaceutically acceptable carriers and excipients suitable for use in the topical formulations provided herein include, but are not limited to, aqueous vehicles, water-miscible vehicles, non-aqueous vehicles, antimicrobial agents or preservatives against the growth of microorganisms, stabilizers, solubility enhancers, isotonic agents, buffering agents, antioxidants, local anesthetics, suspending and dispersing agents, wetting or emulsifying agents, complexing agents, sequestering or chelating agents, penetration enhancers, cryoprotectants, lyoprotectants, thickening agents, and inert gases.

The pharmaceutical compositions may also be administered topically by electroporation, iontophoresis, phonophoresis, sonophoresis, or microneedle or needle-free injection, such as POWDERJECT™ (Chiron Corp., Emeryville, Calif.), and BIOJECT™ (Bioject Medical Technologies Inc., Tualatin, Oreg.).

The pharmaceutical compositions provided herein may be provided in the forms of ointments, creams, and gels. Suitable ointment vehicles include oleaginous or hydrocarbon vehicles, including such as lard, benzoinated lard, olive oil, cottonseed oil, and other oils, white petrolatum; emulsifiable or absorption vehicles, such as hydrophilic petrolatum, hydroxystearin sulfate, and anhydrous lanolin; water-removable vehicles, such as hydrophilic ointment; water-soluble ointment vehicles, including polyethylene glycols of varying molecular weight; emulsion vehicles, either water-in-oil (W/O) emulsions or oil-in-water (O/W) emulsions, including cetyl alcohol, glyceryl monostearate, lanolin, and stearic acid (see, Remington: The Science and Practice of Pharmacy, supra). These vehicles are emollient but generally require addition of antioxidants and preservatives.

Suitable cream base can be oil-in-water or water-in-oil. Cream vehicles may be water-washable, and contain an oil phase, an emulsifier, and an aqueous phase. The oil phase is also called the “internal” phase, which is generally comprised of petrolatum and a fatty alcohol such as cetyl or stearyl alcohol. The aqueous phase usually, although not necessarily, exceeds the oil phase in volume, and generally contains a humectant. The emulsifier in a cream formulation may be a nonionic, anionic, cationic, or amphoteric surfactant.

Gels are semisolid, suspension-type systems. Single-phase gels contain organic macromolecules distributed substantially uniformly throughout the liquid carrier. Suitable gelling agents include crosslinked acrylic acid polymers, such as carbomers, carboxypolyalkylenes, CARBOPOL®; hydrophilic polymers, such as polyethylene oxides, polyoxyethylene-polyoxypropylene copolymers, and polyvinylalcohol; cellulosic polymers, such as hydroxypropyl cellulose, hydroxyethyl cellulose, hydroxypropyl methylcellulose, hydroxypropyl methylcellulose phthalate, and methylcellulose; gums, such as tragacanth and xanthan gum; sodium alginate; and gelatin. In order to prepare a uniform gel, dispersing agents such as-alcohol or glycerin can be added, or the gelling agent can be dispersed by trituration, mechanical mixing, and/or stirring.

The pharmaceutical compositions provided herein may be administered rectally, urethrally, vaginally, or perivaginally in the forms of suppositories, pessaries, bougies, poultices or cataplasm, pastes, powders, dressings, creams, plasters, contraceptives, ointments, solutions, emulsions, suspensions, tampons, gels, foams, sprays, or enemas. These dosage forms can be manufactured using conventional processes as described in Remington: The Science and Practice of Pharmacy, supra.

Rectal, urethral, and vaginal suppositories are solid bodies for insertion into body orifices, which are solid at ordinary temperatures but melt or soften at body temperature to release the active ingredient(s) inside the orifices. Pharmaceutically acceptable carriers utilized in rectal and vaginal suppositories include bases or vehicles, such as stiffening agents, which produce a melting point in the proximity of body temperature, when formulated with the pharmaceutical compositions provided herein; and antioxidants as described herein, including bisulfite and sodium metabisulfite. Suitable vehicles include, but are not limited to, cocoa butter (theobroma oil), glycerin-gelatin, carbowax (polyoxyethylene glycol), spermaceti, paraffin, white and yellow wax, and appropriate mixtures of mono-, di- and triglycerides of fatty acids, hydrogels, such as polyvinyl alcohol, hydroxyethyl methacrylate, polyacrylic acid; glycerinated gelatin. Combinations of the various vehicles may be used. Rectal and vaginal suppositories may be prepared by the compressed method or molding. The typical weight of a rectal and vaginal suppository is about 2 to about 3 g.

The pharmaceutical compositions provided herein may be administered ophthalmically in the forms of solutions, suspensions, ointments, emulsions, gel-forming solutions, powders for solutions, gels, ocular inserts, and implants.

The pharmaceutical compositions provided herein may be administered intranasally or by inhalation to the respiratory tract. The pharmaceutical compositions may be provided in the form of an aerosol or solution for delivery using a pressurized container, pump, spray, atomizer, such as an atomizer using electrohydrodynamics to produce a fine mist, or nebulizer, alone or in combination with a suitable propellant, such as 1,1,1,2-tetrafluoroethane or 1,1,1,2,3,3,3-heptafluoropropane. The pharmaceutical compositions may also be provided as a dry powder for insufflation, alone or in combination with an inert carrier such as lactose or phospholipids; and nasal drops. For intranasal use, the powder may comprise a bioadhesive agent, including chitosan or cyclodextrin.

Solutions or suspensions for use in a pressurized container, pump, spray, atomizer, or nebulizer may be formulated to contain ethanol, aqueous ethanol, or a suitable alternative agent for dispersing, solubilizing, or extending release of the active ingredient provided herein, a propellant as solvent; and/or a surfactant, such as sorbitan trioleate, oleic acid, or an oligolactic acid.

The pharmaceutical compositions provided herein may be micronized to a size suitable for delivery by inhalation, such as about 50 micrometers or less, or about 10 micrometers or less. Particles of such sizes may be prepared using a comminuting method known to those skilled in the art, such as spiral jet milling, fluid bed jet milling, supercritical fluid processing to form nanoparticles, high pressure homogenization, or spray drying.

Capsules, blisters and cartridges for use in an inhaler or insufflator may be formulated to contain a powder mix of the pharmaceutical compositions provided herein; a suitable powder base, such as lactose or starch; and a performance modifier, such as l-leucine, mannitol, or magnesium stearate. The lactose may be anhydrous or in the form of the monohydrate. Other suitable excipients or carriers include dextran, glucose, maltose, sorbitol, xylitol, fructose, sucrose, and trehalose. The pharmaceutical compositions provided herein for inhaled/intranasal administration may further comprise a suitable flavor, such as menthol and levomenthol, or sweeteners, such as saccharin or saccharin sodium.

The pharmaceutical compositions provided herein for topical administration may be formulated to be immediate release or modified release, including delayed-, sustained-, pulsed-, controlled-, targeted, and programmed release.

D. Modified Release

The pharmaceutical compositions provided herein may be formulated as a modified release dosage form. As used herein, the term “modified release” refers to a dosage form in which the rate or place of release of the active ingredient(s) is different from that of an immediate dosage form when administered by the same route. Modified release dosage forms include delayed-, extended-, prolonged-, sustained-, pulsatile-, controlled-, accelerated- and fast-, targeted-, programmed-release, and gastric retention dosage forms. The pharmaceutical compositions in modified release dosage forms can be prepared using a variety of modified release devices and methods known to those skilled in the art, including, but not limited to, matrix controlled release devices, osmotic controlled release devices, multiparticulate controlled release devices, ion-exchange resins, enteric coatings, multilayered coatings, microspheres, liposomes, and combinations thereof. The release rate of the active ingredient(s) can also be modified by varying the particle sizes and polymorphorism of the active ingredient(s).

Examples of modified release include, but are not limited to, those described in U.S. Pat. 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.

1. Matrix Controlled Release Devices

The pharmaceutical compositions provided herein in a modified release dosage form may be fabricated using a matrix controlled release device known to those skilled in the art (see, Takada et al in “Encyclopedia of Controlled Drug Delivery,” Vol. 2, Mathiowitz Ed., Wiley, 1999).

In one embodiment, the pharmaceutical compositions provided herein in a modified release dosage form is formulated using an erodible matrix device, which is water-swellable, erodible, or soluble polymers, including synthetic polymers, and naturally occurring polymers and derivatives, such as polysaccharides and proteins.

Materials useful in forming an erodible matrix include, but are not limited to, chitin, chitosan, dextran, and pullulan; gum agar, gum arabic, gum karaya, locust bean gum, gum tragacanth, carrageenans, gum ghatti, guar gum, xanthan gum, and scleroglucan; starches, such as dextrin and maltodextrin; hydrophilic colloids, such as pectin; phosphatides, such as lecithin; alginates; propylene glycol alginate; gelatin; collagen; and cellulosics, such as ethyl cellulose (EC), methylethyl cellulose (MEC), carboxymethyl cellulose (CMC), CMEC, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), cellulose acetate (CA), cellulose propionate (CP), cellulose butyrate (CB), cellulose acetate butyrate (CAB), CAP, CAT, hydroxypropyl methyl cellulose (HPMC), HPMCP, HPMCAS, hydroxypropyl methyl cellulose acetate trimellitate (HPMCAT), and ethylhydroxy ethylcellulose (EHEC); polyvinyl pyrrolidone; polyvinyl alcohol; polyvinyl acetate; glycerol fatty acid esters; polyacrylamide; polyacrylic acid; copolymers of ethacrylic acid or methacrylic acid (EUDRAGIT®, Rohm America, Inc., Piscataway, N.J.); poly(2-hydroxyethyl-methacrylate); polylactides; copolymers of L-glutamic acid and ethyl-L-glutamate; degradable lactic acid-glycolic acid copolymers; poly-D-(−)-3-hydroxybutyric acid; and other acrylic acid derivatives, such as homopolymers and copolymers of butylmethacrylate, methylmethacrylate, ethylmethacrylate, ethylacrylate, (2-dimethylaminoethyl)methacrylate, and (trimethylaminoethyl)methacrylate chloride.

In further embodiments, the pharmaceutical compositions are formulated with a non-erodible matrix device. The active ingredient(s) is dissolved or dispersed in an inert matrix and is released primarily by diffusion through the inert matrix once administered. Materials suitable for use as a non-erodible matrix device included, but are not limited to, insoluble plastics, such as polyethylene, polypropylene, polyisoprene, polyisobutylene, polybutadiene, polymethylmethacrylate, polybutylmethacrylate, chlorinated polyethylene, polyvinylchloride, methyl acrylate-methyl methacrylate copolymers, ethylene-vinyl acetate copolymers, ethylene/propylene copolymers, ethylene/ethyl acrylate copolymers, vinyl chloride 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, polyvinyl chloride, plasticized nylon, plasticized polyethylene terephthalate, natural rubber, silicone rubbers, polydimethylsiloxanes, silicone carbonate copolymers, and; hydrophilic polymers, such as ethyl cellulose, cellulose acetate, crospovidone, and cross-linked partially hydrolyzed polyvinyl acetate,; and fatty compounds, such as carnauba wax, microcrystalline wax, and triglycerides.

In a matrix controlled release system, the desired release kinetics can be controlled, for example, via the polymer type employed, the polymer viscosity, the particle sizes of the polymer and/or the active ingredient(s), the ratio of the active ingredient(s) versus the polymer, and other excipients or carriers in the compositions.

The pharmaceutical compositions provided herein in a modified release dosage form may be prepared by methods known to those skilled in the art, including direct compression, dry or wet granulation followed by compression, melt-granulation followed by compression.

2. Osmotic Controlled Release Devices

The pharmaceutical compositions provided herein in a modified release dosage form may be fabricated using an osmotic controlled release device, including one-chamber system, two-chamber system, asymmetric membrane technology (AMT), and extruding core system (ECS). In general, such devices have at least two components: (a) the core which contains the active ingredient(s); and (b) a semipermeable membrane with at least one delivery port, which encapsulates the core. The semipermeable membrane controls the influx of water to the core from an aqueous environment of use so as to cause drug release by extrusion through the delivery port(s).

In addition to the active ingredient(s), the core of the osmotic device optionally includes an osmotic agent, which creates a driving force for transport of water from the environment of use into the core of the device. One class of osmotic agents water-swellable hydrophilic polymers, which are also referred to as “osmopolymers” and “hydrogels,” including, but not limited to, hydrophilic vinyl and acrylic polymers, polysaccharides such as calcium alginate, polyethylene oxide (PEO), polyethylene glycol (PEG), polypropylene glycol (PPG), poly(2-hydroxyethyl methacrylate), poly(acrylic) acid, poly(methacrylic) acid, polyvinylpyrrolidone (PVP), crosslinked PVP, polyvinyl alcohol (PVA), PVA/PVP copolymers, PVA/PVP copolymers with hydrophobic monomers such as methyl methacrylate and vinyl acetate, hydrophilic polyurethanes containing large PEO blocks, sodium croscarmellose, carrageenan, hydroxyethyl cellulose (HEC), hydroxypropyl cellulose (HPC), hydroxypropyl methyl cellulose (HPMC), carboxymethyl cellulose (CMC) and carboxyethyl, cellulose (CEC), sodium alginate, polycarbophil, gelatin, xanthan gum, and sodium starch glycolate.

The other class of osmotic agents is osmogens, which are capable of imbibing water to affect an osmotic pressure gradient across the barrier of the surrounding coating. Suitable osmogens include, but are not limited to, inorganic salts, such as magnesium sulfate, magnesium chloride, calcium chloride, sodium chloride, lithium chloride, potassium sulfate, potassium phosphates, sodium carbonate, sodium sulfite, lithium sulfate, potassium chloride, and sodium sulfate; sugars, such as dextrose, fructose, glucose, inositol, lactose, maltose, mannitol, raffinose, sorbitol, sucrose, trehalose, and xylitol,; organic acids, such as ascorbic acid, benzoic acid, fumaric acid, citric acid, maleic acid, sebacic acid, sorbic acid, adipic acid, edetic acid, glutamic acid, p-toluenesulfonic acid, succinic acid, and tartaric acid; urea; and mixtures thereof.

Osmotic agents of different dissolution rates may be employed to influence how rapidly the active ingredient(s) is initially delivered from the dosage form. For example, amorphous sugars, such as MANNOGEM™ EZ (SPI Pharma, Lewes, Del.) can be used to provide faster delivery during the first couple of hours to promptly produce the desired therapeutic effect, and gradually and continually release of the remaining amount to maintain the desired level of therapeutic or prophylactic effect over an extended period of time. In this case, the active ingredient(s) is released at such a rate to replace the amount of the active ingredient metabolized and excreted.

The core may also include a wide variety of other excipients and carriers as described herein to enhance the performance of the dosage form or to promote stability or processing.

Materials useful in forming the semipermeable membrane include various grades of acrylics, vinyls, ethers, polyamides, polyesters, and cellulosic derivatives that are water-permeable and water-insoluble at physiologically relevant pHs, or are susceptible to being rendered water-insoluble by chemical alteration, such as crosslinking. Examples of suitable polymers useful in forming the coating, include plasticized, unplasticized, and reinforced cellulose acetate (CA), cellulose diacetate, cellulose triacetate, CA propionate, cellulose nitrate, cellulose acetate butyrate (CAB), CA ethyl carbamate, CAP, CA methyl carbamate, CA succinate, cellulose acetate trimellitate (CAT), CA dimethylaminoacetate, CA ethyl carbonate, CA chloroacetate, CA ethyl oxalate, CA methyl sulfonate, CA butyl sulfonate, CA p-toluene sulfonate, agar acetate, amylose triacetate, beta glucan acetate, beta glucan triacetate, acetaldehyde dimethyl acetate, triacetate of locust bean gum, hydroxylated ethylene-vinylacetate, EC, PEG, PPG, PEG/PPG copolymers, PVP, HEC, HPC, CMC, CMEC, HPMC, HPMCP, HPMCAS, HPMCAT, poly(acrylic) acids and esters and poly(methacrylic) acids and esters and copolymers thereof, starch, dextran, dextrin, chitosan, collagen, gelatin, polyalkenes, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinyl esters and ethers, natural waxes, and synthetic waxes.

Semipermeable membrane may also be a hydrophobic microporous membrane, wherein the pores are substantially filled with a gas and are not wetted by the aqueous medium but are permeable to water vapor, as disclosed in U.S. Pat. No. 5,798,119. Such hydrophobic but water-vapor permeable membrane are typically composed of hydrophobic polymers such as polyalkenes, polyethylene, polypropylene, polytetrafluoroethylene, polyacrylic acid derivatives, polyethers, polysulfones, polyethersulfones, polystyrenes, polyvinyl halides, polyvinylidene fluoride, polyvinyl esters and ethers, natural waxes, and synthetic waxes.

The delivery port(s) on the semipermeable membrane may be formed post-coating by mechanical or laser drilling. Delivery port(s) may also be formed in situ by erosion of a plug of water-soluble material or by rupture of a thinner portion of the membrane over an indentation in the core. In addition, delivery ports may be formed during coating process, as in the case of asymmetric membrane coatings of the type disclosed in U.S. Pat. Nos. 5,612,059 and 5,698,220.

The total amount of the active ingredient(s) released and the release rate can substantially by modulated via the thickness and porosity of the semipermeable membrane, the composition of the core, and the number, size, and position of the delivery ports.

The pharmaceutical compositions in an osmotic controlled-release dosage form may further comprise additional conventional excipients or carriers as described herein to promote performance or processing of the formulation.

The osmotic controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art (see, Remington: The Science and Practice of Pharmacy, supra; Santus and Baker, J. Controlled Release 1995, 35, 1-21; Verma et al., Drug Development and Industrial Pharmacy 2000, 26, 695-708; Verma et al., J. Controlled Release 2002, 79, 7-27).

In certain embodiments, the pharmaceutical compositions provided herein are formulated as AMT controlled-release dosage form, which comprises an asymmetric osmotic membrane that coats a core comprising the active ingredient(s) and other pharmaceutically acceptable excipients or carriers. See, U.S. Pat. No. 5,612,059 and WO 2002/17918. The AMT controlled-release dosage forms can be prepared according to conventional methods and techniques known to those skilled in the art, including direct compression, dry granulation, wet granulation, and a dip-coating method.

In certain embodiments, the pharmaceutical compositions provided herein are formulated as ESC controlled-release dosage form, which comprises an osmotic membrane that coats a core comprising the active ingredient(s), a hydroxylethyl cellulose, and other pharmaceutically acceptable excipients or carriers.

3. Multiparticulate Controlled Release Devices

The pharmaceutical compositions provided herein in a modified release dosage form may be fabricated a multiparticulate controlled release device, which comprises a multiplicity of particles, granules, or pellets, ranging from about 10 μm to about 3 mm, about 50 μm to about 2.5 mm, or from about 100 μm to about 1 mm in diameter. Such multiparticulates may be made by the processes know to those skilled in the art, including wet-and dry-granulation, extrusion/spheronization, roller-compaction, melt-congealing, and by spray-coating seed cores. See, for example, Multiparticulate Oral Drug Delivery; Marcel Dekker: 1994; and Pharmaceutical Pelletization Technology; Marcel Dekker: 1989.

Other excipients or carriers as described herein may be blended with the pharmaceutical compositions to aid in processing and forming the multiparticulates. The resulting particles may themselves constitute the multiparticulate device or may be coated by various film-forming materials, such as enteric polymers, water-swellable, and water-soluble polymers. The multiparticulates can be further processed as a capsule or a tablet.

4. Targeted Delivery

The pharmaceutical compositions provided herein may also be formulated to be targeted to a particular tissue, receptor, or other area of the body of the subject to be treated, including liposome-, resealed erythrocyte-, and antibody-based delivery systems. Examples include, but are not limited to, U.S. Pat. Nos. 6,316,652; 6,274,552; 6,271,359; 6,253,872; 6,139,865; 6,131,570; 6,120,751; 6,071,495; 6,060,082; 6,048,736; 6,039,975; 6,004,534; 5,985,307; 5,972,366; 5,900,252; 5,840,674; 5,759,542; and 5,709,874.

Methods of Use

Provided herein are methods for treating or preventing a hepatitis C viral infection in a subject, which comprises administering to a subject a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In one embodiment, the subject is a mammal. In another embodiment, the subject is a human.

Additionally, provided herein is a method for inhibiting replication of a virus in a host, which comprises contacting the host with a therapeutically effective amount of the compound of Formula I, II, III, I′, II′, I″, II″, Ia, IIa, or IIIa, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In one embodiment, the host is a cell. In another embodiment, the host is a human cell. In yet another embodiment, the host is a mammal. In still another embodiment, the host is human.

In certain embodiments, administration of a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or more reduction in the replication of the virus relative to a subject without administration of the compound, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the administration by a method known in the art, e.g., determination of viral titer.

In certain embodiments, administration of a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, 75, 100-fold or more reduction in the replication of the virus relative to a subject without administration of the compound, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the administration by a method known in the art.

In certain embodiments, administration of a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or more reduction in the viral titer relative to a subject without administration of the compound, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the administration by a method known in the art.

In certain embodiments, administration of a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, 75, 100 or more fold reduction in the viral titer relative to a subject without administration of the compound, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the administration by a method known in the art.

Further provided herein is a method for inhibiting the replication of an HCV virus, which comprises contacting the virus with a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

In certain embodiments, the contacting of the virus with a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or more reduction in the virus titer relative to the virus without such contact, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the initial contact, by a method known in the art.

In certain embodiments, the contacting of the virus with a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, 75, 100-fold or more reduction in the virus titer relative to the virus without such contact, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the initial contact, by a method known in the art.

In certain embodiments, the contacting of the virus with a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, 99% or more reduction in the viral titer relative to the virus without such contact, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the initial contact by a method known in the art.

In certain embodiments, the contacting of the virus with a therapeutically effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, results in a 1, 2, 3, 4, 5, 10, 15, 20, 25, 50, 75, 100 or more fold reduction in the viral titer relative to the virus without such contact, as determined at 1 day, 2 days, 3 days, 4 days, 5 days, 10 days, 15 days, or 30 days after the initial contact, by a method known in the art.

Also provided herein is a method for treating, preventing, or ameliorating one or more symptoms of a liver disease or disorder associated with an HCV infection, comprising administering to a subject a therapeutically effective amount of the compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof. Non-limiting examples of diseases associated with HCV infection include chronic hepatitis, cirrhosis, hepatocarcinoma, or extra hepatic manifestation.

Provided herein is a method for inhibiting the activity of a polymerase, which comprises contacting the polymerase with an effective amount of a compound provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof, or a pharmaceutically acceptable salt, solvate, or prodrug thereof. In one embodiment, the polymerase is hepatitis C NS5B polymerase.

Depending on the condition, disorder, or disease, to be treated and the subject's condition, a compound provided herein may be administered by oral, parenteral (e.g., intramuscular, intraperitoneal, intravenous, ICV, intrasternal injection or infusion, subcutaneous injection, or implant), inhalation, nasal, vaginal, rectal, sublingual, or topical (e.g., transdermal or local) routes of administration, and may be formulated, alone or together, in suitable dosage unit with pharmaceutically acceptable carriers, adjuvants and vehicles appropriate for each route of administration.

The dose may be in the form of one, two, three, four, five, six, or more sub-doses that are administered at appropriate intervals per day. The dose or sub-doses can be administered in the form of dosage units containing from about 0.1 to about 1000 milligram, from about 0.1 to about 500 milligrams, or from 0.5 about to about 100 milligram active ingredient(s) per dosage unit, and if the condition of the patient requires, the dose can, by way of alternative, be administered as a continuous infusion.

In certain embodiments, an appropriate dosage level is about 0.01 to about 100 mg per kg patient body weight per day (mg/kg per day), about 0.01 to about 50 mg/kg per day, about 0.01 to about 25 mg/kg per day, or about 0.05 to about 10 mg/kg per day, which may be administered in single or multiple doses. A suitable dosage level may be about 0.01 to about 100 mg/kg per day, about 0.05 to about 50 mg/kg per day, or about 0.1 to about 10 mg/kg per day. Within this range the dosage may be about 0.01 to about 0.1, about 0.1 to about 1.0, about 1.0 to about 10, or about 10 to about 50 mg/kg per day.

Combination Therapy

The compounds provided herein may also be combined or used in combination with other therapeutic agents useful in the treatment and/or prevention of an HCV infection.

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.

As used herein, the term “synergistic” includes a combination of a compound provided herein and another therapy (e.g., a prophylactic or therapeutic agent) which has been or is currently being used to treat, prevent, or manage a disease or disorder, which is more effective than the additive effects of the therapies. A synergistic effect of a combination of therapies (e.g., a combination of prophylactic or therapeutic agents) permits the use of lower dosages of one or more of the therapies and/or less frequent administration of said therapies to a subject with a disorder. The ability to utilize lower dosages of a therapy (e.g., a prophylactic or therapeutic agent) and/or to administer said therapy less frequently reduces the toxicity associated with the administration of said therapy to a subject without reducing the efficacy of said therapy in the prevention or treatment of a disorder). In addition, a synergistic effect can result in improved efficacy of agents in the prevention or treatment of a disorder. Finally, a synergistic effect of a combination of therapies (e.g., a combination of prophylactic or therapeutic agents) may avoid or reduce adverse or unwanted side effects associated with the use of either therapy alone.

The compound provided herein can be administered in combination or alternation with another therapeutic agent, such as an anti-HCV agent. In combination therapy, effective dosages of two or more agents are administered together, whereas in alternation or sequential-step therapy, an effective dosage of each agent is administered serially or sequentially. The dosages given will depend on absorption, inactivation and excretion rates of the drug as well as other factors known to those of skill in the art. It is to be noted that dosage values will also vary with the severity of the condition to be alleviated. It is to be further understood that for any particular subject, specific dosage regimens and schedules should be adjusted over time according to the individual need and the professional judgment of the person administering or supervising the administration of the compositions.

It has been recognized that drug-resistant variants of HCV can emerge after prolonged treatment with an antiviral agent. Drug resistance most typically occurs due to the mutation of a gene that encodes for an enzyme used in viral replication. The efficacy of a drug against the viral infection can be prolonged, augmented, or restored by administering the compound in combination or alternation with a second, and perhaps third, antiviral compound that induces a different mutation from that caused by the principle drug. Alternatively, the pharmacokinetics, biodistribution or other parameters of the drug can be altered by such combination or alternation therapy. In general, combination therapy is typically preferred over alternation therapy because it induces multiple simultaneous stresses on the virus.

In certain embodiments, the compound provided herein is combined with one or more agents selected from the group consisting of an interferon, ribavirin, amantadine, an interleukin, a NS3 protease inhibitor, a cysteine protease inhibitor, a phenanthrenequinone, a thiazolidine, a benzanilide, a helicase inhibitor, a polymerase inhibitor, a nucleotide analogue, a nucleoside analogue, a gliotoxin, a cerulenin, an antisense phosphorothioate oligodeoxynucleotide, an inhibitor of IRES-dependent translation, and a ribozyme.

In certain embodiments, the compound provided herein is combined with a HCV protease inhibitor, including, but not limited to, Medivir HCV protease inhibitor (Medivir/Tobotec); ITMN-191 (InterMune), SCH 503034 (Schering), VX950 (Vertex); substrate-based NS3 protease inhibitors as disclosed in WO 98/22496; Attwood et al., Antiviral Chemistry and Chemotherapy 1999, 10, 259-273; DE 19914474; WO 98/17679; WO 99/07734; non-substrate-based NS3 protease inhibitors, such as 2,4,6-trihydroxy-3-nitro-benzamide derivatives (Sudo et al., Biochem. Biophys. Res. Commun. 1997, 238, 643-647), RD3-4082, RD3-4078, SCH 68631, and a phenanthrenequinone (Chu et al., Tetrahedron Letters 1996, 37, 7229-7232); SCH 351633 (Chu et al., Bioorganic and Medicinal Chemistry Letters 1999, 9, 1949-1952); Eglin c, a potent polymerase inhibitor (Qasim et al., Biochemistry 1997, 36, 1598-1607).

Other suitable protease inhibitors for the treatment of HCV include those disclosed in, for example, U.S. Pat. No. 6,004,933, which discloses a class of cysteine protease inhibitors of HCV endopeptidase 2.

Additional hepatitis C virus NS3 protease inhibitors include those disclosed in, for example, Llinàs-Brunet et al., Bioorg. Med. Chem. Lett. 1998, 8, 1713-1718; Steinkühler et al., Biochemistry 1998, 37, 8899-8905; U.S. Pat. Nos. 5,538,865; 5,990,276; 6,143,715; 6,265,380; 6,323,180; 6,329,379; 6,410,531; 6,420,380; 6,534,523; 6,642,204; 6,653,295; 6,727,366; 6,838,475; 6,846,802; 6,867,185; 6,869,964; 6,872,805; 6,878,722; 6,908,901; 6,911,428; 6,995,174; 7,012,066; 7,041,698; 7,091,184; 7,169,760; 7,176,208; 7,208,600; U.S. Pat. App. Pub. Nos.: 2002/0016294, 2002/0016442; 2002/0037998; 2002/0032175; 2004/0229777; 2005/0090450; 2005/0153877; 2005/176648; 2006/0046956; 2007/0021330; 2007/0021351; 2007/0049536; 2007/0054842; 2007/0060510; 2007/0060565; 2007/0072809; 2007/0078081; 2007/0078122; 2007/0093414; 2007/0093430; 2007/0099825; 2007/0099929; 2007/0105781; WO 98/17679; WO 98/22496; WO 99/07734; WO 00/059929; WO 00/09543; WO 02/060926; WO 02/08187; WO 02/008251; WO 02/008256; WO 02/08198; WO 02/48116; WO 02/48157; WO 02/48172; WO 03/053349; WO 03/064416; WO 03/064456; WO 03/099274; WO 03/099316; WO 2004/032827; WO 2004/043339; WO 2005/037214; WO 2005/037860; WO 2006/000085; WO 2006/119061; WO 2006/122188; WO 2007/001406; WO 2007/014925; WO 2007/014926; and WO 2007/056120.

Other protease inhibitors include thiazolidine derivatives, such as RD-1-6250, RD4 6205, and RD4 6193, which show relevant inhibition in a reverse-phase HPLC assay with an NS3/4A fusion protein and NS5A/5B substrate (Sudo et al., Antiviral Research 1996, 32, 9-18); thiazolidines and benzanilides identified in Kakiuchi et al., FEBS Lett. 1998, 421, 217-220; Takeshita et al., Analytical Biochemistry 1997, 247, 242-246.

Suitable helicase inhibitors include, but are not limited to, those disclosed in U.S. Pat. No. 5,633,358; and WO 97/36554.

Suitable nucleotide polymerase inhibitors include, but are not limited to, gliotoxin (Ferrari et al., Journal of Virology 1999, 73, 1649-1654), and the natural product cerulenin (Lohmann et al., Virology 1998, 249, 108-118).

Suitable interfering RNA (iRNA) based antivirals include, but are not limited to, short interfering RNA (siRNA) based antivirals, such as Sirna-034 and those described in WO/03/070750, WO 2005/012525, and U.S. Pat. Pub. No. 2004/0209831.

Suitable antisense phosphorothioate oligodeoxynucleotides (S-ODN) complementary to sequence stretches in the 5′ non-coding region (NCR) of HCV virus include, but are not limited to those described in Alt et al., Hepatology 1995, 22, 707-717, and nucleotides 326-348 comprising the 3′ end of the NCR and nucleotides 371-388 located in the core coding region of HCV RNA (Alt et al., Archives of Virology 1997, 142, 589-599; Galderisi et al., Journal of Cellular Physiology 1999, 181, 251-257);

Suitable inhibitors of IRES-dependent translation include, but are not limited to, those described in Japanese Pat. Pub. Nos.: JP 08268890 and JP 10101591.

Suitable ribozymes include those disclosed in, for example, U.S. Pat. Nos. 6,043,077; 5,869,253 and 5,610,054.

Suitable nucleoside analogs include, but are not limited to, the compounds described in U.S. Pat. Nos. 6,660,721; 6,777,395; 6,784,166; 6,846,810; 6,927,291; 7,094,770; 7,105,499; 7,125,855; and 7,202,224; U.S. Pat. Pub. Nos. 2004/0121980; 2005/0009737; 2005/0038240; and 2006/0040890; WO 99/43691; WO 01/32153; WO 01/60315; WO 01/79246; WO 01/90121, WO 01/92282, WO 02/18404; WO 02/32920, WO 02/48165, WO 02/057425; WO 02/057287; WO 2004/002422, WO 2004/002999, and WO 2004/003000.

Other miscellaneous compounds that can be used as second agents include, for example, 1-amino-alkylcyclohexanes (U.S. Pat. No. 6,034,134), alkyl lipids (U.S. Pat. No. 5,922,757), vitamin E and other antioxidants (U.S. Pat. No. 5,922,757), squalene, amantadine, bile acids (U.S. Pat. No. 5,846,964), N-(phosphonacetyl)-L-aspartic acid (U.S. Pat. No. 5,830,905), benzenedicarboxamides (U.S. Pat. No. 5,633,388), polyadenylic acid derivatives (U.S. Pat. No. 5,496,546), 2′,3′-dideoxyinosine (U.S. Pat. No. 5,026,687), benzimidazoles (U.S. Pat. No. 5,891,874), plant extracts (U.S. Pat. Nos. 5,725,859; 5,837,257; and 6,056,961), and piperidines (U.S. Pat. No. 5,830,905).

In certain embodiments, one or more compounds provided herein are administered in combination or alternation with an anti-hepatitis C virus interferon, including, but not limited to, INTRON® A (interferon alfa-2b) and PEGASYS® (Peginterferon alfa-2a); ROFERON® A (recombinant interferon alfa-2a), INFERGEN® (interferon alfacon-1), and PEG-INTRON® (pegylated interferon alfa-2b). In one embodiment, the anti-hepatitis C virus interferon is INFERGEN®, IL-29 (PEG-Interferon lambda), R7025 (Maxy-alpha), BELEROFON®, oral interferon alpha, BLX-883 (LOCTERON®), omega interferon, MULTIFERON®, medusa interferon, ALBUFERON®, or REBIF®.

In certain embodiments, one or more compounds provided herein are administered in combination or alternation with an anti-hepatitis C virus polymerase inhibitor, such as ribavirin, viramidine, NM 283 (valopicitabine), PSI-6130, R1626, HCV-796, or R7128.

In certain embodiments, the one or more compounds provided herein are administered in combination with ribavirin and an anti-hepatitis C virus interferon, such as INTRON® A (interferon alfa-2b), PEGASYS® (Peginterferon alfa-2a), ROFERON® A (recombinant interferon alfa-2a), INFERGEN® (interferon alfacon-1), and PEG-INTRON® (pegylated interferon alfa-2b),

In certain embodiments, one or more compounds provided herein are administered in combination or alternation with an anti-hepatitis C virus protease inhibitor, such as ITMN-191, SCH 503034, VX950 (telaprevir), or Medivir HCV protease inhibitor.

In certain embodiments, one or more compounds provided herein are administered in combination or alternation with an anti-hepatitis C virus vaccine, including, but not limited to, TG4040, PEVIPRO™, CGI-5005, HCV/MF59, GV1001, IC41, and INNO0101 (E1).

In certain embodiments, one or more compounds provided herein are administered in combination or alternation with an anti-hepatitis C virus monoclonal antibody, such as AB68 or XTL-6865 (formerly HepX-C); or an anti-hepatitis C virus polyclonal antibody, such as cicavir.

In certain embodiments, one or more compounds provided herein are administered in combination or alternation with an anti-hepatitis C virus immunomodulator, such as ZADAXIN® (thymalfasin), NOV-205, or oglufanide.

In certain embodiments, one or more compounds provided herein are administered in combination or alternation with NEXAVAR®, doxorubicin, PI-88, amantadine, JBK-122, VGX-410C, MX-3253 (celgosivir), SUVUS® (BIVN-401 or virostat), PF-03491390 (formerly IDN-6556), G126270, UT-231B, DEBIO-025, EMZ702, ACH-0137171, MitoQ, ANA975, AVI-4065, bavituximab (tarvacin), ALINIA® (nitrazoxanide) or PYN17.

In certain embodiments, the compounds provided herein can be combined with one or more steroidal drugs known in the art, including, but not limited to the group including, aldosterone, beclometasone, betamethasone, deoxycorticosterone acetate, fludrocortisone, hydrocortisone (cortisol), prednisolone, prednisone, methylprednisolone, dexamethasone, and triamcinolone.

In certain embodiments, the compounds provided herein can be combined with one or more antibacterial agents known in the art, including, but not limited to the group including amikacin, amoxicillin, ampicillin, arsphenamine, azithromycin, aztreonam, azlocillin, bacitracin, carbenicillin, cefaclor, cefadroxil, cefamandole, cefazolin, cephalexin, cefdinir, cefditorin, cefepime, cefixime, cefoperazone, cefotaxime, cefoxitin, cefpodoxime, cefprozil, ceftazidime, ceftibuten, ceftizoxime, ceftriaxone, cefuroxime, chloramphenicol, cilastin, ciprofloxacin, clarithromycim, clindamycin, cloxacillin, colistin, dalfopristin, demeclocycline, dicloxacillin, dirithromycin, doxycycline, erythromycin, enrofloxacin, ertepenem, ethambutol, flucloxacillin, fosfomycin, furazolidone, gatifloxacin, geldanamycin, gentamicin, herbimycin, imipenem, isoniazid, kanamycin, levofloxacin, linezolid, lomefloxacin, loracarbef, mafenide, moxifloxacin, meropenem, metronidazole, mezlocillin, minocycline, mupirocin, nafcillin, neomycin, netilmicin, nitrofurantoin, norfloxacin, ofloxacin, oxytetracycline, penicillin, piperacillin, platensimycin, polymyxin B, prontocil, pyrazinamide, quinupristine, rifampin, roxithromycin, spectinomycin, streptomycin, sulfacetamide, sulfamethizole, sulfamethoxazole, teicoplanin, telithromycin, tetracycline, ticarcillin, tobramycin, trimethoprim, troleandomycin, trovafloxacin, and vancomycin.

In certain embodiments, the compounds provided herein can be combined with one or more antifungal agents known in the art, including, but not limited to the group including amorolfine, amphotericin B, anidulafungin, bifonazole, butenafine, butoconazole, caspofungin, ciclopirox, clotrimazole, econazole, fenticonazole, filipin, fluconazole, isoconazole, itraconazole, ketoconazole, micafungin, miconazole, naftifine, natamycin, nystatin, oxyconazole, ravuconazole, posaconazole, rimocidin, sertaconazole, sulconazole, terbinafine, terconazole, tioconazole, and voriconazole.

In certain embodiments, the compounds provided herein can be combined with one or more anticoagulants known in the art, including, but not limited to the group including acenocoumarol, argatroban, bivalirudin, lepirudin, fondaparinux, heparin, phenindione, warfarin, and ximelagatran.

In certain embodiments, the compounds provided herein can be combined with one or more thrombolytics known in the art, including, but not limited to the group including anistreplase, reteplase, t-PA (alteplase activase), streptokinase, tenecteplase, and urokinase.

In certain embodiments, the compounds provided herein can be combined with one or more non-steroidal anti-inflammatory agents known in the art, including, but not limited to, aceclofenac, acemetacin, amoxiprin, aspirin, azapropazone, benorilate, bromfenac, carprofen, celecoxib, choline magnesium salicylate, diclofenac, diflunisal, etodolac, etoricoxib, faislamine, fenbufen, fenoprofen, flurbiprofen, ibuprofen, indometacin, ketoprofen, ketorolac, lornoxicam, loxoprofen, lumiracoxib, meclofenamic acid, mefenamic acid, meloxicam, metamizole, methyl salicylate, magnesium salicylate, nabumetone, naproxen, nimesulide, oxyphenbutazone, parecoxib, phenylbutazone, piroxicam, salicyl salicylate, sulindac, sulfinpyrazone, suprofen, tenoxicam, tiaprofenic acid, and tolmetin.

In certain embodiments, the compounds provided herein can be combined with one or more antiplatelet agents known in the art, including, but not limited to, abciximab, cilostazol, clopidogrel, dipyridamole, ticlopidine, and tirofibin.

The compounds provided herein can also be administered in combination with other classes of compounds, including, but not limited to, endothelin converting enzyme (ECE) inhibitors, such as phosphoramidon; thromboxane receptor antagonists, such as ifetroban; potassium channel openers; thrombin inhibitors, such as hirudin; growth factor inhibitors, such as modulators of PDGF activity; platelet activating factor (PAF) antagonists; anti-platelet agents, such as GPIIb/IIIa blockers (e.g., abciximab, eptifibatide, and tirofiban), P2Y(AC) antagonists (e.g., clopidogrel, ticlopidine and CS-747), and aspirin; anticoagulants, such as warfarin; low molecular weight heparins, such as enoxaparin; Factor VIIa Inhibitors and Factor Xa Inhibitors; renin inhibitors; neutral endopeptidase (NEP) inhibitors; vasopeptidase inhibitors (dual NEP-ACE inhibitors), such as omapatrilat and gemopatrilat; HMG CoA reductase inhibitors, such as pravastatin, lovastatin, atorvastatin, simvastatin, NK-104 (a.k.a. itavastatin, nisvastatin, or nisbastatin), and ZD-4522 (also known as rosuvastatin, atavastatin, or visastatin); squalene synthetase inhibitors; fibrates; bile acid sequestrants, such as questran; niacin; anti-atherosclerotic agents, such as ACAT inhibitors; MTP Inhibitors; calcium channel blockers, such as amlodipine besylate; potassium channel activators; alpha-adrenergic agents; beta-adrenergic agents, such as carvedilol and metoprolol; antiarrhythmic agents; diuretics, such as chlorothiazide, hydrochlorothiazide, flumethiazide, hydroflumethiazide, bendroflumethiazide, methylchlorothiazide, trichloromethiazide, polythiazide, benzothiazide, ethacrynic acid, ticrynafen, chlorthalidone, furosenide, muzolimine, bumetanide, triamterene, amiloride, and spironolactone; thrombolytic agents, such as tissue plasminogen activator (tPA), recombinant tPA, streptokinase, urokinase, prourokinase, and anisoylated plasminogen streptokinase activator complex (APSAC); anti-diabetic agents, such as biguanides (e.g., metformin), glucosidase inhibitors (e.g., acarbose), insulins, meglitinides (e.g., repaglinide), sulfonylureas (e.g., glimepiride, glyburide, and glipizide), thiozolidinediones (e.g., troglitazone, rosiglitazone, and pioglitazone), and PPAR-gamma agonists; mineralocorticoid receptor antagonists, such as spironolactone and eplerenone; growth hormone secretagogues; aP2 inhibitors; phosphodiesterase inhibitors, such as PDE III inhibitors (e.g., cilostazol) and PDE V inhibitors (e.g., sildenafil, tadalafil, and vardenafil); protein tyrosine kinase inhibitors; antiinflammatories; antiproliferatives, such as methotrexate, FK506 (tacrolimus), mycophenolate mofetil; chemotherapeutic agents; immunosuppressants; anticancer agents and cytotoxic agents (e.g., alkylating agents, such as nitrogen mustards, alkyl sulfonates, nitrosoureas, ethylenimines, and triazenes); antimetabolites, such as folate antagonists, purine analogues, and pyrimidine analogues; antibiotics, such as anthracyclines, bleomycins, mitomycin, dactinomycin, and plicamycin; enzymes, such as L-asparaginase; farnesyl-protein transferase inhibitors; hormonal agents, such as glucocorticoids (e.g., cortisone), estrogens/antiestrogens, androgens/antiandrogens, progestins, and luteinizing hormone-releasing hormone antagonists, and octreotide acetate; microtubule-disruptor agents, such as ecteinascidins; microtubule-stabilizing agents, such as pacitaxel, docetaxel, and epothilones A-F; plant-derived products, such as vinca alkaloids, epipodophyllotoxins, and taxanes; and topoisomerase inhibitors; prenyl-protein transferase inhibitors; and cyclosporins; steroids, such as prednisone and dexamethasone; cytotoxic drugs, such as azathioprine and cyclophosphamide; TNF-alpha inhibitors, such as tenidap; anti-TNF antibodies or soluble TNF receptor, such as etanercept, rapamycin, and leflunimide; and cyclooxygenase-2 (COX-2) inhibitors, such as celecoxib and rofecoxib; and miscellaneous agents such as, hydroxyurea, procarbazine, mitotane, hexamethylmelamine, gold compounds, platinum coordination complexes, such as cisplatin, satraplatin, and carboplatin.

In certain embodiments, the pharmaceutical compositions provided herein further comprise a second antiviral agent as described herein. In one embodiment, the second antiviral is selected from the group consisting of an interferon, ribavirin, an interleukin, an NS3 protease inhibitor, a cysteine protease inhibitor, a phenanthrenequinone, a thiazolidine, a benzanilide, a helicase inhibitor, a polymerase inhibitor, a nucleotide analogue, a nucleoside analogue, a gliotoxin, a cerulenin, an antisense phosphorothioate oligodeoxynucleotide, an inhibitor of IRES-dependent translation, and a ribozyme. In another embodiment, the second antiviral agent is an interferon. In yet another embodiment, the t interferon is selected from the group consisting of pegylated interferon alpha 2a, interferon alphcon-1, natural interferon, ALBUFERON®, interferon beta-1a, omega interferon, interferon alpha, interferon gamma, interferon tau, interferon delta, and interferon gamma-1b.

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.

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 provided herein, including a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug 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 an enantiomeric pair, an individual diastereomer, or a mixture of diastereomers thereof; or a pharmaceutically acceptable salt, solvate, or prodrug 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, needleless 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 seated 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.

The disclosure will be further understood by the following non-limiting examples.

EXAMPLES

As used herein, the symbols and conventions used in these processes, schemes and examples, regardless of whether a particular abbreviation is specifically defined, are consistent with those used in the contemporary scientific literature, for example, the Journal of the American Chemical Society or the Journal of Biological Chemistry. Specifically, but without limitation, the following abbreviations may be used in the examples and throughout the specification: g (grams); mg (milligrams); mL (milliliters); μL (microliters); mM (millimolar); μM (micromolar); Hz (Hertz); MHz (megahertz); mmol (millimoles); hr (hours); min (minutes); TLC (thin layer chromatography); HPLC (high performance liquid chromatography); SCX (strong cation exchange); MS (mass spectrometry); ESI (electrospray ionization); Rt (retention time); SiO2 (silica); THF (tetrahydrofuran); CD3OD (deuterated methanol); CDCl3 (deuterated chloroform); DCE (dichloroethane); DCM (dichloromethane); DMF (dimethyformamide); DMSO (dimethylsulfoxide); EtOAc (ethyl acetate); CHCl3 (chloroform); DMF (N,N-dimethylformamide); DMA (N,N-dimethylacetamide); MeOH (methanol); EtOH (ethanol); HCl (hydrochloric acid); LiOH (lithium hydroxide); NaOH (sodium hydroxide); KOH (potassium hydroxide); Cs2CO3 (cesium carbonate); DIPEA (N,N-diisopropylethylamine); TEA (trietlylamine); DBU (1,8-diazabicyclo[5.4.0]undec-7-ene; CDI (carbonyldiimidazole); TBTU (O-(benzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium tetrafluoroborate); Ac (acetyl); Me (methyl); Et (ethyl); tBu (tert-butyl); Boc (tert-butoxylcarbony); Bn (benzyl); and Ts (tosylate).

For all of the following examples, standard work-up and purification methods known to those skilled in the art can be utilized. Unless otherwise indicated, all temperatures are expressed in ° C. (degrees Centigrade). All reactions conducted at room temperature unless otherwise noted. Synthetic methodologies illustrated in Schemes 4 to 6 are intended to exemplify the applicable chemistry through the use of specific examples and are not indicative of the scope of the disclosure.

Intermediate 1 6-fluoro-1H-benzo[d][1,3]oxazine-2,4-dione

A solution of 5-fluoroanthranilic acid (10 g, 64.46 mmol; Aesar, Fluorochem, ABCR Product List, Eur. Pat. Appl. No. EP 647 614, Apr. 12, 1995, the contents of which are hereby incorporated by reference in their entirety) in anhydrous tetrahydrofuran (200 ml) was treated with triphosgene (12.5 g, 42.158 mmol) and stirred at 50° C. overnight. The yellow solution was cooled down to 0° C., saturated sodium hydrogen carbonate solution was added to pH 7-8, and the mixture was extracted with ethyl acetate (150 ml×2).

The combined organic layers were dried on anhydrous sodium sulfate, ethyl acetate was evaporated, and the residue was triturated with diethyl ether. The suspension was cooled down for 30 min, the precipitate was collected by filtration, washed with diethyl ether, and dried under vacuum to give intermediate 1 (11 g, 90%), which was a white powder. Intermediate 1 was characterized by the following spectroscopic data: 1NMR (d6-DMSO, 400 MHz) δ (ppm) 6.86 (dd, J=9.66 Hz and J=2.42 Hz, 1H), 7.09 (td, J=8.8 Hz and 2.42 Hz, 1H), 7.98 (dd, J=8.8 Hz and 6 Hz, 1H), 11.88 (brs, NH); and MS (ESI, EI+) m/z=182 (MH+).

Intermediate 2 1-(2-cyclopropylethyl)-6-fluorobenzo[d]-[1,3]oxazine-2,4-dione

Under nitrogen atmosphere, 6-fluoro-1H-benzo[d][1,3]oxazine-2,4-dione (2.2 g, 12.3 mmol), triphenylphosphine (3.5 g, 13.4 mmol) and cyclopropyl ethanol were stirred together in THF and treated with diethyl azodicarboxylate (40% in toluene, 6.14 ml, 13.4 mmol). The reaction was stirred overnight at room temperature and under nitrogen atmosphere. The THF was evaporated, and the crude compound was purified by silica gel chromatography (ethyl acetate/petroleum ether) to give intermediate 2 (2.54 g, 84%) which was a white powder. Intermediate 2 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.34-0.41 (m, 3H), 0.65-0.89 (m,2H), 1.54 (q, J=7.28 Hz, 2H), 4.08 (t, J=7.28 Hz, 2H), 7.17 (td, J=8.54 Hz and J=2.14 Hz, 1H), 7.47 (dd, J=11.3 Hz and J=2.14 Hz, 1H), 8.07 (dd, J=8.54 Hz and J=6.22 Hz, 1H); and MS (ESI, EI) m/z=222 (MH) mass of acid derivate.

Intermediate 3 1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carbonitrile

At room temperature and under nitrogen atmosphere, 1-(2-cyclopropylethyl)-6-fluorobenzo[d]-[1,3]oxazine-2,4-dione (2.5 g, 10.04 mmol), ethyl cyano acetate (1.07 ml, 10.04 mmol) in anhydrous DMF (30 ml) were treated with ethyldiisopropylamine (3.5 ml, 20.08 mmol). The reaction was stirred overnight at 150° C. The DMF was evaporated, and the residue was partitioned between 1M aqueous hydrochloric acid and ethyl acetate, the solvent was evaporated under reduced pressure, and the residue was triturated with 1M aqueous hydrochloric acid, isolated by filtration, washed with water until the filtrate was neutral, then dried at 45° C. under vacuum to give intermediate 3, which was a beige powder. Intermediate 3 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.0-0.029 (m, 2H), 0.34-0.40 (m, 2H), 0.71-0.81 (m, 1H), 1.47 (q, J=7.13 Hz, 2H), 4.23 (t, J=7.13 Hz, 2H), 7.16 (td, J=8.71 Hz and J=2.18 Hz, 1H), 7.46 (dd, J=11.88 Hz and J=2.18 Hz, 1H), 2.13 (dd, J=8.91 Hz and J=6.54 Hz, 1H); 19F NMR (d6-DMSO,376 MHz) δ (ppm) −103.72 (s, 1F); and MS (ESI, EI+) m/z=273 (MH+).

Intermediate 4 N-(2-bromophenyl)-1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carboxamidine

At room temperature and under nitrogen atmosphere, 1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carbonitrile (1.8 g, 6.6 mmol) and 2-bromo-aniline (3.4 g, 19.83 ml) in anhydrous 1,4-dioxane (30 ml) were stirred for 10 min, then the trimethyl aluminium in toluene (10 ml, 19.83 mmol) was slowly added. The reaction mixture was stirred at 85° C. overnight. The 1,4-dioxane was evaporated, the residue was slowly partitioned between 1M aqueous hydrochloric acid and ter-butyl-methyl ether, the organic layer was washed with saturated sodium hydrogen carbonate solution. The solvent was evaporated, and the residue was triturated in pentane, collected by filtration, washed with pentane then dried under vacuum to give intermediate 4. (2.73 g, 81%), which was a beige powder. Intermediate 4 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.04-0.07 (m, 2H), 0.4-0.41 (m, 2H), 0.74-0.85 (m, 1H), 1.5 (q, J=7.50 Hz, 2H), 4.22 (t, J=7.5 Hz, 2H), 7.03 (td, J=8.02 and J=1.29 Hz, 1H), 7.29-7.35 (m, 2H), 7.50 (td, J=7.50 Hz and J=1.29 Hz, 1H), 7.6 (d, J=7.50 Hz, 1H), 7.8 (d, J=8.02 Hz and J=1.29 H, 1H), 7.86 (br s, 1H), 8.19 (dd, J=7.24 Hz and J=8.80 Hz, 1H), 11.16 (br s, 1H), 13.92 (br s, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −106.44 (s, 1F); and MS (ESI, EI+) m/z=444 (MH+).

Intermediate 5 N-(2-diethylphosphonophenyl)-1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carboxamidine

To a solution of N-(2-bromophenyl)-1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carboxamidine (2.5 g, 5.6 mmol) in anhydrous acetonitrile, triethyl phosphite (2.94 ml, 16.9 mmol) and palladium acetate (126.3 mg, 0.56 mmol) were added under nitrogen atmosphere.

The reaction mixture was heated under microwaves radiations to 160° C. for 1 hour, then filtered through 0.45 μm filter and acetonitrile was evaporated. The residue was partitioned between ethyl acetate and 1M aqueous hydrochloric acid, the organic layer was washed with phosphate buffer solution pH 7. The ethyl acetate was evaporated under reduced pressure, and the residue purified by silica gel chromatography (petroleum ether/ethyl acetate) to give intermediate 5 (2 g, 70%), which was a beige solid. Intermediate 5 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=502 (MH+).

Intermediate 6 Diethyl(1-amino-4-nitrophenyl)phosphonate

2-iodo-4-nitro aniline (4 g, 7.57 mmol), triethyl phosphite (3.6 ml, 15.1 mmol) and palladium acetate (340 mg, 1.51 mmol) were mixed together in acetonitrile (36 ml), in a microwave tube. The vessel was sealed and placed in a microwave to react at 160° C., for 30 min. After cooling to room temperature, acetonitrile was evaporated. The residue obtained was diluted in ethyl acetate, washed with hydrochloric solution (1N) and phosphate buffer solution (pH 7). The solvent was evaporated and the crude material purified by silica gel chromatography (petroleum ether/ethyl acetate) to yield intermediate 6 (4 g, 92%), which was a brown solid. Intermediate 6 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 1.21 (t, J=7.1 Hz, 6H), 3.92 (m, 4H), 5.13 (s, NH2), 6.50 (t, J=7.8 Hz, 1H), 6.61 (m, 2H); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) 21.68 (s, 1P); and MS (ESI, EI+) m/z=275(MH+).

Intermediate 7 Diethyl(1,4-diaminophenyl)phosphonate

Intermediate 6 (1 g, 3.65 mmol) was dissolved in methanol (5 ml) Pd/C was added under nitrogen. After several vacuum/nitrogen cycles, hydrogen was introduced at atmospheric pressure. The reaction mixture was stirred at room temperature, under hydrogen, overnight. The reaction mixture was then filtered through celite and concentrated to dryness, to yield intermediate 7 (860 mg, 97%), which was a brown solid. Intermediate 7 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 1.20 (t, J=7.05 Hz, 6H), 3.87-4.00 (m, 4H), 4.45 (s, NH2), 5.15 (s, NH2), 6.50 (t, J=8 Hz, 1H), 6.58-6.60 (m, 2H); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) 21.46 (s, 1P); and MS (ESI, EI+) m/z=245 (MH+).

Intermediate 8 Diethyl(1-amino-4-methanesulfonaminephenyl)phosphonate

To a stirred solution of intermediate 7 (860 mg, 3.52 mmol), triethylamine (600 μl, 4.22 mmol) in dichloromethane (7 ml) at 0° C., was added methane sulfonyl chloride (327 μl, 4.22 mmol) under nitrogen. The reaction mixture was stirred at room temperature, over night. The mixture was then quenched with phosphate buffer solution (pH 7). The organic layer was separated, concentrated and the crude material was purified by silica gel chromatography (dichloromethane/methanol) to yield intermediate 8 (624 mg, 55%), which was a beige solid. Intermediate 8 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 1.34 (t, J=7.07 Hz, 6H), 2.94 (s, 3H), 4.04-4.22 (m, 4H), 5.14 (s, NH2), 6.57 (s, NH), 6.66 (t, J=7.26, 1H), 7.29 (d, J=8.79 Hz, 1H), 7.39 (d, J=14.9 Hz, 1H); 31P NMR (d6-DMSO, 162 MHz) δ ppm) 19.44 (s, 1P); and MS (ESI, EI+) m/z=323 (MH+).

Intermediate 9 Methyl 2-phenylpropanoate

Under nitrogen atmosphere, acetyl chloride was slowly added to 100 ml of methanol at 0° C. The solution was stirred for 10 min at 0° C., then warmed up to room temperature. The 2-phenylpropionic acid (146.07 mmol, 2 eq) was slowly added, the reaction mixture was stirred at room temperature for 15 min then heated to reflux for 3 hours. The solvent was removed under reduced pressure. The residue was dissolved in ethyl acetate, washed with water, buffer pH 7 then water. The organic layer was dried on sodium sulphate and evaporated to give intermediate 9 (11 g; 90%), which was a yellow oil. Intermediate 9 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 1.52 (d, J=7.24 Hz, 3H), 3.67 (s, 3H), 3.74 (q, J=7.24 Hz, 1H), 7.26-7.34 (m, 5H).

Intermediate 10 1-iodo-3,3-dimethylbutane

Under nitrogen atmosphere, sodium iodide (0.829 mmol, 10 eq) was added to a solution of 1-chloro-3,3 dimethyl butane (0.083 mmol, 1 eq) in anhydrous acetone (140 ml). The reaction mixture was stirred to reflux for 72 hours. The solvent was removed, and the residue was partitioned between diethyl ether and aqueous saturated thio sulfate solution. The organic layer was dried on sodium sulphate, and distillated on atmosphere pressure to yield intermediate 10. The product was collected at 140° C. to 160° C. (11 g, 64%), which was an orange oil. Intermediate 10 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.91 (s, 9H), 1.88 (t, J=12 Hz, 2H), 3.17 (t, J=12 Hz, 2H).

Intermediate 11 Methyl 2,5,5-trimethyl-2-phenylhexanoate

Under nitrogen atmosphere, lithium bis(trimethylsilyl) amide solution 1M in THF (13.4 mmol, 1.1 eq)was added to a solution of intermediate 9 (12.18 mmol, 1 eq) in THF (50 ml) at −40° C. The stirring was applied at −40° C. until complete formation of anion. At −40° C., and under nitrogen atmosphere, intermediate 10 was slowly added, then the reaction mixture was warmed to room temperature, and stirred until complete disappearance of starting material (3 hours). The reaction was quenched with 1M aqueous hydrochloric acid, and extracted with MTBE. The organic layer was washed with aqueous saturated thiosulfate solution, water then dried on sodium sulphate. The solvent was removed under reduced pressure to give intermediate 11 (2.5 g, 85%), which was a brown oil. Intermediate 11 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.81 (s, 9H), 0.98-1.03 (m, 2H), 1.47 (s, 3H), 1.80-1.87 (m, 1H), 3.60 (s, 3H), 7.16-7.21 (m, 1H), 7.24-7.27 (m, 4H).

Intermediate 12 2,5,5-trimethyl-2-phenylhexanoic acid

At room temperature, a mixture of intermediate 11 (10.2 mmol, 1 eq) and lithium hydroxide (51 mmol, 5 eq) in dioxane and water, were stirred until complete disappearance of starting material. T-butyl-methyl-ether was added, the aqueous layer was acidified with 1M aqueous hydrochloric acid until pH 1, and the product was extracted three times with MTBE. The combined organics layers were dried on sodium sulphate and evaporated to give intermediate 12 (2 g, 90%), which was a yellow oil. Intermediate 12 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=233 (MH).

Intermediate 13 Diethyl 2-(2,5,5-trimethyl-2-phenylhexanoyl)malonate

Under nitrogen atmosphere, thionyl chloride (27.31 mmol, 4 eq) was added dropwise to a solution of intermediate 12 and few drops of DMF in dichloromethane (15 ml) and few drops of DMF. The reaction mixture was stirred to room temperature for 2.5 hours, then the solvent was evaporated, and two co-evaporation with toluene were done to give the expected acid chloride, stocked under nitrogen atmosphere.

Under nitrogen atmosphere, magnesium chloride (6.83 mmol, 1 eq) and triethylamine (14.3 mmol, 2 eq) was added successively to a solution of diethylmalonate (6.83 mmol, 1 eq) in anhydrous acetonitrile (15 ml) at 0° C. The suspension was stirred at 0° C. for 15 min then warmed up to room temperature for 2H.

At 0° C., under nitrogen atmosphere, the acid chloride dissolved in anhydrous acetonitrile (15 ml) was added dropwise to the magnesium salt, then the reaction mixture was heated to 50° C. overnight. The solvent was removed and the residue was partitioned between ethyl acetate and 1M aqueous hydrochloric acid, washed with buffer pH 7. The solvent was removed and the residue was purified by chromatography on silica gel to yield intermediate 13 (74%), which was a yellowish oil. Intermediate 13 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.84 (s, 9H), 1.04-1.11 (m, 1H), 1.20-1.30 (m, 7H), 1.53 (s, 3H), 1.93-1.97 (m, 2H), 3.36 (s, 1H), 4.02-4.07 (m, 2H), 4.19-4.24 (m, 2H), 4.52 (s, 1H), 7.20-7.38 (m, 5H); and MS (ESI, EI+) m/z=375 (MH).

Intermediate 14 Ethyl 3,4-dihydro-1-hydroxy-4-methyl-4-(3,3-dimethylbutyl)-3-oxonaphtalene-2-carboxylate

Under nitrogene atmosphere, the intermediate 13 (5.31 mmol, 1 eq) in methanesulfonic acid was stirred at room temperature until complete disappearance of starting material (2 hours). The reaction mixture was quenched with water and extracted three times with ethyl acetate. The combined organics layers were dried on sodium sulphate, the solvent was removed under reduced pressure, and the residue was purified by chromatography on silica gel, to yield intermediate 14. (1.16 g, 66%), which was a yellow oil. Intermediate 14 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.49-0.55 (m, 1H), 0.75 (d, J=7.24 Hz, 9H), 0.84-0.92 (m, 2H), 1.22-1.51 (m, 6H), 1.76-1.97 (m, 1H), 2.25-2.32 (m, 1H), 4.44-4.52 (m, 2H), 7.39-7.44 (m, 2H), 7.56-7.62 (m, 1H), 8.21 (dd, J=7.24 Hz and J=33.63 Hz, 1H), 15.03 (s, 0.5H), 15.15 (s, 0.5H); and MS (ESI, EI+) m/z=331 (MH+).

Intermediate 15 4-hydroxy-1-methyl-1-(3,3-dimethylbutyl)naphtalen-2(2H,4H)-one

The intermediate 15 (3.027 mmol, 1 eq) in dioxane (15 ml) was treated with 2M aqueous hydrochloric acid (15 ml) at 100° C. for 5 hours. The reaction mixture was cooled down to room temperature, extracted three times with dichloromethane. The combined organics layers were dried on sodium sulphate and evaporated under reduced pressure. The residue was triturated with hexane (10 ml), the precipitate was collected by filtration, washed with hexane, and dried under vacuum to give intermediate 15 (522 mg, 67%), which was a yellow solid. Intermediate 15 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.40-0.47 (m, 0.5H), 0.72 (s, 5H), 0.81 (s, 4H), 0.83-0.91 (m, 1H), 1.03-1.12 (m, 0.5H), 1.52 (s, 1.5H), 1.57 (s, 1.5H), 1.77-1.93 (m, 1H), 1.98-2.06 (m, 0.5H), 2.22-2.30 (m, 0.5H), 3.74 (s, 1H), 5.32 (br s, 1H), 7.35-7.70 (m, 3H), 8.09-8.13 (m, 1H); and MS (ESI, EI+) m/z=259 (MH+).

Intermediate 16 Dimethyl trithiocarbonate methylsulfate salt

In a sealed tube, under nitrogen atmosphere, the dimethyl trithiocarbonate (0.2 mmol, 1 eq) and dimethyl sulphate (0.2 mmol, 1 eq) were stirred at 90° C. for 1 hour. The reaction mixture was cooled down to room temperature, triturated with diethyl ether. The precipitate was collected by filtration, washed with diethyl ether and dried under vacuum to give intermediate 16 (5.12 g, 97%), which was a white solid. Intermediate 16 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 2.13 (s, 3H), 3.17 (s, 3H), 3.37 (s, 3H).

Intermediate 17 2-(bis(methylthio)methylene)-4-methyl-4-(3,3-dimethylbutyl)naphthalene-1,3(4H)-dione

Under nitrogene atmosphere, the intermediate 15 (0.19 mmol, 1 eq), the intermediate 16 (0.387 mmol, 2 eq) and pyridine (0.387 mmol, 2 eq) in dioxane (8 ml) were heated to 120° C. for 2 hours. After cooling to room temperature, MTBE was added to the reaction mixture, and the solid was eliminated by filtration. The filtrate was evaporated under reduced pressure, and the residue was purified by chromatography on silica gel to give intermediate 17 (49.8 mg, 71%), which was a yellow solid. Intermediate 17 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.58-0.68 (m, 1H), 0.74 (s, 9H), 0.82-0.93 (m, 1H), 1.55 (s, 3H), 1.72-1.80 (m, 1H), 2.20-2.27 (m, 1H), 2.57 (s, 6H), 7.39-7.41 (m, 2H), 7.56 (t; J=7.76 Hz, 1H), 8.22 (d, J=7.76 Hz, 1H).

MS (ESI, EI+) m/z=363 (MH+).

Intermediate 18 (N-(2-diethylphosphonophenyl)-4-methanesulfonaminyl-carboxaminyl)-8-hydroxy-3-methyl-3-(3,3-dimethylbutyl)naphthalene-2(4H)-one

In a sealed tube and under nitrogen atmosphere, the intermediate 17 (0.138 mmol, 1 eq) and the intermediate 8 (0.138 mmol, 1 eq) in dioxane (11 ml) were stirred at 85° C. overnight. The reaction mixture was cooled down to 0° C. and saturated with ammonia gas, then heated to 100° C. for 2.5 hours. The dioxane was removed under reduced pressure to give intermediate 18, which was a white solid. Intermediate 18 was characterized by the following spectroscopic data: 1H NMR (Acetone-d6, 400 MHz) δ (ppm) in agreement with expected compound; 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −13.80 (s, 1P); and MS (ESI, EI+) m/z=606 (MH+).

Intermediate 19 6-fluoro-1H-benzo[d][1,3]oxazine-2,4-dione

A solution of 5-fluoroanthranilic acid (64.46 mmol) in anhydrous tetrahydrofuran (200 ml) was treated with triphosgene (42.16 mmol) and stirred at 50° C. overnight. The yellow solution was cooled down to 0° C., then a saturated sodium hydrogen carbonate solution was added to pH 7/8, and the mixture was extracted with ethyl acetate. The combined organic layers were dried on anhydrous sodium sulfate, the ethyl acetate was evaporated, and the residue was triturated with diethyl ether. The suspension was cooled down for 30 min, the precipitate was collected by filtration, washed with diethyl ether, and dried under vacuum to give intermediate 19, which was a white powder. Intermediate 19 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 6.86 (dd, J=9.66 Hz and J=2.42 Hz, 1H), 7.09 (td, J=8.8 Hz and 2.42 Hz, 1H), 7.98 (dd, J=8.8 Hz and 6 Hz, 1H), 11.88 (brs, NH); and MS (ESI, EI+) m/z=182 (MH+).

Intermediate 20 1-(2-cyclopropylethyl)-6-fluorobenzo[d]-[1,3]oxazine-2,4-dione

Under nitrogen atmosphere, intermediate 19 (12.3 mmol), triphenylphosphine (13.4 mmol) and cyclopropyl ethanol were stirred together in THF and treated with diethyl azodicarboxylate (40% in toluene, 13.4 mmol). The reaction was stirred overnight at room temperature and under nitrogen atmosphere. The THF was removed, and the crude compound was purified by silica gel chromatography to give intermediate 20, which was a white powder. Intermediate 20 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.34-0.41 (m, 3H), 0.65-0.89 (m,2H), 1.54 (q, J=7.28 Hz, 2H), 4.08 (t, J=7.28 Hz, 2H), 7.17 (td, J=8.54 Hz and J=2.14 Hz, 1H), 7.47 (dd, J=11.3 Hz and J=2.14 Hz, 1H), 8.07 (dd, J=8.54 Hz and J=6.22 Hz, 1H); and MS (ESI, EI) m/z=222 (MH) mass of acid derivate.

Intermediate 21 1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carbonitrile

At room temperature and under nitrogen atmosphere, intermediate 20 (10.04 mmol), ethyl cyano acetate (1.07 ml, 10.04 mmol) in anhydrous DMF (30 ml) were treated with ethyl diisopropylamine (20.08 mmol). The reaction was stirred overnight at 150° C. The DMF was removed, and the residue was partitioned between 1M aqueous hydrochloric acid and ethyl acetate, the solvent was evaporated under reduced pressure, and the residue was triturated with 1M aqueous hydrochloric acid, isolated by filtration, washed with water until the filtrate was neutral, then dried at 45° C. under vacuum to give intermediate 21, which was a beige powder. Intermediate 21 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.0-0.029 (m, 2H), 0.34-0.40 (m, 2H), 0.71-0.81 (m, 1H), 1.47 (q, J=7.13 Hz, 2H), 4.23 (t, J=7.13 Hz, 2H), 7.16 (td, J=8.71 Hz and J=2.18 Hz, 1H), 7.46 (dd, J=11.88 Hz and J=2.18 Hz, 1H), 2.13 (dd, J=8.91 Hz and J=6.54 Hz, 1H); 19F NMR (DMSO-d6, 400 MHz) δ (ppm) −103.72 (s, 1F); and MS (ESI, EI+) m/z=273 (MH+).

Intermediate 22 N-(2-bromophenyl)-1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carboxamidine

At room temperature and under nitrogen atmosphere, intermediate 21 (6.6 mmol) and 2-bromo-aniline (3.4 g, 19.83 ml) in anhydrous 1,4-dioxane (30 ml) were stirred for 10 min, then the trimethyl aluminium in toluene (19.83 mmol) was slowly added. The reaction mixture was stirred at 85° C. overnight. The 1,4-dioxane was removed, the residue was slowly partitioned between 1M aqueous hydrochloric acid and ter-butyl-methyl ether, the organic layer was washed with saturated sodium hydrogen carbonate solution. The solvent was removed, and the residue was triturated in pentane, collected by filtration, washed with pentane then dried under vacuum to give intermediate 22, which was a beige powder. Intermediate 22 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.04-0.07 (m, 2H), 0.4-0.41 (m, 2H), 0.74-0.85 (m, 1H), 1.5 (q, J=7.50 Hz, 2H), 4.22 (t, J=7.5 Hz, 2H), 7.03 (td, J=8.02 and J=1.29 Hz, 1H), 7.29-7.35 (m, 2H), 7.50 (td, J=7.50 Hz and J=1.29 Hz, 1H), 7.6 (d, J=7.50 Hz, 1H), 7.8 (d, J=8.02 Hz and J=1.29 H, 1H), 7.86 (br s, 1H), 8.19 (dd, J=7.24 Hz and J=8.80 Hz, 1H), 11.16 (br s, 1H), 13.92 (br s, 1H); 19F NMR (DMSO-d6, 376 MHz) δ (ppm) −106.44 (s, 1F); and MS (ESI, EI+) m/z=444 (MH+).

Intermediate 23 N-(2-diethylphosphonophenyl)-1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carboxamidine

To a solution of intermediate 22 (5.6 mmol) in anhydrous acetonitrile, triethyl phosphite (16.9 mmol) and acetate de palladium (0.56 mmol) were added under nitrogen atmosphere. The reaction mixture was heated under microwaves radiations to 160° C. for 1H, then filtered on 0.45 μm filter and the acetonitrile was removed. The residue was partitioned between ethyl acetate and 1M aqueous hydrochloric acid, the organic layer was washed with phosphate buffer solution pH 7. The ethyl acetate was evaporated under reduced pressure, and the residue was purified by silica gel chromatography to give intermediate 23, which was a beige solid. Intermediate 23 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=502 (MH+).

Intermediate 24 (2-amino-5-nitro-phenyl)-phosphonic acid diethyl ester

2-iodo-4-nitro aniline (7.57 mmol), triethyl phosphite (15.1 mmol) and palladium acetate (1.51 mmol) were mixed together in acetonitrile (36 ml), in a microwave tube. The vessel was sealed and placed in a microwave to react at 160° C., for 30 min. After cooling to room temperature, acetonitrile was removed. The residue obtained was diluted in ethyl acetate, washed with hydrochloric solution (1N) and phosphate buffer solution (pH 7). The solvent was removed and the crude material was purified by silica gel chromatography to yield intermediate 24, which was a brown solid. Intermediate 24 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.21 (t, J=7.1 Hz, 6H), 3.92 (m, 4H), 5.13 (s, NH2), 6.50 (t, J=7.8 Hz, 1H), 6.61 (m, 2H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 21.68 (s, 1P); and MS (ESI, EI+) m/z=275 (MH+).

Intermediate 25 (2,5-diaminophenyl)-phosphonic acid diethyl ester

Intermediate 24 (3.65 mmol) was dissolved in methanol (5 ml) Pd/C was added under nitrogen. After several cycles vacuum/nitrogen, hydrogen was introduced at atmospheric pressure. The reaction mixture was stirred at room temperature, under hydrogen, overnight. The reaction mixture was then filtered through celite and concentrated to yield intermediate 25, which was a brown solid. Intermediate 25 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.20 (t, J=7.05 Hz, 6H), 3.87-4.00 (m, 4H), 4.45 (s, NH2), 5.15 (s, NH2), 6.50 (t, J=8 Hz, 1H), 6.58-6.60 (m, 2H); 1P NMR (DMSO-d6, 162 MHz) δ (ppm) 21.46 (s, 1P); and MS (ESI, EI+) m/z=245 (MH+).

Intermediate 26 (2-amino-5-methanesulfonaminylphenyl)-phosphonic acid diethyl ester

To a stirred solution of the intermediate 25 (3.52 mmol), triethylamine (4.22 mmol) in dicloromethane (7 ml) at 0° C. was added methane sulfonyl chloride (4.22 mmol) under nitrogen. The reaction mixture was stirred at room temperature, over night. The mixture was then quenched with phosphate buffer solution (pH 7). The organic layer was separated, concentrated and the crude material was purified by silica gel chromatography (dichloromethane/methanol) to yield intermediate 26, which was a beige solid. Intermediate 26 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.34 (t, J=7.07 Hz, 6H), 2.94 (s, 3H), 4.04-4.22 (m, 4H), 5.14 (s, NH2), 6.57 (s, NH), 6.66 (t, J=7.26, 1H), 7.29 (d, J=8.79 Hz, 1H), 7.39 (d, J=14.9 Hz, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 19.44 (s, 1P); and MS (ESI, EI+) m/z=323 (MH+).

Intermediate 27 2-phenylpropionic acid methyl ester

Under nitrogen atmosphere, acetyl chloride was slowly added to 100 ml of methanol at 0° C. The solution was stirred for 10 min at 0° C., then warmed up to room temperature. The 2-phenylpropionic acid (146.07 mmol) was slowly added, the reaction mixture was stirred at room temperature for 15 min then heated to reflux for 3 hours. The solvent was removed under reduced pressure. The residue was dissolved in ethyl acetate, washed with water, buffer pH 7 then water. The organic layer was dried on sodium sulphate and evaporated to give intermediate 27, which was a yellow oil. Intermediate 27 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.52 (d, J=7.24 Hz, 3H), 3.67 (s, 3H), 3.74 (q, J=7.24 Hz, 1H), 7.26-7.34 (m, 5H).

Intermediate 28 1-iodo-3,3-dimethylbutane

Under nitrogen atmosphere, sodium iodide (0.829 mmol, 10 eq) was added to a solution of 1-chloro-3,3 dimethyl butane (0.083 mmol, 1 eq) in anhydrous acetone (140 ml). The reaction mixture was stirred to reflux for 72 hours. The solvent was removed, and the residue was partitioned between diethyl ether and aqueous saturated thiosulfate solution The organic layer was dried on sodium sulphate, and distillated at atmospheric pressure (140° C. to 160° C.) to yield intermediate 28, which was an orange oil. Intermediate 28 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.91 (s, 9H), 1.88 (t, J=12 Hz, 2H), 3.17 (t, J=12 Hz, 2H).

Intermediate 29 2,5,5-trimethyl-2-phenyl-hexanoic acid methyl ester

Under nitrogen atmosphere, lithium bis(trimethylsilyl) amide solution 1M in THF (13.4 mmol) was added to a solution of intermediate 27 (12.18 mmol) in THF (50 ml) at −40° C. The stirring was applied at −40° C. until complete formation of anion. At −40° C., and under nitrogen atmosphere, intermediate 10 was slowly added, then the reaction mixture was warmed to room temperature, and stirred until complete disappearance of starting material (3 hours). The reaction was quenched with 1M aqueous hydrochloric acid, and extracted with TBDME. The organic layer was washed with aqueous saturated thio sulfate solution, water then dried on sodium sulphate. The solvent was removed under reduced pressure to give intermediate 29, which was a brown oil. Intermediate 29 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.81 (s, 9H), 0.98-1.03 (m, 2H), 1.47 (s, 3H), 1.80-1.87 (m, 1H), 3.60 (s, 3H), 7.16-7.21 (m, 1H), 7.24-7.27 (m, 4H).

Intermediate 30 2,5,5-trimethyl-2-phenylhexanoic acid

At room temperature, a mixture of intermediate 29 (10.2 mmol) and lithium hydroxide (51 mmol) in dioxane and water, were stirred until complete disappearance of starting material. t-butyl-methyl-ether was added, the aqueous layer was acidified with 1M aqueous hydrochloric acid until pH 1 and the product was extracted thrice with TBDME. The combined organics layers were dried over sodium sulphate and evaporated to give intermediate 30, which was a yellow oil. Intermediate 30 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=233 (MH).

Intermediate 31 2-(2,5,5-trimethyl-2-phenyl-hexanoyl)malonic acid diethyl ester

Under nitrogen atmosphere, thionyl chloride (27.31 mmol) was added dropwise to a solution of intermediate 30 in dichloromethane (15 ml) and a few drops of DMF. The reaction mixture was stirred to room temperature for 2.5 hours. The solvent was then evaporated and two co-evaporations with toluene were done to give the expected acid chloride, stocked under nitrogen atmosphere. Under nitrogen atmosphere, magnesium chloride (6.83 mmol) and triethylamine (14.3 mmol) were added successively to a solution of diethylmalonate (6.83 mmol) in anhydrous acetonitrile (15 ml) at 0° C. The suspension was stirred at 0° C. for 15 min and warmed up to room temperature for 2 hours. At 0° C., under nitrogen atmosphere, the acid chloride dissolved in anhydrous acetonitrile (15 ml) was added dropwise to the magnesium salt. The reaction mixture was heated to 50° C. overnight. The solvent was removed and the residue was partitioned between ethyl acetate and 1M aqueous hydrochloric acid, washed with phosphate buffer solution (pH 7). The solvent was removed and the residue was purified by chromatography on silica gel to yield intermediate 31, which was a yellowish oil. Intermediate 31 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.84 (s, 9H), 1.04-1.11 (m, 1H), 1.20-1.30 (m, 7H), 1.53 (s, 3H), 1.93-1.97 (m, 2H), 3.36 (s, 1H), 4.02-4.07 (m, 2H), 4.19-4.24 (m, 2H), 4.52 (s, 1H), 7.20-7.38 (m, 5H); and MS (ESI, EI+) m/z=375 (MH).

Intermediate 32 4-(3,3-dimethylbutyl)-1-hydroxy-4-methyl-3-oxo-3,4-dihydronaphtalene-2-carboxylic acid ethyl ester

Under nitrogene atmosphere, the intermediate 31 (5.31 mmol) was stirred at room temperature in methanesulfonic acid until complete disappearance of starting material (2 hours). The reaction mixture was quenched with water and extracted thrice with ethyl acetate. The combined organics layers were dried on sodium sulphate. The solvent was removed under reduced pressure, and the residue was purified by chromatography on silica gel, to yield intermediate 32, which was a yellow oil. Intermediate 32 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.49-0.55 (m, 1H), 0.75 (d, J=7.24 Hz, 9H), 0.84-0.92 (m, 2H), 1.22-1.51 (m, 6H), 1.76-1.97 (m, 1H), 2.25-2.32 (m, 1H), 4.44-4.52 (m, 2H), 7.39-7.44 (m, 2H), 7.56-7.62 (m, 1H), 8.21 (dd, J=7.24 Hz and J=33.63 Hz, 1H), 15.03 (s, 0.5H), 15.15 (s, 0.5H); and MS (ESI, EI+) m/z=331 (MH+).

Intermediate 33 1-(3,3-dimethylbutyl)-4-hydroxy-1-methyl-1H-naphtalen-2-one

The intermediate 32 (3.027 mmol) in dioxane (15 ml) was treated with 2M aqueous hydrochloric acid (15 ml) at 100° C. for 5 hours. The reaction mixture was cooled down to room temperature, extracted three times with dichloromethane. The combined organics layers were dried on sodium sulphate and evaporated under reduced pressure. The residue was triturated with hexane (10 ml) the precipitate was collected by filtration, washed with hexane, and dried under vacuum to give intermediate 33, which was a yellow solid. Intermediate 33 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.40-0.47 (m, 0.5H), 0.72 (s, 5H), 0.81 (s, 4H), 0.83-0.91 (m, 1H), 1.03-1.12 (m, 0.5H), 1.52 (s, 1.5H), 1.57 (s, 1.5H), 1.77-1.93 (m, 1H), 1.98-2.06 (m, 0.5H), 2.22-2.30 (m, 0.5H), 3.74 (s, 1H), 5.32 (br s, 1H), 7.35-7.70 (m, 3H), 8.09-8.13 (m, 1H); and MS (ESI, EI+) m/z=259 (MH+).

Intermediate 34 Dimethyl trithiocarbonate methylsulfate salt

In a sealed tube, under nitrogen atmosphere, the dimethyl trithiocarbonate (0.2 mmol) and dimethyl sulphate (0.2 mmol) were stirred at 90° C. for 1 hour. The reaction mixture was cooled down to room temperature, triturated with diethyl ether. The precipitate was collected by filtration, washed with diethyl ether and dried under vacuum to give intermediate 34, which was a white solid. Intermediate 34 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 2.13 (s, 3H), 3.17 (s, 3H), 3.37 (s, 3H).

Intermediate 35 2-(bis(methylthio)methylene)-4-methyl-4-(3,3-dimethylbutyl)naphthalene-1,3(4H)-dione

Under nitrogene atmosphere, the intermediate 33 (0.19 mmol), the intermediate 34 (0.387 mmol) and pyridine (0.387 mmol) dissolved in dioxane (8 ml) were heated to 120° C. for 2 hours. After cooling down to room temperature, TBDME was added to the reaction mixture, and the solid was eliminated by filtration. The filtrate was evaporated under reduced pressure, and the residue was purified by chromatography on silica gel to give intermediate 35, which was a yellow solid. Intermediate 35 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.58-0.68 (m, 1H), 0.74 (s, 9H), 0.82-0.93 (m, 1H), 1.55 (s, 3H), 1.72-1.80 (m, 1H), 2.20-2.27 (m, 1H), 2.57 (s, 6H), 7.39-7.41 (m, 2H), 7.56 (t; J-7.76 Hz, 1H), 8.22 (d, J=7.76 Hz, 1H); and MS (ESI, EI+) m/z=363 (MH+).

Intermediate 36 (N-(2-diethylphosphonophenyl)-4-methanesulfonaminyl-carboxaminyl)-8-hydroxy-3-methyl-3-(3,3-dimethylbutyl)naphthalene-2(4H)-one

In a sealed tube and under nitrogen atmosphere, the intermediate 35 (0.138 mmol) and the intermediate 26 (0.138 mmol) in dioxane (11 ml) were stirred at 85° C. overnight. The reaction mixture was cooled down to 0° C. and saturated with ammonia gas, then heated to 100° C. for 2.5 hours. The dioxane was removed under reduced pressure to give intermediate 36, which was a white solid. Intermediate 36 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.37-0.44 (t, J=12.38 Hz, 1H), 0.68 (s, 9H), 1.02-1.06 (t, J=7.02 Hz, 1H), 1.10-1.20 (m,6H), 1.40 (s,3H), 1.72-1.82 (m,1H), 2.15-2.22 (m, 1H), 3.04 (s,3H), 3.95-4.1 (m, 4H), 7.35-7.38 (t, J=7.33, 1H), 7.50-7.57 (m, 4H), 7.73-7.77 (d, J=15.38 Hz, 1H), 8.05-8.10 (m, 1H), 10.17 (s, 1H), 11.17 (s, 1H), 13.14 (s, 1H), 13.41 (s, 1H); 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −13.80 (s, 1P); and MS (ESI, EI+) m/z=606 (MH+).

Intermediate 37 Methyl 2,5-dimethylphenylhexanoate

Intermediate 37 was synthesized from intermediate 27 and 1-iodo-3-methylbutene as described for intermediate 29, which was yellow oil. Intermediate 37 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 0.89 (m, 7H), 1.04-1.11 (m, 2H), 1.55 (s, 3H), 1.88-1.96 (m, 1H), 2.02-2.1 (m, 1H), 3.66 (s, 3H), 7.22-7.36 (m, 5H); and MS (ESI, EI+) m/z=235 (MH+).

Intermediate 38 2,5-dimethyl-2-phenylhexanoic acid

Intermediate 37 (12.3 mmol) and lithium hydroxide (61.5 mmol) were dissolved in dioxane (10 ml). The reaction mixture was refluxed for 4 hours. t-butyl-methyl-ether was added, the aqueous layer was acidified with 1M aqueous hydrochloric acid until pH 1 and the reaction mixture was extracted thrice with 10 ml of TBDME. The combined organic layers were dried over sodium sulphate and evaporated to give intermediate 38, which was an orange solid. Intermediate 38 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 0.88-0.90 (m, 6H), 1.08-1.12 (m, 2H), 1.50-1.52 (m, 1H), 1.57 (s, 3H), 1.91-1.99 (m, 1H), 2.02-2.10 (m, 1H), 7.24-7.28 (m,1H), 7.33-7.41 (m, 4H); and MS (ESI, EI) m/z=219 (MH).

Intermediate 39 Diethyl 2-(2,5-dimethyl-2-phenylhexanoyl)malonate

Intermediate 39 was synthesized from intermediate 38 and diethyl malonate as described for intermediate 31, which was a yellowish oil. Intermediate 39 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 0.81-0.87 (a 6H), 1.04-1.11 (m, 1H), 1.13-1.22 (m, 6H), 1.26-1.30 (m, 1H), 1.44-1.51 (m, 1H), 1.52 (s, 3H), 1.94-1.98 (m, 2H), 4.02-4.14 (m, 4H),4.18-4.24 (m, 1H), 4.51 (s, 1H), 7.26-7.35 (m, 5H); and MS (ESI, EI) m/z=361 (MH).

Intermediate 40 Ethyl 3,4-dihydro-1-hydroxy-4-methyl-4-(3-methylbutyl)-3-oxonaphtalene-2-carboxylate

Intermediate 40 was synthesized from intermediate 39 as described for intermediate 32, which was a yellow oil. Intermediate 40 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 0.47-0.59 (m, 1H), 0.70-0.76 (m, 6H), 0.82-0.87 (m, 7H), 1.04-1.12 (m, 1H), 1.15-1.20 (m, 6H), 1.31-1.39 (m, 1H), 1.44-1.49 (m, 3H), 1.53 (s, 3H), 1.91-1.98 (m, 2H), 2.20-2.36 (m, 1H), 4.04-4.12 (m, 4H), 4.43-4.50 (m, 3H), 7.23-7.27 (m, 3H), 7.32-7.35 (m, 1H), 7.39-7.45 (m, 2.36), 7.55-7.62 (m, 1H), 8.14-8.23 (m, 1H), 15.05 (s, 0.5H), 15.15 (s, 0.5H); and MS (ESI, EI+) m/z=317 (MH+).

Intermediate 41 4-hydroxy-1-methyl-1-(3-methylbutylnaphtalen-2(2H,4H)-one

Intermediate 41 was synthesized from intermediate 40 as described for intermediate 33, which was a white solid. Intermediate 41 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.34 (brs, 1H), 0.64-069 (dd,J2=8.36 Hz, J4=7.01 Hz, 7H), 1.24-1.31 (m, 1H), 1.41 (s, 3H),1.83-1.93 (m, 1H), 2.01-2.08 (m, 1H), 5.67 (s, 1H),7.34-7.39 (t, J=7.35 Hz, 1H,) 7.53-7.62 (m, 2H), 7.89 (s, 1H), 11.55 (s, 1H).

Intermediate 42 2-(bis(methylthio)methylene)-4-methyl-4-(3-methylbutyl)naphthalene-1,3(4H)-dione

Intermediate 42 was synthesized from intermediate 41 as described for intermediate 35, which was a yellow oil. Intermediate 42 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 0.62-0.70 (m, 1H), 0.72-0.76 (t, J=7.60 Hz, 6H), 0.82-0.91 (m, 1H), 1.35-1.40 (m, 1H), 1.55 (s, 3H), 1.74-1.82 (t, J=12.88 Hz, 1H), 2.20-2.28 (t, J=12.85 Hz, 1H), 2.55 (s, 6H), 7.37-7.42 (m, 2H), 7.55-7.59 (t, J=7.40 Hz, 1H), 8.22 (d, J=7.40 Hz, 1H); and MS (ESI, EI+) m/z=349 (MH+).

Intermediate 43 5-methanesulfonylamino-2-ureidophenyl)-phosphonic acid diethyl ester

To a stirred solution of intermediate 26 (1 eq) in chloroform (5 ml) and TEA (2. eq) was added N,N-carbodiimidazole (2 eq) at 0° C. The reaction mixture was stirred at 0° C. for 30 min and at room temperature overnight. The mixture was cooled down to 0° C. and 2.5 ml of 28% aq ammonia were added. The reaction mixture was stirred at room temperature overnight. The solvents were evaporated. The crude material was purified using preparative HPLC to yield intermediate 43, which was a beige solid. Intermediate 43 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 1.21-1.25 (t, J=7.10 Hz, 6H), 2.92 (s, 3H), 4.00-4.06 (m, 4H), 6.44 (s, 2H), 7.35-7.40 (m, 2H), 8.01-8.05 (t, J=8.04 Hz), 8.67 (s, 1H), 9.63 (brs, 1H); and MS (ESI, EI) m/z=364 (MH).

Intermediate 44 2,4-dimethyl-2-phenyl-pentanoic acid methyl ester

Intermediate 44 was synthesized from intermediate 27 and 1-iodo-2-methylpropane as described for intermediate 29, which was a yellow oil. Intermediate 44 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.75 (d, J=6.52 Hz, 3H), 0.79 (d, J=6.52 Hz, 3H), 1.48 (s, 3H), 1.55-1.59 (m, 1H), 1.78-1.96 (m, 2H), 3.56 (s, 3H), 7.21-7.31 (m, 5H).

Intermediate 45 2,4-dimethyl-2-phenyl-pentanoic acid

Intermediate 45 was synthesized from intermediate 44 as described for intermediate 30, which was a yellow oil. Intermediate 45 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.73 (d, J=6.20 Hz, 3H), 0.83 (d, J=6.20 Hz, 3H), 1.46 (s, 3H), 1.56-1.59 (m, 1H), 1.75-1.93 (m, 2H), 7.21-7.34 (m, 5H).

Intermediate 46 2-(2,4-dimethyl-2-phenyl-pentanoyl)malonic acid diethyl ester

Intermediate 46 was synthesized from intermediate 45 as described for intermediate 31, which was a yellow oil. Intermediate 46 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.51 (d, J=6.66 Hz, 3H), 0.84 (d, J=6.66 Hz, 3H), 0.98-1.02 (t, J=7.12 Hz, 3H), 1.09-1.13 (t, J=7.12 Hz, 3H), 1.41-1.44 (m, 1H), 1.53 (s, 3H), 1.80-1.83 (m, 2H), 3.86-3.91 (q, J=6.97 Hz, 2H), 4.01-4.10 (q, J=6.97 Hz, 2H), 4.70 (s, 1H), 7.25-7.35 (m, 5H).

Intermediate 47 1-hydroxy-4-isobutyl-4-methyl-3-oxo-3,4-dihydronaphtalene-2-carboxylic acid ethyl ester

Intermediate 47 was synthesized from intermediate 46 as described for intermediate 32, which was a yellow oil. Intermediate 47 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.44 (d, J=6.92 Hz, 3H),0.51 (d, J=6.92 Hz, 3H), 0.60 (d, J=6.92 Hz, 3H),0.84 (d, J=6.92 Hz, 3H),1.00-1.02 (t, J=7.10 Hz, 3H), 1.26-1.30 (t, J=7.08 Hz, 3H), 1.43 (s, 3H),1.53 (s, 3H), 1.80-1.83 (m, 1H), 1.97-2.12 (m, 2H), 3.46 (m, 1H), 3.87-3.89 (q, J=7.10 Hz, 2H), 4.02-4.10 (m, 2H), 4.28-4.31 (q, J=7.31 Hz, 2H), 4.70 (s, 1H), 7.25-7.66 (m, 8H), 13.81 (brs, 1H) (mixture of tautomeres).

Intermediate 48 4-hydroxy-1-isobutyl-1-methyl-1H-naphtalene-2-one

Intermediate 48 was synthesized from intermediate 47 as described for intermediate 33, which was a yellow solid. Intermediate 48 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.40 (d, J=6.12 Hz, 3H), 0.62 (d, J=6.12 Hz, 3H), 1.04-1.11 (m, 1H), 1.34 (s, 3H), 1.88-2.10 (m, 2H), 5.66 (s, 1H), 7.34-7.38 (t, J=7.59 Hz, 1H) 7.55 (t, J=7.59 Hz, 1H), 7.60 (d, J=7.59 Hz, 1H), 7.89 (d, J=7.59 Hz, 1H), 11.80 (brs, 1H).

Intermediate 49 2-(bis(methylsulfanyl)methylene)-4-isobutyl-4-methyl-naphthalene-1,3(4H)-dione

Intermediate 49 was synthesized from intermediate 48 and intermediate 34 as described for intermediate 35, which was a yellow solid. Intermediate 49 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.48-0.55 (m, 6H),1.22-1.34 (m, 1H), 1.43 (s, 3H), 1.80-1.92 (m, 2H), 2.53 (m, 6H), 7.39-7.43 (t, J=7.06 Hz, 1H), 7.57-7.69 (m, 2H), 8.03 (d, J=7.50 Hz, 1H).

Intermediate 50 (3-chloro-4-fluoro-phenyl)methanol

To a stirred solution of 3-chloro-4-fluoro-benzaldehyde (31.5 mmol) in MeOH (350 ml) was slowly added sodium borohydride (31.5 mmol). The reaction mixture was stirred at room temperature for 2 hours. Methanol was evaporated. The residue was solubilised in water and AcOEt. Organic phase was washed with brine, dried over Na2SO4, filtered and evaporated to yield Intermediate 50, which was a colourless oil. Intermediate 50 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.46 (d, J=5.45 Hz, 2H), 5.31-5.34 (t, J=5.45 Hz, 1H), 7.29-7.34 (m, 2H), 7.47 (d, J=6.78 Hz, 1H).

Intermediate 51 4-bromomethyl-2-chloro-1-fluoro-benzene

Intermediate 50 (1.25 mmol), triphenylphosphine (1.87 mmol) and carbon tetrabromide (1.87 mmol) were stirred in anhydrous DCM, under nitrogen for 3 hours. The solvent was removed under reduced pressure and the crude material was purified by chromatography on silica gel to give intermediate 51, which was a colourless oil. Intermediate 51 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 4.43 (s, 2H), 7.13 (t, J=8.37 Hz, 1H), 7.26-7.28 (m, 1H), 7.44-7.47 (d, J=6.70 Hz, 1H).

Intermediate 52 3-(3-chloro-4-fluoro-phenyl)-2-methyl-2-phenylpropionic acid methyl ester

Intermediate 52 was synthesized from intermediate 27 and intermediate 51 as described for intermediate 29, which was a colourless oil. Intermediate 52 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 1.49 (s, 3H), 3.12-3.35 (m, 2H), 3.70 (s, 3H), 6.70-6.72 (m, 1H), 6.88-6.93 (m, 2H), 7.24-7.30 (m, 5H); and 19F NMR (CDCl3, 376 MHz) δ (ppm) −119.06 (s, 1F).

Intermediate 53 3-(3-chloro-4-fluoro-phenyl)-2-methyl-2-phenylpropionic acid

Intermediate 53 was synthesized from intermediate 52 as described for intermediate 30, which was a pale yellow oil. Intermediate 53 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 1.33 (s, 3H), 3.13 (d, J=13.44 Hz, 1H), 3.28 (d, J=13.44 Hz, 1H), 7.19-7.35 (m, 8H), 12.60 (brs, 1H); and 19F NMR (DMSO-d6, 376 MHz) δ (ppm) −119.06 (s, 1F).

Intermediate 54 2-[3-(3-chloro-4-fluoro-phenyl)-2-methyl-2-phenylpropionyl]-malonic acid diethyl ester

Intermediate 54 was synthesized from intermediate 53 as described for intermediate 31, which was a colourless oil. Intermediate 54 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 1.15-1.19 (t, J=8 Hz, 3H), 1.24-1.27 (t, J=8 Hz, 3H), 1.57 (s, 3H), 3.15-3.24 (q, J=12 Hz, 2H), 4.04-4.10 (m, 2H), 4.19-4.24 (q, J=12 Hz, 2H), 4.50 (s, 1H), 6.50-6.52 (m, 1H), 6.54 (d, J=4.01 Hz, 1H), 6.69 (t, J=4.00 Hz, 1H), 7.13-7.16 (d, J=4.00 Hz, 2H), 7.27-7.37 (m, 3H); 19F NMR (CDCl3, 376 MHz) δ (ppm) −119.06 (s, 1F); and MS (ESI, EI) m/z=433 (MH).

Intermediate 55 4-(3-chloro-4-fluoro-benzyl)-2-methyl-2-phenylpropionyl]-malonic acid diethyl ester

Intermediate 55 was synthesized from intermediate 54 as described for intermediate 32, which was a colourless oil. Intermediate 55 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 1.27 (t, J=6.87 Hz, 3H), 1.60 (s, 3H), 3.21 (d, J=13.11 Hz, 1H), 3.30 (d, J=13.11 Hz, 1H), 4.26 (q, J=6.87 Hz, 2H), 6.50-6.53 (m, 1H), 6.70-6.72 (dd, J=1.94 Hz and J=8.16 Hz, 1H), 7.02 (t, J=8.93 Hz, 1H), 7.43 (t, J=7.38 Hz, 1H), 7.69 (d, J=7.38 Hz, 1H), 7.84 (d, J=8.16 Hz, 2H), 13.64 (brs, 1H); 19F NMR (DMSO-d6, 376 MHz) δ (ppm) −119.49 (s, 1F); and MS (ESI, EI) m/z=387 (MH).

Intermediate 56 1-(3-chloro-4-fluoro-benzyl)-4-hydroxy-1-methyl-1H-naphtalen-2-one

Intermediate 56 was synthesized from intermediate 55 as described for intermediate 33, which was a pale yellow solid. Intermediate 56 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 1.63 (s, 3H), 3.30 (m, 2H), 6.50-6.53 (m, 1H), 6.70-6.72 (dd, J=1.94 Hz and J=8.16 Hz, 1H), 7.02 (t, J=8.93 Hz, 1H), 7.43 (t, J=7.38 Hz, 1H), 7.69-7.91 (m, 3H), 11.76 (brs, 1H); 19F NMR (DMSO-d6, 376 MHz) δ (ppm) −119.69 (s, 1F); and MS (ESI, EI) m/z=315 (MH).

Intermediate 57 1-(3-chloro-4-fluoro-benzyl)-4-hydroxy-1-methyl-1H-naphtalen-2-one

Intermediate 57 was synthesized from intermediate 56 and intermediate 34 as described for intermediate 35, which was a yellow oil. Intermediate 57 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 1.73 (s, 3H), 2.40 (s, 6H), 3.02 (d, J=12.86 Hz, 1H), 3.35 (d, J=12.86 Hz, 1H), ), 6.31-6.35 (m, 1H), 6.63 (d, J=8.01 Hz, 1H), 6.69-6.73 (t, J=8.63 Hz, 1H), 7.40-7.43 (t, J=7.60 Hz, 1H), 7.49 (d, J=8.05 Hz, 1H), 7.60-7.64 (t, J=7.60 Hz, 1H), 8.11 (d, J=7.63 Hz, 1H); 19F NMR (CDCl3, 376 MHz) δ (ppm) −118.68 (s, 1F); and MS (ESI, EI+) m/z=421 (MH+).

Intermediate 58 1-(3,3-dimethyl-butyl)-4-methoxy-2-oxo-1,2-dihydro-pyridine-3-carbonitrile

To a stirred solution of 4-methoxy-2-oxo-1,2-dihydro-2-pyridine carbonitrile (20 mmol) in anhydrous acetonitrile (100 ml), was added potassium carbonate (30 mmol) under nitrogen. Intermediate 10 (26 mmol) diluted in acetonitrile (50 ml) was then added at room temperature. The reaction mixture was refluxed for 2 days. Acetonitrile was removed under reduced pressure. Water (100 ml) and ethyl acetate (200 ml) were added. The organic phase was extracted, dried over Na2SO4, filtered and evaporated. The crude material was purified by chromatography on silica gel to give intermediate 58, which was a white solid. Intermediate 58 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) (ppm) 0.99 (s, 9H), 1.57-1.61 (m, 2H), 3.91-3.95 (m, 2H), 3.99 (s, 3H), 6.09 (d, J=7.68 Hz, 1H), 7.53 (d, J=7.68 Hz, 1H); and MS (ESI, EI+) m/z=335(MH+).

Intermediate 59 1-(3,3-dimethyl-butyl)-4-hydroxy-2-oxo-1,2-dihydro-pyridine-3-carbonitrile

Intermediate 58 (4.5 mmol) was dissolved in dioxane (45 ml) and sodium hydroxide 1N (22.5 ml) was added. The reaction mixture was stirred at 60° C. for 2 hours. HCl (2N) was added until the formation of a white precipitate (pH=1). The precipitate was dissolved in AcOEt. The organic phase was separated, washed with brine, dried over Na2SO4, filtered and evaporated to yield intermediate 59, which was a white solid. Intermediate 59 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.91 (s, 9H), 1.42-1.46 (m, 2H), 3.80-3.84 (m, 2H), 6.01 (d, J=7.40 Hz, 1H), 7.85 (d, J=7.40 Hz, 1H), 12.59 (brs, 1H); and MS (ESI, EI+) m/z=221 (MH+).

Intermediate 60 1-(3,3-dimethyl-butyl)-4-hydroxy-5-iodo-2-oxo-1,2-dihydro-pyridine-3-carbonitrile

Intermediate 59 (3.41 mmol) was suspended in anyhydrous acetonitrile (35 ml) under nitrogen. N-iodosuccinimide (4.10 mmol) was added followed by trifluoroacetic acid (1.02 mmol) at room temperature. The reaction mixture was stirred at room temperature for 16 hours. Acetonitrile was removed under reduced pressure. Dichloromethane was added followed by saturated solution of sodium metabisulfite. The organic phase was separated, washed with brine, dried over Na2SO4, filtered and evaporated to yield intermediate 60, which was a pale yellow solid. Intermediate 60 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.91 (s, 9H), 1.42-1.46 (m, 2H), 3.80-3.82 (m, 2H), 8.29 (s, 1H), 12.59 (brs, 1H); and MS (ESI, EI+) m/z=347 (MH+).

Intermediate 61 1-(3,3-dimethyl-butyl-4-hydroxy -2-oxo-5-phenyl-1,2-dihydro-pyridine-3-carbonitrile

To a stirred solution of intermediate 60 (0.87 mmol) in anhydrous THF (1.8 ml) was added phenylboronic acid (2.6 mmol) and sodium carbonate solution (2M) (1.8 ml) under nitrogen. The reaction mixture was degazed and tetrakis (triphenylphosphine) palladium (0) (0.04 mmol) was added. The reaction mixture was stirred for 16 hours at 90° C. Water Was added to the mixture. The aqueous phase was washed with EtOAc, acidified to pH 1 with HCl 1N and then extracted with EtOAc. The organic phase was dried over Na2SO4, filtered and evaporated. The crude material was purified by chromatography on silica gel to give intermediate 61, which was a pale yellow solid. Intermediate 61 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.91 (s, 9H), 1.42-1.46 (m, 2H), 3.80-3.82 (m, 2H), 7.29-7.41 (m, 5H), 8.29 (s, 1H), 12.60 (brs, 1H); and MS (ESI, EI+) m/z=297 (MH+).

Intermediate 62 1-(3,3-dimethyl-butyl)-4-hydroxy-2-oxo-5-(3-thiophenyl)-1,2-dihydro-pyridine-3-carbonitrile

Intermediate 60 (0.29 mmol), 3-thiophene boronic acid (0.35 mmol), potassium carbonate (1.16 mmol), and tetrakis (triphenylphosphine) palladium (0) (0.015 mmol) dissolved in anh tetrahydrofuran were stirred at 90° C. under nitrogen for 16 hours. Water and EtOAc were added and the organic phase was separated, washed with brine dried over Na2SO4, filtered and evaporated. The crude material was purified by silica gel chromatography to give intermediate 62, which was a beige solid. Intermediate 62 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) (ppm) 0.92 (s, 9H), 1.44-1.48 (m, 2H), 3.75-3.79 (m, 2H), 7.39 (d, J=5.05 Hz, 1H), 7.46-7.48 (m, 1H), 7.83-7.85 (m, 2H).

Intermediate 63 1-(3,3-dimethyl-butyl)-4-hydroxy-2-oxo-5-pyrazol-1-yl-1,2-dihydro-pyridine-3-carbonitrile

Intermediate 60 (0.87 mmol), pyrazole (1.21 mmol), copper iodide (0.17 mmol) and cesium carbonate (2.61 mmol) dissolved in anh DMF (2 ml) were stirred in a sealed tube at 120° C. for 2 days. The reaction mixture was cooled down to room temperature, ethyl acetate and methanol were added followed by Dowex® until pH 7. The mixture was filtered though celite, washed with brine, dried over Na2SO4, filtered and evaporated to yield intermediate 63, which was a brown solid. Intermediate 63 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=287 (MH+).

Intermediate 64 1-(3,3-dimethyl-butyl)-5-iodo-2-oxo-4-methoxy-1,2-dihydro-pyridine-3-carbonitrile

Intermediate 64 was synthesized from intermediate 58 as described for intermediate 60. Yellow solid. 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.91 (s, 9H), 1.43-1.47 (m, 2H), 3.83-3.87 (m, 2H), 4.25 (s, 3H), 8.44 (s, 1H). Intermediate 64 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=361 (MH+).

Intermediate 65 1-(3,3-dimethyl-butyl-4-hydroxy-2-oxo-5-(2-thiophenyl)-1,2-dihydro-pyridine-3-carbonitrile

Intermediate 64 (0.56 mmol), 2-thiophene boronic acid (1.11 mmol), potassium carbonate (2.24 mmol), and tetrakis (triphenylphosphine) palladium (0) (0.03 mmol) dissolved in anh tetrahydrofuran (1.11 ml) were stirred at 90° C. under nitrogen for 16 hours. NaOH (1N) aq solution was then added and the mixture was stirred at 90° C. for 3 hours. The reaction mixture was cooled down to room temperature, water and AcOEt were added. Organic phase was washed with water, dried over Na2SO4, filtered and concentrated under reduced pressure. The crude material was dissolved in AcOEt, filtered over celite, washed with AcOEt/MeOH 15% and concentrated under reduced pressure to yield intermediate 65, which was a yellow oil. Intermediate 65 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=303 (MH+).

Intermediate 66 2-[(furan-3-ylmethyl)-amino]-3,3-dimethyl-butyric acid methyl ester

Intermediate 66 was synthesized from methyl-2-amino-3,3-dimethyl butanoate (4.82 mmol, Bionet) and 3-furaldehyde (4.82 mmol, Aldrich) as described for intermediate 88, which was a yellow oil. Intermediate 66 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.87 (s, 9H), 2.13 (brs, 1H), 2.82 (s, 1H), 3.32 (d, J=13.69 Hz, 1H), 3.55 (d, J=13.69 Hz, 1H), 3.60 (s, 3H), 6.39 (s, 1H), 7.47 (s, 1H), 7.55 (s, 1H).

Intermediate 67 4-hydroxy-N-methyl-butyramide

The γ-butyrolactone (0.013 mol, Aldrich) and methylamine 2M in methanol (0.066 mol) were stirred at 60° C. in a sealed MPS tube over 5 days. Then, the reaction mixture was evaporated to give intermediate 67, which was a colorless oil. Intermediate 67 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.6 (quintuplet, J=7.01 Hz, 2H), 2.07 (t, J=7.54 Hz, 2H), 2.53 (d, J=4.65 Hz, 3H), 3.34 (q, J=6.32 Hz, 2H), 4.44 (t, J=5.16 Hz, 1H), 7.67 (brs, 1H).

Intermediate 68 4-hydroxy-butyramide

The γ-butyrolactone (0.013 mol, Aldrich) was put in methanol and the reaction mixture was saturated with ammonia gas. This mixture was stirred at 60° C. in a sealed MPS tube over 2 days. Then, the reaction mixture was evaporated and washed with hot ethyl acetate. This hot solution was filtered and cooled down to 0° C. The white solid was filtered and dried under vacuo to give intermediate 68, which was a white solid. Intermediate 68 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.60 (quintuplet, J=6.91 Hz, 2H), 2.06 (t, J=7.47 Hz, 2H), 3.35 (t, J=6.30 Hz, 2H), 4.43 (t, J=5.14 Hz, 1H), 6.68 (brs, 1H), 7.22 (brs, 1H).

Intermediate 69 (2-([1-hydroxy-4-methyl-4-(3-methylbutyl)-3-oxo-3,4-dihydro-naphtalene-2-carboximidoyl]-amino)-5-methanesulfonamino-phenyl)-phosphonic acid diethyl ester

Intermediate 69 was obtained as a by-product from example 20 synthesis, which was a white solid. Intermediate 69 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.55-0.62 (m, 1H), 0.73-0.78 (m, 6H), 0.85-0.96 (m, 1H), 1.29-1.36 (m, 6H), 1.37 (m,1H), 1.59 (s, 3H), 1.81-1.87 (t, J=12.35 Hz, 1H), 2.28-2.36 (m, 1H), 3.06 (s, 3H), 4.15-4.22 (q, J=7.24 Hz, 4H), 5.36-5.40 (d, J=15.87 Hz, 1H), 7.38-7.45 (m, 3H), 7.54-7.57 (t, J=7.50 Hz, 1H), 7.86 (d, J=8.40 Hz, 1H), 8.16 (d, J=15.87 Hz, 1H), 8.25-8.28 (t, J=7.47 Hz, 1H), 9.90 (brs, 1H), 11.71 (d, J=57.8 Hz, 1H), 13.87 (d, J=49.02 Hz, 1H); 31P NMR (CDCl3, 162 MHz) δ (ppm) −7.02 (d, J=4.68 Hz, 1P); and MS (ESI, EI+) m/z=592 (MH+).

Example 1 1-(2-cyclopropylethyl)-(1-ethoxy-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

Intermediate 5 (90 mg, 0.18 mmol) was dissolved in dimethylacetamide (1 ml), heated under microwaves radiations to 200° C. for 4.5 hours. After cooling, the reaction mixture was added dropwise to 25 ml buffer pH 7 (0.1M). The product was extracted in ethyl acetate, after evaporation of the organic layer, the residue was purified by silica gel chromatography (petroleum ether/ethyl acetate) to give Example 1 (24.3 mg, 40%), which was a white powder. Example 1 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.07-0.10 (m, 2H), 0.39-0.44 (m, 2H), 0.76-0.87 (m, 1H), 1.21 (t, J=6.99 Hz, 3H), 1.56 (q, J=7.36 Hz, 2H), 3.99-4.05 (m, 2H), 4.33-4.36 (m, 2H), 7.40-8.08 (m, 7H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −120.47 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) −0.04 (s, 1P); and MS (ESI, EI+) m/z=456 (MH+).

Example 2 1-(2-cyclopropylethyl)-(1-ethoxy-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline sodium salt

Example 1 (10 mg, 0.0018 mmol), NaOH 0.1M (187 μl, 1 eq) were stirred in dioxane (2 ml) and water (2 ml) at 40° C. for 10 min and freeze dried to give Example 2, which was a white powder. Example 2 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.06-0.15 (m, 2H), 0.40-0.46 (m, 2H), 0.73-0.82 (m, 1H), 1.08 (t, J=7.01 Hz, 3H), 1.43-1.49 (m, 2H), 3.45-3.56 (m, 1H), 3.63-3.73 (m, 1H), 4.01-4.22 (m, 2H), 7.10 (t, J=7.4 Hz, 1H), 7.16 (t, J=7.4 Hz, 1H), 7.27-7.35 (m, 2H), 7.46-7.55 (m, 2H), 7.76 (dd, J=9.54 Hz and J=2.72 Hz, 1H), 14.9 (s, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −124.59 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) 3.24 (s, 1P); and MS (ESI, EI+) m/z=456 (MH+).

Example 3 1-(2-cyclopropylethyl)-(1-hydroxy-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

The chromatography column of example 2 was flushed with methanol, the methanol was concentrated and the residue was precipitated in a minimum of methanol, filtered, washed with cold methanol and dried under reduced pressure, to give Example 3 (185 mg), which was a white powder. Example 3 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.06-0.09 (m, 2H), 0.38-0.43 (m, 2H), 0.75-0.82 (m, 1H), 1.54 (q, J=7.22 Hz, 2H), 4.28 (t, J=7.22 Hz, 2H), 7.39-7.47 (m, 2H), 7.54-7.64 (m, 2H), 7.68 (t, J=7.72 Hz, 1H), 7.74-7.84 (m, 2H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −120.77 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) −5.51 (s, 1P); and MS (ESI, EI+) m/z=428 (MH+).

Example 4 1-(2-cyclopropylethyl)-(1-hydroxy-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline sodium salt

Example 4 was obtained from example 3 as described for example 2 and was a white powder. Example 4 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.06-0.09 (m, 2H), 0.38-0.42 (m, 2H), 0.75-0.85 (m, 1H), 1.54 (q, J=7.24 Hz, 2H), 4.29 (t, J=7.24 Hz, 2H), 7.15-7.22 (m, 2H), 7.41 (t, J=7.50 Hz, 1H), 7.51-7.56 (m, 3H), 7.84 (dd, J=9.31 Hz and J=2.72 Hz, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −122.02 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) −13.61 (s, 1P); and MS (ESI, EI+) m/z=428 (MH+).

Example 5 1-(2-cyclopropylethyl)-(1-methoxy-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

At room temperature and under nitrogen atmosphere, 1-(2-cyclopropylethyl)-(1-ethoxy-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline (52 mg, 0.114 mmol) was dissolved in dichloromethane (2 ml), then trimethylsilyl bromide was slowly added and stirred for 1 hour at room temperature. The dichloromethane was evaporated under reduced pressure. To the residue, trimethyl phosphite was added under nitrogen atmosphere. The reaction mixture was sealed and heated under pressure to 90° C. overnight. After cooling the reaction mixture was added to an 1M aqueous hydrochloric acid solution, the suspension was stirred at room temperature for 2 hours, collected by filtration, washed to pH 6-7 with water, and dried under vacuum to give Example 5 (30 mg, 66%), which was a white powder. Example 5 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.06-0.10 (m, 2H), 0.39-0.43 (m, 2H), 0.78-0.84 (m, 1H), 1.56 (q, J=7.24 Hz, 2H), 3.63 (d, J=12.68 Hz, 3H), 4.27-4.38 (m, 2H), 7.50 (td, J=7.5 Hz and J=2.07 Hz, 1H), 7.58-7.72 (m, 3H), 7.79-7.92 (m, 3H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −120.22 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) −1.84 (s, 1P); and MS (ESI, EI+) m/z=442 (MH+).

Example 6 1-(2-cyclopropylethyl)-(1-methoxy-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline sodium salt

Example 6 was obtained from example 5 as described for example 2 and was a white powder. Example 6 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.08-0.12 (m, 2H), 0.39-0.44 (m, 2H), 0.73-0.83 (m, 1H), 1.46 (q, J=7.24 Hz, 2H), 3.26 (d, J=12.42 Hz, 3H), 4.07-4.21 (m, 2H), 7.10-7.19 (m, 2H), 7.28-7.35 (m, 2H), 7.48-7.57 (m, 2H), 7.76 (dd, J=9.57 Hz and J=2.59 Hz, 1H), 14.91 (s, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −124.08 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) 5.50 (s, 1P); and MS (ESI, EI+) m/z=442 (MH+).

Example 7 1-(2-cyclopropylethyl)-(1-amino-1-oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

To a suspension of example 3 (85 mg, 0.199 mmol) in dichloromethane (7 ml), thionyl chloride (43.5 μl, 0.6 mmol) was added under nitrogen. The reaction mixture was stirred at room temperature for one hour. Dichloromethane was evaporated, the residue obtained was dissolved in new dichloromethane (7 ml). The reaction mixture was cooled down to 0° C. and saturated with ammonia gas at atmospheric pressure. The mixture was stirred for three hours. The precipitate was filtered, washed with dichloromethane. The precipitate was let to stir for one hour with water, removed by filtration and washed with water. The precipitate was then stirred for one hour with dichloromethane, removed by filtration, washed with dichloromethane and dried to yield Example 7 (40.7 mg, 48%), which was a white powder. Example 7 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.07 (q, J=4.6 Hz, 2H), 0.40 (q, J=8.01 Hz, 2H), 0.82 (m, 1H), 1.55 (q, J=7.35 Hz, 2H), 4.29 (t, J=7.35 Hz, 2H), 5.38 (d, J=4.73 Hz, 2H), 7.15 (t, J=8.40 Hz, 1H), 7.43-7.50 (m, 3H), 7.69 (t, J=7.35 Hz, 1H), 7.80 (dd, J=7.88 Hz and J=6.83 Hz, 1H), 8.23 (t, J=7.88 Hz, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −104.67 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) −1.39 (s, 1P); and MS (ESI, EI+) m/z=427 (MH+).

Example 8 1-(2-cyclopropylethyl)-(1-fluoro-1oxo-4H-benzo[1,2,4]phosphadiazine 3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

To a suspension of example 3 (85 mg, 0.199 mmol) in dichloromethane (7 ml) at −75° C., DAST (53 μl, 0.4 mmol) was slowly added. The reaction mixture was stirred at −70° C. for one hour and warmed up to 0° C. HCl 0.5N (5 ml) and water (5 ml) were added. Organic layer was separated and concentrated under reduced pressure. The crude material was purified by silica gel chromatography (petroleum ether/ethyl acetate) to give Example 8 (29 mg, 34%), which was a white solid. Example 8 was characterized by the following spectroscopic data: 1H NMR (d6-CDCl3, 400 MHz) δ (ppm) 0.16 (d, J=5.44 Hz, 2H), 0.54 (d, J=7.17 Hz, 2H),0.82 (m, 1H), 1.69 (q, J=7.48 Hz, 2H), 4.35 (t, J=8.16 Hz, 2H), 7.05-7.35 (m, 2H), 7.43 (t, J=8.16 Hz, 1H), 7.47 (t, J=7.48 Hz, 1H), 7.74 (t, J=8.16 Hz, 1H), 7.80 (dd, J=7.48 Hz, 1H), 8.33 (t, J=8.16 Hz, 1H), 14.4 (s, 1H), 17.4 (s, 1H); 19F NMR (d6-CDCl3, 376 MHz) δ (ppm) −26.90; −29.50 (d, J=978.89 Hz, 1F); 31P NMR (d6-CDCl3, 162 MHz) δ (ppm) 1.87-7.91 (d, J=978.33 Hz, 1P); and MS (ESI, EI+) m/z=428 (MH).

Example 9 1-(2-cyclopropylethyl)-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

1-(2-cyclopropylethyl)-6-fluoro-1,2-dihydro-4-hydroxy-2-oxoquinoline-3-carbonitrile (65 mg, 0.23 mmol), diethyl(1-amino-4-methanesulfonaminephenyl)phosphonate (154 mg, 0.47 mmol) and trimethylaluminium (716 μl, 1.43 mmol) were mixed together in 1,4-dioxane (1.2 ml) and stirred at 85° C. overnight. The reaction mixture was quenched with water (4 ml). A brown solid was collected and washed with aqueous solution of HCl (2N) The crude material was purified using preparative reverse phase HPLC to Example 9 (37 mg, 28%), which was a brown solid. Example 9 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.78-0.83 (m, 1H), 1.16-1.22 (t, J=7.01 Hz, 3H), 1.55 (q, J=7.45 Hz, 2H), 3.06 (s, 3H), 4.03 (t, J=7.11 Hz, 2H), 4.30 (m, 2H), 7.18 (t, J=8.3 Hz, 1H), 7.50-7.62 (m, 4H), 8.23 (t, J=6.56 Hz, 1H), 10.18 (s, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −103.5 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) −0.24 (s, 1P); and MS (ESI, EI+) m/z=549(MH+).

Example 10 1-(2-cyclopropylethyl-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

To as stirred solution of example 9 (45 mg, 0.082 mmol) in dichloromethane (4 ml) was added trimethylsilyl bromide (87 μl, 0.65 mmol) under nitrogen. The reaction mixture was stirred at room temperature overnight. Trimethylsilyl bromide (1 eq) was added every 5 hours until the expected product was formed. The reaction mixture was then quenched with water. A white solid was filtered, washed with water and dried to yield Example 10 (25 mg, 59%), which was a white solid. Example 10 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.03 (s, 2H), 0.36 (s, 2H), 0.79 (s, 1H), 1.51 (s, 2H), 3.03(s, 3H), 4.25 (s, 2H), 7.11 (s, 1H), 7.44-7.51 (m, 4H), 8.19 (s, 1H), 10.06 (s, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −104.80 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) −6.88 (s, 1P); and MS (ESI, EI+) m/z=521 (MH+).

Example 11 1-(2-cyclopropylethyl)-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

To a stirred solution of example 10 (22 mg, 0.042 mmol) in dichloromethane (2 ml) and a few drops of dimethylformamide, oxalyl chloride (6 μl, 0.0063 mmol) was added dropwise under nitrogen. The reaction mixture was stirred at room temperature under nitrogen for 24 hours. Methanol (1 ml) was then added and the mixture stirred for one hour. Solvents were concentrated under reduced pressure and the crude material purified using preparative reverse phase HPLC to give Example 11 (20 mg, 89%), which was a white solid. Example 11 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.06 (s, 2H), 0.39 (s, 2H), 0.82 (s, 1H), 1.54 (s, 2H), 3.06 (s, 3H), 3.59 (d, J=13.25 Hz, 3H), 4.30 (s, 2H), 7.17 (s, 1H), 7.49-7.59 (m, 4H), 8.21 (t, J=6.18 Hz, 1H), 10.2 (s, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −103.43 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) 1.57 (s, 1P); and MS (ESI, EI+) m/z=535 (MH+).

Example 12 1-(2-cyclopropylethyl)-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline sodium salt

Example 12 was obtained from example 11 as described for example 2 and was a yellow powder. Example 12 was characterized by the following spectroscopic data: 1H NMR (d6-DMSO, 400 MHz) δ (ppm) 0.06 (s, 2H), 0.39 (s, 2H), 0.82 (s, 1H), 1.54 (s, 2H), 3.06 (s, 3H), 3.59 (d, J=13.25 Hz, 3H), 4.30 (s, 2H), 7.17 (s, 1H), 7.49-7.59 (m, 4H), 8.21 (t, J=6.18 Hz, 1H), 10.2 (s, 1H); 19F NMR (d6-DMSO, 376 MHz) δ (ppm) −103.43 (s, 1F); 31P NMR (d6-DMSO, 162 MHz) δ (ppm) 1.98 (s, 1P); and MS (ESI, EI+) m/z=535 (MH+).

Example 13 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-3,3-dimethylbutyl)naphthalene-2(4H)-one

In a sealed tube and under nitrogen atmosphere, the trimethyl aluminium in toluene (0.924 mmol, 7 eq) was slowly added to the intermediate 18 (0.132 mmol, 1 eq) in 1,4 dioxane. The reaction mixture was stirred at 185° C. overnight. The reaction was quenched with water. A solid was isolated by filtration, washed with water then purified by chromatography on silica gel to give Example 13 (10 mg, 14%), which was a white solid. Example 13 was characterized by the following spectroscopic data: 1H NMR (Acetone-d6, 400 MHz) δ (ppm) 0.53-0.60 (m, 1H), 0.81 (2s, 9H), 0.84-0.92 (m, 1H), 1.32 (t, J=6.95 Hz, 3H), 1.58 (2s, 3H), 1.96-1.99 (m, 1H), 2.31-2.04 (m, 1H), 3.10 (s, 3H), 4.14-4.21 (m, 2H), 7.47 (td, J=6.95 Hz and J=2.12 Hz, 1H), 7.61-7.70 (m, 3H), 7.79-7.85 (m, 2H), 8.87 (d, J=7.86 Hz, 1H), 15.34 (br s, 1H); 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −0.91 (s, 1P); and MS (ESI, EI+) m/z=560 (MH+).

Example 14 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3,3-dimethylbutyl)naphthalene-2(4H)-one

Example 14 was synthesised from example 13, as described for example 10, except for heating to reflux for 2 hours and was a white powder. Example 14 was characterized by the following spectroscopic data: 1H NMR (Acetone-d6, 400 MHz) δ (ppm) according to expected structure; 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −4.05 (s, 1P); and MS (ESI, EI +) m/z=532 (MH+).

Example 15 1-(2-cyclopropylethyl)-(1-ethoxy-1-oxo-4H-7-(N-ethyl)-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-6-fluoro-dihydro-4-hydroxy-2-oxoquinoline

Example 15 was obtained as a by-product from the synthesis of example 20 and was a brown solid. Example 15 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.06 (m, 2H), 0.37 (m, 2H), 0.78-0.83 (m, 1H), 1.16-1.22 (t, J=7.01 Hz, 3H), 1.32 (t, J=6.20 Hz, 3H) 1.55 (q, J=7.45 Hz, 2H), 3.06 (s, 3H), 4.03 (q, J=7.11 Hz, 2H), 4.17 (m, 2H),4.30 (m, 2H), 7.18 (t, J=8.3 Hz, 1H), 7.50-7.62 (m, 4H), 8.23 (t, J=6.56 Hz, 1H), 10.18 (s, 1H); 19F NMR (DMSO-d6, 376 MHz) δ (ppm) −103.25 (s, 1F); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) −0.80 (s, 1P); and MS (ESI, EI+) m/z=577 (MH+). Example 15 is equivalent to Compound 13.

Example 16 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3,3-dimethylbutyl)naphthalene-2(4H)-one

In a sealed tube and under nitrogen atmosphere, the trimethyl aluminium in toluene (7 eq) was slowly added to the intermediate 36 (1 eq) in 1,4 dioxane. The reaction mixture was stirred at 185° C. overnight. The reaction was quenched with water. A solid was isolated by filtration, washed with water then purified by chromatography on silica gel to give Example 16, which was a white solid. Example 16 was characterized by the following spectroscopic data: 1H NMR (Acetone-d6, 400 MHz) δ (ppm) 0.53-0.60 (m, 1H), 0.81 (2s, 9H), 0.84-0.92 (m, 1H), 1.32 (t, J=6.95 Hz, 3H), 1.58 (2s, 3H), 1.96-1.99 (m, 1H), 2.31-2.04 (m, 1H), 3.10 (s, 3H), 4.14-4.21 (m, 2H), 7.47 (td, J=6.95 Hz and J=2.12 Hz, 1H), 7.61-7.70 (m, 3H), 7.79-7.85 (m, 2H), 8.87 (d, J=7.86 Hz, 1H), 15.34 (br s, 1H); 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −0.91 (s, 1P); and MS (ESI, EI+) m/z=560 (MH+). Example 16 is equivalent to Compound II-41.

Example 17 1-(1-ethoxy-1-oxo-4H-7-N-ethyl-methanesulfonaminyl-benzo-[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3,3-dimethylbutyl)naphthalene-2(4H)-one

Example 17 was obtained as a by-product from the synthesis of Example 16, which was a white solid. Example 17 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.53-0.60 (m, 1H), 0.74 (2s, 9H), 0.84-0.92 (m, 1H), 1.30 (t, J=7.00 Hz, 3H), 1.32 (t, J=6.20 Hz, 3H), 1.58 (s, 3H), 1.96-1.99 (m, 1H), 2.31-2.04 (m, 1H), 3.02 (s, 3H), 3.86-3.91 (q, J=7.05 Hz, 2H), 4.17 (m, 2H), 7.47 (t, J=6.91 Hz, 1H), 7.66-7.68 (m, 3H), 7.86 (d, J=9.22 Hz, 1H), 7.93 (d, J=13.83 Hz, 1H), 8.27 (d, J=6.91 Hz, 1H), 15.38 (brs, 1H); 31P NMR (CDCl3, 162 MHz) δ (ppm) −1.37 (s, 1P); and MS (ESI, EI+) m/z=588 (MH+). Example 17 is equivalent to Compound II-44.

Example 18 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3,3-dimethylbutyl)naphthalene-2(4H)-one

Example 18 was synthesized from Example 16. To as stirred solution of example 16 (45 mg, 0.082 mmol) in dichloromethane (4 ml) was added trimethylsilyl bromide (87 μl, 0.65 mmol) under nitrogen. The reaction mixture was stirred at room temperature overnight. Trimethylsilyl bromide (1 eq) was added every 5 hours until the expected product was formed. The reaction mixture was then quenched with water. A white solid was filtered, washed with water and dried to yield example 18, which is a white solid. Example 18 was characterized by the following spectroscopic data: 1H NMR (Acetone-d6, 400 MHz) δ (ppm), 0.35-0.45 (m, 1H), 0.69 (s, 9H), 0.74-0.78 (m, 1H), 1.86-1.92 (m, 1H), 2.17-2.24 (m, 1H), 3.03 (s, 1H), 7.40-7.50 (m, 4H), 7.62 (m, 2H), 8.13-8.15 (d, J=7.73 Hz, 1H), 10.06 (s, 1H), 14.32 (s, 1H), 14.84 (s, 1H); 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −7.30 (s, 1P); and MS (ESI, EI+) m/z=532 (MH+). Example 18 is equivalent to Compound II-21.

Example 19 1-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3,3-dimethylbutyl)naphthalene-2(4H)-one

Example 19 was synthesized from example 18 as described for example 11, which was a white solid. Example 19 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.38-0.44 (m, 1H), 0.69 (s, 9H), 0.72-0.76 (m, 1H), 1.51 (s, 3H), 3.07 (s, 3H), 4.01-4.03 (t, J=12.05 Hz, 3H), 7.46-7.66 (m, 6H), 8.15 (d, J=7.60 Hz, 1H), 10.19 (s, 1H), 15.02 (s, 1H), 15.48 (s, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 1.15-1.23 (d, J=13.7 Hz, 1P); and MS (ESI, EI+) m/z=546 (MH+). Example 19 is equivalent to Compound II-1.

Example 20 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3-methylbutyl)naphthalene-2(4H)-one

Intermediate 42 (1 eq) and intermediate 26 (1 eq), dissolved in dioxane, were stirred under nitrogen in a sealed tube, at 80° C., for 16 hours. The reaction mixture was then saturated with ammonia (NH3) at 0° C. and heated at 100° C. for 16 hours. The reaction mixture was concentrated under reduced pressure and purified using preparative HPLC to give Example 20, which was a white solid. Example 20 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.51-0.62 (m, 1H), 0.71-0.78 (m, 6H), 0.80-0.87 (m, 1H), 1.35-1.41 (m, 4H), 1.61 (s, 3H), 1.84-1.94 (m, 1H), 2.27-2.37 (m, 1H), 3.08 (s, 3H), 4.17-4.21 (m, 2H), 7.33-7.39 (m, 1H), 7.42-7.47 (m, 2H), 7.59-7.63 (m, 1H), 7.71-7.76 (m, 2H), 7.91-7.95 (m, 1H), 8.25-8.29 (m, 1H), 15.30 (d, J=6.0 Hz, 0.15H), 15.50 (d, J=12.0 Hz, 0.5 H), 15.90 (d, J=5.08 Hz, 0.5H), 16.12 (d, J=10.62 Hz, 0.5H); 31P NMR (CDCl3, 162 MHz) δ (ppm) 0.41-0.51 (m, 1P); and MS (ESI, EI+) m/z=546 (MH+). Example 20 is equivalent to Compound II-42.

Example 21 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3-methylbutyl)naphthalene-2(4H)-one

Example 20 (1 eq) and tetramethylsilylbromide (10 eq) dissolved in 1,2-dichloroethane (10 ml), were stirred in sealed tube, at 60° C., for 2 hours. The reaction mixture was then evaporated, diluted in methanol (1 ml) and added to HCl 1N. (15 ml). The precipitate was filtered, washed with water and dried over P2O5 to give Example 21, which was a yellow solid. Example 21 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.53-0.56 (m, 1H), 0.72-0.77 (dd, J=7.37 Hz, J=6.61 Hz, 6H), 0.81-0.90 (m, 1H), 1.35-1.39 (m, 1H), 1.57 (s, 3H), 1.86-1.92 (t, J=11.15 Hz, 3H), 2.29-2.38 (m, 1H), 3.12 (s, 3H), 7.32-7.36 (t, J=8.07 Hz, 1H), 7.41-7.46 (m, 2H), 7.59-7.62 (t, J=7.70 Hz, 1H), 7.70 (d, 8.08, 1H), 7.85 (d, J=15.78 Hz, 1H), 8.26 (s, 2H), 15.23 (s, 0.5H), 15.43 (s, 0.5H), 16.36 (brs, 1H); 31P NMR (CDCl3, 162 MHz) δ (ppm) −7.15 (s, 1P); and MS (ESI, EI+) m/z=518 (MH+). Example 21 is equivalent to Compound II-22.

Example 22 1-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-(3-methylbutyl)naphthalene-2(4H)-one

Example 22 was synthesized from example 21 as described for example 11, which was a white solid. Example 22 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.52-0.60 (m, 1H), 0.71-0.78 (m, 6H), 0.82-0.86 (m, 1H), 1.33-1.42 (m, 1H), 1.55-1.59 (m, 3H), 1.86-1.93 (m, 1H), 2.30-2.36 (m, 1H), 3.08 (d, J=3.30 Hz), 3.79-3.85 (m, 3H), 7.34-7.45 (m, 3H), 7.60-7.63 (t, J=7.34 Hz, 1H), 7.72-7.80 (m, 2H), 8.00-8.11 (m, 1H), 8.26-8.29 (m, 1H), 15.28 (d, J=5.42 Hz, 0.5H), 15.50 (d, J=9.04 Hz, 0.5 H), 16.05 (s, 0.5H), 16.29 (d, J=9.04 Hz, 0.5 H); 31P NMR (CDCl3, 162 MHz) δ (ppm) 2.25-2.34 (m, 1P); and MS (ESI, EI+) m/z=532 (MH+). Example 22 is equivalent to Compound II-2.

Example 23 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-isobutyl-naphthalene-2(4H)-one

Example 23 was synthesized from intermediate 26 and intermediate 49 as described for example 20 and was a white lyophilized powder. Example 23 was characterized by the following spectroscopic data: 1H NMR (Acetone-d6, 400 MHz) δ (ppm) 0.54-0.57 (m, 3H), 0.64-0.66 (m, 3H), 1.22 (m, 1H), 1.32 (s, 3H), 1.53 (s, 3H), 2.35 (m, 2H), 3.11 (s, 3H), 4.14-4.19 (m, 2H), 7.45-7.49 (t, J=7.00 Hz, 1H), 7.64-7.67 (m, 3H), 7.81 (d, J=7.62 Hz, 1H), 7.86 (m, 1H), 8.29 (d, J=7.10 Hz, 1H), 9.02 (brs, 1H), 15.30 (brs, 0.5H), 15.45-15.48 (m, 1H), 15.70 (brs, 0.5H); 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −1.00 (s, 1P); and MS (ESI, EI+) m/z=532 (MH+). Example 23 is equivalent to Compound II-42.

Example 24 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-isobutyl-naphthalene-2(4H)-one

Example 24 was synthesized from example 23 as described for example 21, which was a beige powder. Example 24 was characterized by the following spectroscopic data: MS (ESI, EI+) m/z=504 (MH+). Example 24 is equivalent to Compound II-45.

Example 25 1-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-8-hydroxy-3-methyl-3-isobutyl-naphthalene-2(4H)-one

Example 25 was synthesized from example 24 as described for example 11, which was a white lyophilized powder. Example 25 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 0.56-0.66 (m, 6H), 1.27 (m, 1H), 1.53 (s, 3H), 1.95-1.98 (m, 1H), 2.37-2.40 (m, 1H), 3.08 (s, 3H), 3.77-3.86 (m, 3H), 7.35-7.47 (m, 3H), 7.59-7.76 (m, 4H), 8.28 (m, 1H), 15.31 (s, 0.5H), 15.53-15.58 (d, J=18.10 Hz, 0.5H), 15.96-16.04 (d, J=29.60 Hz, 0.5H), 16.28-16.32 (d, J=16.26 Hz, 0.5H); 31P NMR (CDCl3, 162 MHz) δ (ppm) 1.97 (s, 1P), 2.02 (s, 1P); and MS (ESI, EI+) m/z=518 (MH+). Example 25 is equivalent to Compound II-46.

Example 26 1(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-3-(2-chloro-3-fluorobenzyl)-8-hydroxy-3-methyl-naphthalene-2(4H)-one

Example 26 was synthesized from intermediate 26 and intermediate 57 as described for example 20, which was a white lyophilized powder. Example 26 was characterized by the following spectroscopic data: 1H NMR (CDCl3, 400 MHz) δ (ppm) 1.34-1.42 (m, 3H), 1.75 (s, 3H), 3.08 (s, 3H), 3.46-3.50 (m, 2H), 4.16-4.21 (m, 2H), 6.68-6.75 (m, 2H), 7.29-7.40 (m, 1H), 7.43-7.60 (m, 3H), 7.65-7.70 (m, 3H), 8.15 (d, J=7.91 Hz, 1H), 15.16 (s, 1H), 15.30 (d, J=13.41 Hz, 1H), 15.55-15.67 (t, J=23.50 Hz, 1H); 31P NMR (CDCl3, 162 MHz) δ (ppm) −0.23 (s, 1P), −0.01 (s, 1P), 0.23 (s, 1P) (mixture of diastereoisomers); 19F NMR (CDCl3, 376 MHz) δ (ppm) −118.51 (s, 1F), −118.44 (s, 1F), −118.33 (s, 1F), −118.10 (s, 1F) (mixture of diastereoisomers); and MS (ESI, EI) m/z=616 (MH). Example 26 is equivalent to Compound II-43.

Example 27 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-3-(2-chloro-3-fluorobenzyl)-8-hydroxy-3-methyl-naphthalene-2(4H)-one

Example 27 was synthesized from example 26 as described for example 21, which was a white lyophilized powder. Example 27 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.62 (s, 3H), 3.02 (s, 3H), 3.19-3.55 (m, 2H), 6.58-6.63 (m, 2H), 7.00 (t, J=7.46 Hz, 1H), 7.40-7.52 (m, 4H), 7.64-7.67 (t, J=6.74 Hz, 1H), 7.80 (d, J=7.07 Hz, 1H), 7.98 (d, J=7.50 Hz, 1H), 10.03 (s, 1H), 13.91 (s, 1H), 14.61 (s, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) −8.15 (s, 1P); 19F NMR (DMSO-d6, 376 MHz) δ (ppm) −119.67 (s, 1); and MS (ESI, EI) m/z=588 (MH). Example 27 is equivalent to Compound II-29.

Example 28 1-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-3-(2-chloro-3-fluorobenzyl)-8-hydroxy-3-methyl-naphthalene-2(4H)-one

Example 28 was synthesized from example 27 as described for example 11, which was a white lyophilized powder. Example 28 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 1.63 (s, 3H), 3.04 (s, 3H), 3.19-3.55 (m, 2H), 3.56 (s, 3H), 6.56 (brs, 1H), 6.67 (brs, 1H), 7.00 (t, J=7.80 Hz, 1H), 7.43-7.81 (m, 6H), 7.9 (d, J=7.80 Hz, 1H), 10.17 (brs, 1H), 14.78 (brs, 1H), 15.13 (brs, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 1.21 (s, 1P); 19F NMR (DMSO-d6, 376 MHz) δ (ppm) −119.50 (s, 1F); and MS (ESI, EI+) m/z=604 (MH+). Example 28 is equivalent to Compound II-9.

Example 29 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-iodo-pyridin-2-one

Example 29 was synthesized from intermediate 26 and intermediate 60 as described for example 20, which was a white lyophilized powder. Example 29 was characterized by the following spectroscopic data: 1H NMR (Acetone-d6, 400 MHz) δ (ppm) 1.05(s, 9H), 1.40 (t, J=6.9 Hz, 3H), 1.68-1.72 (m, 2H), 2.86 (s, 3H), 3.90 (m, 2H), 4.10-4.14 (m, 2H), 7.50-7.54 (m, 3H), 8.4 (s, 1H), 10.2 (brs, 1H); 31P NMR (Acetone-d6, 162 MHz) δ (ppm) −1.00 (s, 1P); and MS (ESI, EI+) m/z=623 (MH+). Example 29 is equivalent to Compound III-1.

Example 30

Example 30 was synthesized from intermediate 26 and intermediate 61 as described for example 20. Three compounds were obtained from the purification by preparative HPLC.

Example 30a 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-phenyl-pyridin-2-one

Example 30a is a white lyophilized powder. Example 30a was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.97 (s, 9H), 1.16 (t, J=6.86 Hz, 3H), 1.61 (t, J=7.96 Hz, 2H), 3.01 (s, 3H), 3.66-3.70 (m, 2H), 4.02-4.05 (m, 2H), 7.36-7.39 (m, 1H), 7.39-7.43 (t, J=7.15 Hz, 2H), 7.50-7.55 (m, 5H), 8.21 (s, 1H), 9.51 (brs, 1H), 14.19 (brs, 0.5H), 17.75 (brs, 0.5H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 0.27 (s, 1P); and MS (ESI, EI) m/z=571 (MH). Example 30a is equivalent to Compound III-2.

Example 30b 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl-4-hydroxy-pyridin-2-one

Example 30b is a white lyophilized powder. Example 30b was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.95 (s, 9H), 1.16 (t, J=7.20 Hz, 3H), 1.56 (t, J=8.06 Hz, 2H), 2.99 (s, 3H), 3.80-3.82 (m, 2H), 3.93-3.95 (m, 2H), 6.18 (m, 1H), 7.33-7.48 (m, 4H), 7.94 (m, 1H), 10.16 (s, 1H), 13.94 (brs, 1H), 16.97 (brs, 1 H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 0.68 (s, 1P); and MS (ESI, EI+) m/z=497 (MH+). Example 30b is equivalent to Compound III-3.

Example 30c 1-(1-ethoxy-1-oxo-4H-7-(N-ethyl-methanesulfonaminyl)-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-iodo-pyridin-2-one

Example 30c is a white lyophilized powder. Example 30c was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.97 (s, 9H), 1.16 (t, J=6.86 Hz, 3H), 1.21 (t, J=7.03 Hz, 3H), 1.61 (t, J=7.96 Hz, 2H), 3.01 (s, 3H), 3.38-3.42 (m, 2H), 3.66-3.70 (m, 2H), 4.02-4.05 (m, 2H), 7.36-7.39 (m, 1H), 7.39-7.43 (t, J=7.15 Hz, 2H), 7.50-7.55 (m, 5H), 8.21 (s, 1H), 9.51 (brs, 1H), 16.97 (brs, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) −0.20 (s, 1P); and MS (ESI, EI+) m/z=601 (MH+). Example 30c is equivalent to Compound III-4.

Example 31 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-phenyl-pyridin-2-one

Example 31 was synthesized from example 30a as described for example 21, which was a beige solid. Example 31 was characterized by the following spectroscopic data; and MS (ESI, EI+) m/z=545 (MH+). Example 31 is equivalent to Compound III-5.

Example 32 1-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-3,3-dimethylbutyl)-4-hydroxy-5-phenyl-pyridin-2-one

Example 32 was synthesized from example 31 as described for example 11, which was a white lyophilized powder. Example 32 was characterized by the following spectroscopic data: 1H NMR (MeOD, 400 MHz) δ (ppm) 1.05 (s, 9H), 1.68-1.72 (t, J=8.18 Hz, 3H), 3.04 (s, 3H), 3.62 (d, J=12.27 Hz, 3H), 4.10-4.14 (m, 2H), 7.36-7.46 (m, 4H), 7.53 (d, J=7.06 Hz, 2H), 7.63 (d, J=11.20 Hz, 2H), 7.89 (brs, 1H); 31P NMR (CDCl3, 162 MHz) δ (ppm) 3.36 (s, 1P); and MS (ESI, EI) m/z=557 (MH). Example 32 is equivalent to Compound III-6.

Example 33 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-methoxy-5-phenyl-pyridin-2-one

Example 33 was synthesized from intermediate 26 and intermediate 58 as described for example 20, which was a white lyophilized powder. Example 33 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.93 (s, 9H), 1.07-1.10 (t, J=7.24 Hz, 3H), 1.50 (m, 2H), 3.96 (s, 3H), 3.48-3.50 (m, 1H), 3.72-3.75 (m, 1H), 3.81 (s, 3H), 3.90-3.97 (m, 2H), 6.40 (d, J=7.72 Hz, 1H), 7.19-7.22 (m, 1H), 7.39-7.42 (m, 2H), 7.93 (d, J=7.72 Hz, 1H), 9.85 (s, 1H), 11.40 (s, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 3.91 (s, 1P); and MS (ESI, EI+) m/z=511 (MH+. Example 33 is equivalent to Compound III-7.

Example 34 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-thiophenyl-pyridin-2-one

Example 34 was synthesized from intermediate 26 and intermediate 62 as described for example 20, which was a white lyophilized powder. Example 34 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.98 (s, 9H), 1.15-1.18 (t, J=6.95 Hz, 3H), 1.58-1.62 (t, J=7.94 Hz, 2H), 2.98 (s, 3H), 3.94-3.90 (m, 2H), 3.99-4.01 (m, 2H), 7.47-7.52 (m, 4H), 7.58 (s, 1H), 7.91 (s, 1H), 8.25-8.28 (m, 1H), 14.20 (brs, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 0.43 (s, 1P); and MS (ESI, EI+) m/z=579 (MH+). Example 34 is equivalent to Compound III-8.

Example 35 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-(thiophen-3-yl)-pyridin-2-one

Example 35 was synthesized from example 34 as described for example 21, which was a white lyophilized powder. Example 35 was characterized by the following spectroscopic data; and MS (ESI, EI+) m/z=551(MH+). Example 35 is equivalent to Compound I-70.

Example 36 1-(1-methoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-(thiophen-3-yl)-pyridin-2-one

Example 36 was synthesized from example 35 as described for example 11, which was a white lyophilized powder. Example 36 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.97 (s, 9H), 1.59-1.63 (t, J=8.00 Hz, 2H), 3.04 (s, 3H), 3.50 (d, J=12.08 Hz, 3H), 4.03 (m, 2H), 7.50-7.59 (m, 5H), 7.90 (s, 1H), 8.31 (s, 1H), 10.14 (brs, 1H), 14.22 (brs, 1H), 17.86 (brs, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) 1.91 (s, 1P); and MS (ESI, EI+) m/z=565 (MH+). Example 36 is equivalent to Compound I-71.

Example 37 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-(pyrazol-1-yl)-pyridin-2-one

Example 37 was synthesized from intermediate 26 and intermediate 63 as described for example 20, which was a white lyophilized powder. Example 37 was characterized by the following spectroscopic data: 1H NMR (MeOD, 400 MHz) δ (ppm) 1.03 (s, 9H), 1.32 (m, 3H), 1.69 (m, 2H), 3.04 (s, 3H), 4.06 (m, 4H), 6.51 (s, 1H), 7.48 (m, 2H), 7.64-7.71 (m, 2H), 8.12 (m, 1H); 31P NMR (MeOD, 162 MHz) δ (ppm) 1.82 (s, 1P); and MS (ESI, EI+) m/z=563 (MH+). Example 37 is equivalent to Compound III-9.

Example 38 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-(pyrazol-1-yl)-pyridin-2-one

Example 38 was synthesized from example 37 as described for example 21, which was a white lyophilized powder. Example 38 was characterized by the following spectroscopic data: 1H NMR (MeOD, 400 MHz) δ (ppm) 1.03 (s, 9H), 1.62-1.66 (m, 2H), 2.98 (s, 3H), 3.94 (m, 2H), 6.46 (s, 1H), 7.22 (m, 1H), 7.42 (m, 1H), 7.66 (m, 1H), 7.86 (s, 1H), 8.07 (s, 1H); 31P NMR (MeOD, 162 MHz) δ (ppm) −8.74 (s, 1P); and MS (ESI, EI+) m/z=535 (MH+). Example 38 is equivalent to Compound III-10.

Example 39 1-(1-ethoxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-(thiophen-2-yl)-pyridin-2-one

Example 39 was synthesized from intermediate 26 and intermediate 65 as described for example 20, which was a white lyophilized powder. Example 39 was characterized by the following spectroscopic data: 1H NMR (DMSO-d6, 400 MHz) δ (ppm) 0.94 (s, 9H), 1.17-1.20 (t, J=6.90 Hz, 3H), 1.58-1.62 (t, J=8.30 Hz, 2H), 3.04 (s, 3H), 3.95-4.01 (brs, 4H), 7.08-7.10 (m, 1H), 7.49-7.58 (m, 5H), 8.39 (s, 1H), 10.15 (brs, 1H), 14.32 (brs, 1H), 17.68 (brs, 1H); 31P NMR (DMSO-d6, 162 MHz) δ (ppm) −0.14 (s, 1P); and MS (ESI, EI+) m/z=579 (MH+). Example 39 is equivalent to Compound III-8.

Example 40 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-5-(thiophen-2-yl)-pyridin-2-one

Example 40 was synthesized from example 39 as described for example 21, which was a white lyophilized powder. Example 40 was characterized by the following spectroscopic data: 1H NMR (MeOD, 400 MHz) δ (ppm) 1.04 (s, 9H), 1.64-1.68 (m, 2H), 3.03 (s, 3H), 4.00-4.04 (m, 2H), 7.05 (t, J=4.39 Hz, 1H), 7.36-7.41 (m, 3H), 7.54-7.56 (d, J=9.04 Hz, 1H), 7.67-7.71 (d, J=15.06 Hz, 1H), 7.97 (s, 1H); 31P NMR (MeOD, 162 MHz) δ (ppm) −3.28 (s, 1P); and MS (ESI, EI+) m/z=551 (MH+). Example 40 is equivalent to Compound III-11.

Example 41 1-(1-hydroxy-1-oxo-4H-7-methanesulfonaminyl-benzo[1,2,4]phosphadiazine-3-yl)-1-N-(3,3-dimethylbutyl)-4-hydroxy-pyridin-2-one

Example 41 was synthesized from example 40 following the procedure as described for example 21 and was a white powder. Example 41 was characterized by the following spectroscopic data: 1H NMR (MeOD, 400 MHz) δ (ppm) 1.02 (s, 9H), 1.59-1.60 (m, 2H), 3.01 (s, 3H), 3.91-3.93 (m, 2H), 5.97 (m, 1H), 7.28 (1H), 7.52-7.64 (m, 3H). 31P NMR (MeOD, 162 MHz) δ (ppm) −3.29-3.69 (m, 1P). MS (ESI, EI+) m/z=466 (MH+). Example 41 is equivalent to Compound III-12.

Example 42 HCV Polymerase Assay

The HCV polymerase assay was performed in 96-well streptavidin-coated microtiter plates (Pierce) using 50 nM HCV genotype 1b polymerase (strain J4) from Replizyme, 15 μM bromo-UTP, 1 μg/ml 5′-biotynilated oligo (rU12), 1 μg/ml poly(rA) in 20 mM Tris-HCl pH 7.5, 5 mM MgCl2, 0.5 μg/ml BSA, 1 mM DTT, 0.02 U/μl RNasin, 5% DMSO and 25 mM KCL. The 60-μl reaction was incubated at 35° C. for 60 min and terminated by adding 20 μL 0.5 M EDTA pH 8.0. The BrUTP incorporated onto the biotinylated primer was quantified by ELISA using a peroxidase-labeled anti-BrdU monoclonal antibody (Roche) and TMB (Sigma) substrate and the plates were read at 630 nm with the Tecan Sunrise Stectrophotometer. The compounds were routinely solubilised at a concentration of 15 mM in DMSO and tested at a variety of concentrations in assay buffer containing a final DMSO concentration of 5%. The IC50 values were determined from the percent inhibition versus concentration data using a sigmoidal non-linear regression analysis based on four parameters with Tecan Magellan software.

The biological results are summarized in Table 1 (IC50), wherein A represents a value smaller than 100 nM, B represents a value between 100 nM to 10 μM, and C represents a value greater than 10 μM.

Example 43 HCV Relicon Assay

General procedure: Huh-7 cells containing HCV Con1 subgenomic replicon (GS4.1 cells) were grown in Dulbecco's Modified Eagle Medium (DMEM) supplemented with 10% fetal bovine serum (FBS), 2 mM L-glutamine, 110 mg/L sodium pyruvate, 1× non-essential amino acids, 100 U/mL penicillin-streptomycin, and 0.5 mg/mL G418 (Invitrogen). For dose-response testing, the cells were seeded in 96-well plates at 7.5×103 cells/well in a volume of 50 μL, and incubated at 37° C./5% CO2. Three hours after plating, 50 μL of ten 2-fold serial dilutions of compounds (highest concentration, 75 μM) were added, and cell cultures were incubated at 37° C./5% CO2 in the presence of 0.5% DMSO. Alternatively, compounds were tested at a single concentration of 15 μM. In all cases, Huh-7 cells lacking the HCV replicon served as negative control. The cells were incubated in the presence of compounds for 72 hr after which they were monitored for expression of the NS4A protein by enzyme-linked immunosorbent assay (ELISA). For this, the plates were then fixed for 1 min with acetone/methanol (1:1, v/v), washed twice with phosphate-buffered saline (PBS), 0.1% Tween 20, blocked for 1 hr at room temperature with TNE buffer containing 10% FBS and then incubated for 2 hr at 37° C. with the anti-NS4A mouse monoclonal antibody A-236 (ViroGen) diluted in the same buffer. After washing three times with PBS, 0.1% Tween 20, the cells were incubated 1 hr at 37° C. with anti-mouse immunoglobulin G-peroxidase conjugate in TNE, 10% FBS. After washing as described above, the reaction was developed with O-phenylenediamine (Zymed). The reaction was stopped after 30 min with 2 N H2SO4, and absorbance was read at 492 nm using Sunrise Tecan spectrophotometer. EC50 values were determined from the % inhibition versus concentration data using a sigmoidal non-linear regression analysis based on four parameters with Tecan Magellan software. When screening at a single concentration, the results were expressed as % inhibition at 15 μM.

The biological results are summarized in Table 1 (EC50 and CC50), wherein A represents a value smaller than 100 nM, B represents a value between 100 nM to 10 μM, and C represents a value greater than 10 μM.

TABLE 1 Compound IC50 (μM) EC50 (μM) CC50 (μM) C C C B B C B B C B B C A B C A B C

The examples set forth above are provided to give those of ordinary skill in the art with a complete disclosure and description of how to make and use the claimed embodiments, and are not intended to limit the scope of what is disclosed herein. Modifications that are obvious to persons of skill in the art are intended to be within the scope of the following claims. All publications, patents, and patent applications cited in this specification are incorporated herein by reference as if each such publication, patent or patent application were specifically and individually indicated to be incorporated herein by reference.

Claims

1. A compound of Formula I′ or II″: or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof, wherein:

each R1 is independently H, alkyl, arylalkyl, heteroarylalkyl, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl;
each R2 is independently H, alkyl, aryl-CH2—, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl, or R1 and R2 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
each R4 is independently H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
each R4′ is independently H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
each R5 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R4 and R5 together form a part of a 3-8 membered heterocycloalkyl ring;
each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5 and R6 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
each R5′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, heteroaryl, —NR8R10, alkenyl, or alkynl;
each R6′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5′ and R6′ together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
each R12 is independently F, —OR8, —SR8, —NR8R9, alkyl, or aryl;
each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-4 aryl, heteroaryl, or heterocyclyl; or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9; and
each Y is independently O or S,
wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

2. The compound of claim 1, wherein each pair of R5 and R6 together independently forms a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring.

3. The compound of claim 1, wherein each pair of R5 and R6 together independently forms a benzo ring having formula (A): where

each * is a bond;
each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C1-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9, —OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′—O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9;
each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; and
n is an integer from 1 to 4.

4. The compound of claim 1, wherein each pair of R5′ and R6′ together independently forms a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring.

5. The compound of claim 4, wherein the compound of Formula I′ or II′ has the following formula I″ or II″: wherein

each A is independently CR18 or N;
each A′ is independently CR15R16, NR17, N, CR15, N-oxide, N—OR8—, S or O;
each R15 is independently a bond, H, halogen, —NR10SO2R8, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R10, —(C1-C6 alkylene)-NR9′S(O)2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
R16 is a bond, H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
R17 is a bond, H, alkyl, aryl-CH2—, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, sulfonyl, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
each m is independently an integer from 1 to 3;
R18 is a bond, H, halogen, —NR10SO2R8, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R10, —(C1-C6 alkylene)-NR9′S(O)2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl; and
Z has the following structures:

6. A compound of Formula I, II, or III: or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof;

each R1 is independently alkyl, H, arylalkyl, heteroarylalkyl, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl, sulfonyl, or heteroaryl;
each R2 is independently alkyl, aryl-CH2—, H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, aryl, arylalkyl, alkenyl, alkynyl, heterocyclylalkyl or heteroaryl;
each R5′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, heteroaryl, —NR8R10, alkenyl, or alkynl;
each R6′ is independently H, halogen, cyano, nitro, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
each R5 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl;
each R6 is independently H, halogen, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, —C(O)NR8R9, —C(O)OR8, alkyl, aryl, or heteroaryl, or R5 and R6 together form a part of a 3-8 membered cycloalkyl, aryl, heterocycloalkyl or heteroaryl ring;
each R12 is independently —OR8, —SR8, —NR8R9, alkyl, or aryl;
each R14 is independently H, halogen, alkyl, alkenyl, alkynyl, aryl, heterocyclyl, heteroaryl, cyano, nitro, OH, —NR10SO2R8, —OR8, —NR8R9, —C(O)R8, C(O)NR8R9, —OCH2C(O)NR8R9, —C(O)OR8, —O—(C1-C6 hydroxyalkyl), —O—(C6-C6 alkoxy), —O—(C1-C6 alkylene)-cyano, —O—(C1-C6 alkylene)-C(O)R9′, OCHR9′C(O)O—R8, —OCHR9′C(O)NHOH, —O—(C1-C6 alkyl)-C(O)NR8R9, —O—(C1-C6 alkylene)-NR9′C(O)R8, —O—(C1-C6 alkylene)-NR9′C(O)OR8, —O—(C1-C6 alkylene)-NR9′C(O)NR8R9, —OCHR9′C(O)NR8R9, —O—(C1-C6 alkylene)-S(O)R9′, —O—(C1-C6 alkyl)-S(O)2R9′, —O—(C1-C6 alkylene)-S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2NR8R9, —O—(C1-C6 alkylene)-NR9′S(O)2R8—O—(C1-C6 alkylene)-S(O)2R9′—O—(C1-C6 alkylene)-NR8R9, —(C1-C6 alkylene)-S(O)2R8, —(C1-C6 alkylene)-S(O)2NR8R9, —(C1-C6 alkylene)-S(O)R8, —(C1-C6 alkylene)-C(O)R8, —(C1-C6 alkylene)-C(O)NR8R9, —(C1l-C6 alkylene)-NR9′C(O)R8, —(C1-C6 alkylene)-NR9′S(O)2R8, —(C1-C6 alkylene)-NR9′C(O)OR8, —(C1-C6 alkylene)-NR9′C(O)NR8R9, —(C1-C6 alkylene)-NR9′S(O)2NR8R9, —(C1-C6 alkylene)-C(O)OR8, —(C1-C6 alkylene)-NR8R9, —NR8C(O)R9, —NR9′S(O)2NR8R9, —NR9′S(O)2NR8R10, —S(O)R9′, —S(O)2R9′, or —S(O)2NR8R9;
each n is independently an integer from 1 to 4;
each R8 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, heterocyclyl, C1-6 alkyl-C3-7 cycloalkylene, or C1-10 alkyl-siloxyl;
each R9 is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl, or R8 and R9 together with the N atom to which they are attached form heterocyclyl;
each R9′ is independently hydrogen, C1-6 alkyl, C2-6 alkenyl, C2-6 alkynyl, C3-7 cycloalkyl, C6-14 aryl, heteroaryl, or heterocyclyl; and
each R10 is independently H, alkyl, aryl, sulfonyl, C(O)R8, C(O)OR8 or C(O)NR8 R9;
wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, sulfonyl, or alkyl-cycloalkyl is optionally substituted.

7. The compound of claim 6 according to Formula Ia, IIa, or IIIa: or a single enantiomer, a mixture of an enantiomeric pair, an individual diastereomer, a mixture of diastereomers, or any tautomeric form thereof; or a pharmaceutically acceptable salt, solvate, or prodrug thereof.

8. The compound of claim 6, wherein the compound has the structure of Formula Ia.

9. The compound of claim 6, wherein the compound has the structure of Formula IIa.

10. The compound of claim 6, wherein the compound has the structure of Formula IIIa.

11. The compound of claim 6, wherein each alkyl, aryl, arylalkyl, heteroaryl, alkenyl, alkynyl, cycloalkyl, heterocyclyl, or alkyl-cycloalkyl is unsubstituted.

12. The compound of claim 6, wherein R1 is C1-6 alkyl.

13. The compound of claim 12, wherein R1 is 2-cyclopropylethyl.

14. The compound of claim 6, wherein R2 is hydrogen.

15. The compound of claim 6, wherein R6 is hydrogen or halogen.

16. The compound o claim 15, wherein R6 is fluoro.

17. The compound o claim 6, wherein R12 is C1-6 alkoxy.

18. The compound of claim 17, wherein R12 is methoxy.

19. The compound of claim 17, wherein R12 is ethoxy.

20. The compound of claim 6, wherein R12 is NH2.

21. The compound of claim 6, wherein R12 is OH.

22. The compound of claim 6, wherein R14 is hydrogen.

23. The compound of claim 6, wherein R14 is —NHSO2R8.

24. The compound of claim 23, wherein R8 is C1-6 alkyl.

25. The compound of claim 24, wherein R8 is methyl.

26. The compound of claim 6, wherein R1 is 2-cyclopropylethyl; R2 is hydrogen; R6 is fluoro; R12 is methoxy, ethoxy, fluoro, NH2 or OH; and R14 is hydrogen or —NHSO2Me.

27. The compound of claim 6 according to the following formula:

28. The compound of any of claim 6 according to the following formula:

29. The compound of any of claim 6 according to the following formula:

30. The compound of any of claim 6 according to the following formula:

31. The compound of any of claim 6 according to the following formula:

32. The compound of claim 6 according to the following formula:

33. The compound of any of claim 6 according to the following formula:

34. The compound of any of claim 6 according to the following formula:

35. The compound of any of claim 6 according to the following formula:

36. The compound of any of claim 6 according to the following formula:

37. The compound of any of claim 6 according to the following formula:

38. The compound of claim 6 according to the following formula:

39. A pharmaceutical composition comprising the compound of claim 1 and one or more pharmaceutically acceptable carriers.

40. The pharmaceutical composition of claim 39, farther comprising a second antiviral agent.

41. The pharmaceutical composition of claim 40, wherein the second antiviral agent is selected from the group consisting of an interferon, ribavirin, an interleukin, an NS3 protease inhibitor, a cysteine protease inhibitor, a phenathrenequinone, a thiazolidine, a benzanilide, a helicase inhibitor, a polymerase inhibitor, a nucleotide analogue, a nucleoside analogue, a liotoxin, acerulenin, an antisense phosphorothioate oligodeoxynucleotide, an inhibitor of IRES-dependent translation, and a ribozyme.

42. The pharmaceutical composition of claim 41, wherein the second antiviral agent is an interferon.

43. The pharmaceutical composition of claim 42, wherein the interferon is selected from the group consisting of pegylated interferon alpha 2a, interferon alphcon-1, natural interferon, albuferon, interferon beta-1a, omega interferon, interferon alpha, interferon gamma, interferon tau, interferon delta, and interferon gamma-1b.

44. The pharmaceutical composition of claim 39, wherein the composition is formulated for single dose administration.

45. The pharmaceutical composition of claim 39, wherein the composition is formulated as an oral, parenteral, or intravenous dosage form.

46. The pharmaceutical composition of claim 45, wherein the oral dosage form is a tablet or capsule.

47. The pharmaceutical composition of claim 39, wherein the compound is administered in a dose of about 0.5 milligram to about 1,000 milligram daily.

48. A method for treating or preventing an HCV infection, which comprises administering the compound of claim 1.

49. A method for treating or preventing an HCV infection, which comprises administering the pharmaceutical composition of claim 39.

50. A method of treating, preventing, or ameliorating one or more symptoms of a liver disease or disorder associated with an HCV infection, comprising administering the compound of claim 1.

51. A method of treating, preventing, or ameliorating one or more symptoms of a liver disease or disorder associated with an HCV infection, comprising administering the pharmaceutical composition of claim 39.

52. The method of any of claim 48, wherein the method comprises administering a second antiviral agent, in combination or alternation.

53. The method of any of claim 49, wherein the method comprises administering a second antiviral agent, in combination or alternation.

54. The method of claim 52, wherein the second antiviral agent is selected from the group consisting of an interferon, ribavirin, amantadine, an interleukin, a NS3 protease inhibitor, a cysteine protease inhibitor, a phenathrenequinone, a thiazolidine, a benzanilide, a helicase inhibitor, a polymerase inhibitor, a nucleotide analogue, a nucleoside analogue, a liotoxin, acerulenin, an antisense phosphorothioate ologodeoxynucleotide, an inhibitor of IRES-dependent translation, and a ribozyme.

55. The method of claim 53, wherein the second antiviral agent is selected from the group consisting of an interferon, ribavirin, amantadine, an interleukin, a NS3 protease inhibitor, a cysteine protease inhibitor, a phenathrenequinone, a thiazolidine, a benzanilide, a helicase inhibitor, a polymerase inhibitor, a nucleotide analogue, a nucleoside analogue, a liotoxin, acerulenin, an antisense phosphorothioate ologodeoxynucleotide, an inhibitor of IRES-dependent translation, and a ribozyme.

56. The method of claim 52, wherein the second antiviral agent is an interferon.

57. The method of claim 53, wherein the second antiviral agent is an interferon.

58. The method of claim 56, wherein the interferon is selected from the group consisting of pegylated interferon alpha 2a, interferon alphcon-1, natural interferon, albuferon, interferon beta-1a, omega interferon, interferon alpha, interferon gamma, interferon tau, interferon delta, and interferon gamma-1b.

59. The method of claim 57, wherein the interferon is selected from the group consisting of pegylated interferon alpha 2a, interferon alphcon-1, natural interferon, albuferon, interferon beta-1a, omega interferon, interferon alpha, interferon gamma, interferon tau, interferon delta, and interferon gamma-1b.

60. A method for inhibiting replication of a virus in a host, which comprises contacting the host with the compound of claim 1.

61. A method for inhibiting replication of a virus in a host, which comprises contacting the host with the pharmaceutical composition of claim 39.

62. The method of claim 60, wherein the host is a human.

63. The method of claim 61, wherein the host is a human.

64. A method for inhibiting replication of a virus, which comprises contacting the virus with the compound of claim 1.

65. A method for inhibiting replication of a virus, which comprises contacting the virus with the pharmaceutical composition of claim 39.

66. A method for inhibiting the activity of a polymerase, which comprises contacting the polymerase with the compound of claim 1.

67. A method for inhibiting the activity of a polymerase, which comprises contacting the polymerase with the pharmaceutical composition of claim 39.

68. The method of claim 66, wherein the polymerase is an HCV NS5B polymerase.

69. The method of claim 67, wherein the polymerase is an HCV NS5B polymerase.

Patent History
Publication number: 20090060866
Type: Application
Filed: Aug 27, 2008
Publication Date: Mar 5, 2009
Applicant: Idenix Pharmaceuticals, Inc. (Cambridge, MA)
Inventors: Cyril Dousson (Canet), Dominique Surleraux (Wauthier-Braine), Arlene Roland (Marsillargues), Claire Pierra (Montarnaud), Daniel Da Costa (Saint Jean de Vedas)
Application Number: 12/198,898
Classifications