β-d-2′-deoxy-2′-α-fluoro-2′-β-c-substituted-2-modified-n6-substituted purine nucleotides for HCV treatment

Information

  • Patent Grant
  • 10875885
  • Patent Number
    10,875,885
  • Date Filed
    Wednesday, July 1, 2020
    4 years ago
  • Date Issued
    Tuesday, December 29, 2020
    3 years ago
Abstract
A compound of the structure:
Description
FIELD OF THE INVENTION

The present invention is directed to nucleotide compounds and compositions and uses thereof to treat the Hepatitis C virus (“HCV”).


BACKGROUND OF THE INVENTION

Hepatitis C (HCV) is an RNA single stranded virus and member of the Hepacivirus genus. It is estimated that 75% of all cases of liver disease are caused by HCV. HCV infection can lead to cirrhosis and liver cancer, and if left to progress, liver failure which may require a liver transplant. Approximately 170-200 million people worldwide are infected, with an estimated 3-4 million infections in the United States.


RNA polymerase is a key component in the targeting of RNA single stranded viruses. The HCV non-structural protein NS5B RNA-dependent RNA polymerase is a key enzyme responsible for initiating and catalyzing viral RNA synthesis. As a result, HCV NS5B is an attractive target for the current drug discovery and development of anti-HCV agents. There are two major subclasses of NS5B inhibitors: nucleoside analogs, which are anabolized to their active triphosphates—which act as alternative substrates for the polymerase—and non-nucleoside inhibitors (NNIs), which bind to allosteric regions on the protein. Nucleoside or nucleotide inhibitors mimic natural polymerase substrate and act as chain terminators. They inhibit the initiation of RNA transcription and elongation of a nascent RNA chain.


In addition to targeting RNA polymerase, other RNA viral proteins may also be targeted in combination therapies. For example, HCV proteins that are additional targets for therapeutic approaches are NS3/4A (a serine protease) and NS5A (a non-structural protein that is an essential component of HCV replicase and exerts a range of effects on cellular pathways).


In December 2013, the first nucleoside NS5B polymerase inhibitor sofosbuvir (Sovaldi®, Gilead Sciences) was approved. Sovaldi® is a uridine phosphoramidate prodrug that is taken up by hepatocytes and undergoes intracellular activation to afford the active metabolite; 2′-deoxy-2′-α-fluoro-β-C-methyluridine-5′-triphosphate; see structures below:




embedded image


Sovaldi® is the first drug that has demonstrated safety and efficacy to treat certain types of HCV infection without the need for co-administration of interferon. Sovaldi® is the third drug with breakthrough therapy designation to receive FDA approval.


In 2014, the U.S. FDA approved Harvoni® (ledispasvir, a NS5A inhibitor, and sofosbuvir) to treat chronic hepatitis C virus genotype 1 infection. Harvoni® is the first combination pill approved to treat chronic HCV genotype 1 infection. It is also the first approved regimen that does not require administration with interferon or ribavirin. In addition, the FDA approved simeprevir (Olysio™) in combination with sofosbuvir (Sovaldi®) as a once-daily, all oral, interferon and ribavirin-free treatment for adults with genotype 1 HCV infection.


The U.S. FDA also approved AbbVie's VIEKIRA Pak™ in 2014, a multipill pack containing dasabuvir (a non-nucleoside NS5B polymerase inhibitor), ombitasvir (a NS5A inhibitor), paritaprevir (a NS3/4A inhibitor), and ritonavir. The VIEKIRA Pak™ can be used with or without the ribavirin to treat genotype 1 HCV infected patients including patients with compensated cirrhosis. VIEKIRA Pak™ does not require interferon co-therapy.


In July 2015, the U.S. FDA approved Technivie™ and Daklinza™ for the treatment of HCV genotype 4 and HCV genotype 3 respectively. Technivie™ (Ombitasvir/paritaprevir/ritonavir) was approved for use in combination with ribavirin for the treatment of HCV genotype 4 in patients without scarring and cirrhosis and is the first option for HCV-4 infected patients who do not require co-administration with interferon. Daklinza™ was approved for use with Sovaldi® to treat HCV genotype 3 infections. Daklinza™ is the first drug that has demonstrated safety and efficacy in treating HCV genotype 3 without the need for co-administration of interferon or ribavirin.


In October 2015, the U.S. FDA warned that HCV treatments Viekira Pak and Technivie can cause serious liver injury primarily in patients with underlying advanced liver disease, and required that additional information about safety be added to the label.


Other current approved therapies for HCV include interferon alpha-2b or pegylated interferon alpha-2b (Pegintron®), which can be administered with ribavirin (Rebetol®), NS3/4A telaprevir (Incivek®, Vertex and Johnson & Johnson), boceprevir (Victrelis™, Merck), simeprevir (Olysio™, Johnson & Johnson), paritaprevir (AbbVie), Ombitasvir (AbbVie), (NNI) Dasabuvir (ABT-333) and Merck's Zepatier™ (a single-tablet combination of the two drugs grazoprevir and elbasvir).


Additional NS5B polymerase inhibitors are currently under development. Merck is developing the uridine nucleotide prodrug MK-3682 (formerly Idenix IDX21437). The drug is currently in Phase II combination trials.


United States patents and WO applications which describe nucleoside polymerase inhibitors for the treatment of Flaviviridae, including HCV, include those filed by Idenix Pharmaceuticals (U.S. Pat. Nos. 6,812,219; 6,914,054; 7,105,493; 7,138,376; 7,148,206; 7,157,441; 7,163,929; 7,169,766; 7,192,936; 7,365,057; 7,384,924; 7,456,155; 7,547,704; 7,582,618; 7,608,597; 7,608,600; 7,625,875; 7,635,689; 7,662,798; 7,824,851; 7,902,202; 7,932,240; 7,951,789; 8,193,372; 8,299,038; 8,343,937; 8,362,068; 8,507,460; 8,637,475; 8,674,085; 8,680,071; 8,691,788, 8,742,101, 8,951,985; 9,109,001; 9,243,025; US2016/0002281; US2013/0064794; WO/2015/095305; WO/2015/081133; WO/2015/061683; WO/2013/177219; WO/2013/039920; WO/2014/137930; WO/2014/052638; WO/2012/154321); Merck (U.S. Pat. Nos. 6,777,395; 7,105,499; 7,125,855; 7,202,224; 7,323,449; 7,339,054; 7,534,767; 7,632,821; 7,879,815; 8,071,568; 8,148,349; 8,470,834; 8,481,712; 8,541,434; 8,697,694; 8,715,638, 9,061,041; 9,156,872 and WO/2013/009737); Emory University (U.S. Pat. Nos. 6,348,587; 6,911,424; 7,307,065; 7,495,006; 7,662,938; 7,772,208; 8,114,994; 8,168,583; 8,609,627; US 2014/0212382; and WO2014/1244430); Gilead Sciences/Pharmasset Inc. (U.S. Pat. Nos. 7,842,672; 7,973,013; 8,008,264; 8,012,941; 8,012,942; 8,318,682; 8,324,179; 8,415,308; 8,455,451; 8,563,530; 8,841,275; 8,853,171; 8,871,785; 8,877,733; 8,889,159; 8,906,880; 8,912,321; 8,957,045; 8,957,046; 9,045,520; 9,085,573; 9,090,642; and 9,139,604) and (U.S. Pat. Nos. 6,908,924; 6,949,522; 7,094,770; 7,211,570; 7,429,572; 7,601,820; 7,638,502; 7,718,790; 7,772,208; RE42,015; U.S. Pat. Nos. 7,919,247; 7,964,580; 8,093,380; 8,114,997; 8,173,621; 8,334,270; 8,415,322; 8,481,713; 8,492,539; 8,551,973; 8,580,765; 8,618,076; 8,629,263; 8,633,309; 8,642,756; 8,716,262; 8,716,263; 8,735,345; 8,735,372; 8,735,569; 8,759,510 and 8,765,710); Hoffman La-Roche (U.S. Pat. No. 6,660,721), Roche (U.S. Pat. Nos. 6,784,166; 7,608,599, 7,608,601 and 8,071,567); Alios BioPharma Inc. (U.S. Pat. Nos. 8,895,723; 8,877,731; 8,871,737, 8,846,896, 8,772,474; 8,980,865; 9,012,427; US 2015/0105341; US 2015/0011497; US 2010/0249068; US2012/0070411; WO 2015/054465; WO 2014/209979; WO 2014/100505; WO 2014/100498; WO 2013/142159; WO 2013/142157; WO 2013/096680; WO 2013/088155; WO 2010/108135), Enanta Pharmaceuticals (U.S. Pat. Nos. 8,575,119; 8,846,638; 9,085,599; WO 2013/044030; WO 2012/125900), Biota (U.S. Pat. Nos. 7,268,119; 7,285,658; 7,713,941; 8,119,607; 8,415,309; 8,501,699 and 8,802,840), Biocryst Pharmaceuticals (U.S. Pat. Nos. 7,388,002; 7,429,571; 7,514,410; 7,560,434; 7,994,139; 8,133,870; 8,163,703; 8,242,085 and 8,440,813), Alla Chem, LLC (U.S. Pat. No. 8,889,701 and WO 2015/053662), Inhibitex (U.S. Pat. No. 8,759,318 and WO/2012/092484), Janssen Products (U.S. Pat. Nos. 8,399,429; 8,431,588, 8,481,510, 8,552,021, 8,933,052; 9,006,29 and 9,012,428) the University of Georgia Foundation (U.S. Pat. Nos. 6,348,587; 7,307,065; 7,662,938; 8,168,583; 8,673,926, 8,816,074; 8,921,384 and 8,946,244), RFS Pharma, LLC (U.S. Pat. Nos. 8,895,531; 8,859,595; 8,815,829; 8,609,627; 7,560,550; US 2014/0066395; US 2014/0235566; US 2010/0279969; WO/2010/091386 and WO 2012/158811) University College Cardiff Consultants Limited (WO/2014/076490, WO 2010/081082; WO/2008/062206), Achillion Pharmaceuticals, Inc. (WO/2014/169278 and WO 2014/169280), Cocrystal Pharma, Inc. (U.S. Pat. No. 9,173,893), Katholieke Universiteit Leuven (WO 2015/158913), Catabasis (WO 2013/090420) and the Regents of the University of Minnesota (WO 2006/004637).


Nonetheless, there remains a strong medical need to develop anti-HCV therapies that are safe, effective and well-tolerated. The need is accentuated by the expectation that drug resistance. More potent direct-acting antivirals could significantly shorten treatment duration and improve compliance and SVR rates for patients infected with all HCV genotypes.


It is therefore an object of the present invention to provide compounds, pharmaceutical compositions, and methods and uses to treat and/or prevent infections of HCV.


SUMMARY OF THE INVENTION

It has been discovered that the compounds of Formula I, Formula II, Formula III, Formula IV, Formula V, Formula VI, Formula VII and including β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-N6-(mono- or di-methyl) purine nucleotides, are highly active against the HCV virus when administered in an effective amount to a host in need thereof. The host can be a human or any animal that carries the viral infection.


Disclosed nucleotides include those with nanomolar activity against HCV in vitro and therapeutic indices that range to 25,000 or more.


Surprisingly, the parent N6-(methyl) purine nucleosides of disclosed compounds had not been developed or specifically disclosed as drug candidates prior to this invention. For example, it was reported in 2010 that 3′-azido-N6-dimethyl-2,6-diaminopurine is not substantially deaminated by adenosine deaminase over a long period (120 minutes), and for that reason it had been considered an inappropriate compound to derivatize as a drug (see for example, WO 2010/091386, page 86 and corresponding U.S. Pat. No. 8,609,627).


However, it has now been discovered that compounds of the present invention are anabolized to a 5-monophosphate of the N6-substituted-purine without substantial N6-deamination and then subsequently anabolized at the 6-position to generate active guanine triphosphate compounds, in a manner that provides exceptional activity and therapeutic index.


In particular, it has been discovered that a 5′-stabilized phosphate prodrug or derivative of β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleotide, as well as β-2′-deoxy-2′-α-fluoro-2′-β-methyl-N-dimethyl-2,6-diaminopurine nucleotide, and other β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides as described below, are highly active against HCV. This is surprising because the activity of the parent nucleoside β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine in a replicon assay (EC50=15.7 micromolar) indicates that it is not suitable for use as a human drug due to insufficient activity (in combination with the reference WO 2010/091386, page 86 and corresponding U.S. Pat. No. 8,609,627 that suggests that N6-methyl-2,6-diaminopurines are not deaminated in vivo) however, the stabilized racemic phosphate prodrug (phosphoramidate) exhibits an EC50=26 nanomolar (nM), in a replicon assay, which is at least an 600 fold increase in activity. The corresponding (S)-phosphoramidate exhibits an EC50=4 nM, which is at least a 3,900 fold increase in activity; see the structure below and compound 5-2 in Table 7. With a TC50 greater than one hundred micromolar, the compound thus has a therapeutic index of greater than 25,000. For comparison, Sofosbuvir has an EC50=53 nM, a TC50 greater than one hundred micromolar and a therapeutic index greater than 1,920.




embedded image


Likewise, the activity of the parent nucleoside β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diaminopurine in a replicon assay (EC50=10.7 micromolar, “M”) indicates that it is also not suitable for use as a human drug due to insufficient activity, however, the stabilized racemic phosphate prodrug (phosphoramidate) exhibits an EC50=12 nM, in a replicon assay, which is more than a 890 fold increase in activity. The corresponding (S)-phosphoramidate (compound 25, Table 7) also exhibits an EC50=4 nM, which is at least a 2,600 fold increase in activity; see the structure below. In addition, compound 25 also has a therapeutic index of greater than 25,000.




embedded image


In another example, the compound isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-N-cyclopropyl-amino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate exhibited an EC50=7 nM and the corresponding (S)-phosphoramidate exhibited an EC50=5 nM in a replicon assay; see compound 27 in Table 7 and the structure below.




embedded image


As stated above, the metabolism of the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleoside as a phosphoramidate involves the production of a 5′-monophosphate and the subsequent anabolism of the N6-methyl-2,6-diaminopurine base to generate the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine nucleoside as the 5′-monophosphate. The monophosphate is then further anabolized to the active species; the 5′-triphosphate. The β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine triphosphate has an IC50=0.15 μM against the HCV genotype 1b NS5B polymerase.


Thus, in one embodiment, the invention is:




embedded image



wherein:

    • Y is NR1R2
    • R1 is C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CH2F, CHF2, CF3, CH2CF3, CF2CH3 and CF2CF3), C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl), —OR25, —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28 each of which can be optionally substituted;
    • R2 is hydrogen, C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CHF2, CF3, CH2CF3 and CF2CF3), —(C0-C2alkyl)(C3-C6cycloalkyl), —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(heteroaryl); and


      wherein at least one of R1 and R2 is methyl, CH2F, CHF2 or CF3;
    • R3 is hydrogen,




embedded image



diphosphate, triphosphate, an optionally substituted carbonyl linked amino acid, or —C(O)R3C;


R3A can be selected from O, OH, an —O-optionally substituted aryl, an —O-optionally substituted heteroaryl, or an optionally substituted heterocyclyl;

    • R3B can be selected from O, OH, an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester;


      R3C is alkyl, alkenyl, alkynyl, —(C0-C2)(cycloalkyl), —(C0-C2)(heterocyclo), —(C0-C2)(aryl), —(C0-C2)(heteroaryl), —O-alkyl, —O-alkenyl, —O-alkynyl, —O—(C0-C2)(cycloalkyl), —O—(C0-C2)(heterocyclo), —O—(C0-C2)(aryl), or —O—(C0-C2)(heteroaryl), each of which can be optionally substituted;
    • R4 is a monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug, including but not limited to a phosphoramidate, a thiophosphoramidate, or any other moiety that is metabolized to a monophosphate, diphosphate or triphosphate in vivo in the host human or animal; or
    • R3 and R4 together with the oxygens that they are bonded to can form a 3′,5′-cyclic prodrug, including but not limited to, a 3′,5′-cyclic phosphate prodrug;
    • R12 is CH3, CH2F, CHF2, CF3, or ethynyl.


In one embodiment, the invention is:




embedded image



wherein:

    • Y is NR1R2;
    • R1 is C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CH2F, CHF2, CF3, CH2CF3, CF2CH3 and CF2CF3), C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl), —OR25, —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28 each of which can be optionally substituted;
    • R2 is hydrogen, optionally substituted C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CHF2, CF3, CH2CF3 and CF2CF3), optionally substituted —(C0-C2alkyl)(C3-C6cycloalkyl), optionally substituted —(C0-C2alkyl)(heterocycle), optionally substituted —(C0-C2alkyl)(aryl), optionally substituted —(C0-C2alkyl)(heteroaryl), —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28; and wherein at least one of R1 and R2 is methyl, CH2F, CHF2 or CF3;
    • R3 is hydrogen,




embedded image



diphosphate, triphosphate, an optionally substituted carbonyl linked amino acid, or —C(O)R3C;

    • R3A can be selected from O, OH, an —O-optionally substituted aryl, an —O-optionally substituted heteroaryl, or an optionally substituted heterocyclyl;
    • R3B can be selected from O, OH, an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester;
    • R3C is alkyl, alkenyl, alkynyl, —(C0-C2)(cycloalkyl), —(C0-C2)(heterocyclo), —(C0-C2)(aryl), —(C0-C2)(heteroaryl), —O-alkyl, —O-alkenyl, —O-alkynyl, —O—(C0-C2)(cycloalkyl), —O—(C0-C2)(heterocyclo), —O—(C0-C2)(aryl), —O—(C0-C2)(heteroaryl), —S-alkyl, —S-alkenyl, —S-alkynyl, —S—(C0-C2)(cycloalkyl), —S—(C0-C2)(heterocycle), —S—(C0-C2)(aryl), or —S—(C0-C2)(heteroaryl) each of which can be optionally substituted;
    • R3D is alkyl, alkenyl, alkynyl, —(C0-C2)(cycloalkyl), —(C0-C2)(heterocyclo), —(C0-C2)(aryl), —(C0-C2)(heteroaryl), —O-alkyl, —O-alkenyl, —O-alkynyl, —O—(C0-C2)(cycloalkyl), —O—(C0-C2)(heterocyclo), —O—(C0-C2)(aryl), or —O—(C0-C2)(heteroaryl), each of which can be optionally substituted;
    • R4 is a monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug, including but not limited to a phosphoramidate, a thiophosphoramidate, or any other moiety that is metabolized to a monophosphate, diphosphate or triphosphate in vivo in the host human or animal; or
    • R3 and R4 together with the oxygens that they are bonded to can form a 3′,5′-cyclic prodrug, including but not limited to, a 3′,5′-cyclic phosphate prodrug;
    • R5 is C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CHF2, CF3, CH2CF3 and CF2CF3), C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl), —OR25, —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28 each of which can be optionally substituted;
    • R6 is hydrogen, optionally substituted C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CHF2, CF3, CH2CF3 and CF2CF3), optionally substituted —(C0-C2alkyl)(C3-C6cycloalkyl), optionally substituted —(C0-C2alkyl)(heterocycle), optionally substituted —(C0-C2alkyl)(aryl), optionally substituted —(C0-C2alkyl)(heteroaryl), —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28; or
    • R5 and R6 together with the nitrogen that they are bonded to can form a heterocyclic ring;
    • R12 is CH3, CH2F, CHF2, CF3, or ethynyl;
    • R22 is Cl, Br, F, CN, N3, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C1-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl); —ONHC(═O)OR23, —NHOR24, —OR25, —SR25, —NH(CH2)1-4N(R26)2, —NHNHR26, —N═NR27, —NHC(O)NHNHR27, —NHC(S)NHNHR27, —C(O)NHNHR27, —NR27SO2R28, —SO2NR27R29, —C(O)NR27R29, —CO2R29, —SO2R29,




embedded image



—P(O)H(OR29), —P(O)(OR29)(OR30), —P(O)(OR29)(NR29R30) or —NR5R6;

    • for example including but not limited to the following embodiments, chloro, bromo, fluoro, cyano, azido, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and n-pentyl, 1,1-dimethylpropyl, 2,2-dimtheylpropyl, 3-methylbutyl, 1-methylbutyl, 1-ethylpropyl, vinyl, allyl, 1-butynyl, 2-butynyl, acetylenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —(CH2)-cyclopropyl, —(CH2)-cyclobutyl, —(CH2)-cyclopentyl, —(CH2)-cyclohexyl, aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, tetrahydrofuran, thiolane, pyrazolidine, piperidine, oxane, thiane, —(CH2)-aziridine, —(CH2)-oxirane, —(CH2)-thiirane, —(CH2)-azetidine, —(CH2)-oxetane, —(CH2)-thietane, —(CH2)-pyrrolidine, —(CH2)-tetrahydrofuran, —(CH2)-thiolane, —(CH2)-pyrazolidine, —(CH2)-piperidine, —(CH2)-oxane, —(CH2)-thiane, phenyl, pyridyl, —ONHC(═O)OCH3, —ONHC(═O)OCH2CH3, —NHOH, NHOCH3, —OCH3, OC2H5, —OPh, OCH2Ph, —SCH3, —SC2H5, —SPh, SCH2Ph, —NH(CH2)2NH2, —NH(CH2)2N(CH3)2, —NHNH2, —NHNHCH3, —N═NH, —N═NCH3, —N═NCH2CH3, —NHC(O)NHNH2, —NHC(S)NHNH2, —C(O)NHNH2, —NHSO2CH3, —NHSO2CH2CH3, —SO2NHCH3, —SO2N(CH3)2, —C(O)NH2, —C(O)NHCH3, —C(O)N(CH3)2, —CO2CH3, —CO2CH2CH3, —CO2Ph, —CO2CH2Ph, —SO2CH3, —SO2CH2CH3, —SO2Ph, —SO2CH2Ph,




embedded image



—P(O)H(OH), —P(O)H(OCH3), —P(O)(OH)(OH), —P(O)(OH)(OCH3), —P(O)(OCH3)(OCH3), —P(O)(OH)(NH2), —P(O)(OH)(NHCH3), —P(O)(OH)N(CH3)2, —NHC(O)CH3, —NHC(O)CH2CH3, —NHC(O)CH(CH3)2, —NHC(O)OCH3, —NHC(O)OCH2CH3, —NHC(O)OCH(CH3)2, —NHC(O)OCH2CH2CH3, —NHC(O)OCH2CH2CH2CH3 and —NHC(O)OCH2CH2CH2CH2CH3;

    • R23 is C1-C5alkyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle)-(C0-2 alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) each of which can be optionally substituted;
    • R24 is hydrogen, C1-C6 alkyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C1-C2alkyl)(C3-C6heterocycle) —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted;
    • R25 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted;
    • R26 is independently selected from hydrogen, C1-C6alkyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted;
    • R27 hydrogen or optionally substituted C1-C6 alkyl;
    • R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) each of which can be optionally substituted;
    • R29 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted; or
    • R27 and R29 together with the nitrogen that they are bonded to can form a heterocyclic ring;
    • R30 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted; or
    • R29 and R30 can be bonded together to form a heterocyclic ring;
    • x is 1, 2 or 3.


The metabolism of the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diaminopurine nucleotide involves both the formation of the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diaminopurine nucleoside triphosphate as well as the generation of the corresponding guanine nucleoside triphosphate. See Scheme 2 and 3.


2′-Deoxy-2′-α-fluoro-2′-β-C-substituted-N6-substituted-2,6-diaminopurine nucleotides can be further substituted at the N2-position by alkylation or acylated which can modify the lipophilicity, pharmacokinetics and/or targeting of the nucleotide to the liver. It has been discovered that 2′-deoxy-2′-α-fluoro-2′-β-C-substituted-N6-substituted-2,6-diaminopurine nucleotides modified at the 2-position of the diaminopurine can be dealkylated or deacylated by hepatic enzymes to further increase the specificity of the nucleotide derivatives both in vitro and in vivo, unless the N2-amino group is completely replaced by a different moiety, as described herein, such as fluoro. For example, the nucleoside phosphoramidate 2′-deoxy-2′-α-fluoro-2′-β-methyl-N2-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate is dealkylated to 2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate when incubated with a human liver S9 fraction in vitro, up to 60 minutes, these conditions mimics in vivo conditions. In one embodiment, N2 modifications will increase cell permeability and hepatitic targeting.


Despite the volume of antiviral nucleoside literature and patent filings, the 5′-stabilized phosphate derivative of 2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleoside, 2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diaminopurine nucleoside, and other β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleoside derivatives as described herein have not been specifically disclosed, nor have their advantageous activities been described.


Unless otherwise specified, the compounds described herein are provided in the β-D-configuration. Likewise, when in phosphoramide or thiophosphoramidate form, the amino acid portion can be in the L- or D-configuration. In an alternative embodiment, the compounds can be provided in a β-L-configuration. Likewise, any substituent group that exhibits chirality can be provided in racemic, enantiomeric, diastereomeric form or any mixture thereof. Where a phosphoramidate, thiophosphoramidate or other stabilized phosphorus prodrug in which the phosphorus exhibits chirality is used as the R4 stabilized phosphate prodrug, it can be provided as an R or S chiral phosphorus derivative or a mixture thereof, including a racemic mixture. All of the combinations of these stereoconfigurations are included in the invention described herein.


Accordingly, the present invention includes a compound of Formula I-VII, or a pharmaceutically acceptable composition, salt, or prodrug thereof, as described herein:




embedded image


In one specific embodiment, the parent nucleoside, i.e., the nucleoside wherein R4 is hydrogen and the 5′-position thus has a hydroxyl group, is not substantially deaminated by adenosine deaminase under conditions that mimic the in vivo environment (e.g., ambient temperature and aqueous physiological pH), for a period of 7 minutes, 10 minutes, 30 minutes, 60 minutes or 120 minutes. Unless otherwise stated, the time period is 30 minutes. In this embodiment, the term “not substantially deaminated” means that the parent compound is not converted to the corresponding guanine derivative, or 6-oxo derivative, in an amount sufficient to provide a therapeutic effect in vivo.


Compounds, methods, and compositions are provided for the treatment of a host infected with a HCV virus via administration of an effective amount of the compound or its pharmaceutically acceptable salt.


The compounds and compositions can also be used to treat related conditions such as anti-HCV antibody positive and antigen positive conditions, viral-based chronic liver inflammation, liver cancer resulting from advanced hepatitis C, cirrhosis, chronic or acute hepatitis C, fulminant hepatitis C, chronic persistent hepatitis C and anti-HCV-based fatigue.


The compound or formulations that include the compounds can also be used prophylactically to prevent or restrict the progression of clinical illness in individuals who are anti-HCV antibody or antigen positive or who have been exposed to hepatitis C.


In another embodiment, compounds of Formula Ia are disclosed:




embedded image




    • wherein.


      Y, R3 and R4 are as defined above.





In one embodiment of Formula Ia, R3 is hydrogen.


In one embodiment of Formula Ia, when Y is NR1R2, R1 is methyl and R2 is hydrogen.


In one embodiment of Formula Ia, when Y is NR1R2, both R1 and R2 are methyl.


In one embodiment of Formula Ia, when Y is NR1R2, R1 is methyl and R2 is cyclopropyl.


In another embodiment, compounds of Formula Ib are disclosed:




embedded image




    • wherein:

    • Y, R3 and R4 are as defined above.





In one embodiment of Formula Ib, R3 is hydrogen.


In one embodiment of Formula Ib, when Y is NR1R2, R1 is methyl and R2 is hydrogen.


In one embodiment of Formula Ib, when Y is NR1R2, both R1 and R2 are methyl.


In one embodiment, compounds of Formula II are disclosed:




embedded image




    • wherein:

    • Y, R3, R4, R12 and R22 are as defined above.





In another embodiment, compounds of Formula IIa are disclosed:




embedded image




    • wherein:

    • Y, R3, R4 and R22 are as defined above.





In another embodiment, compounds of Formula IIb are disclosed:




embedded image




    • wherein:

    • Y, R3, R4, and R22 are as defined above.





In one embodiment, compounds of Formula III are disclosed:




embedded image




    • wherein the variables Y, R3, R7, R8, R9a, R9b, R10, R12 and R22 are described herein.





In one embodiment, compounds of Formula IV are disclosed:




embedded image




    • wherein the variables Y, R3, R7, R8, R9a, R9b, R10 and R22 are described herein.





In one embodiment, compounds of Formula V are disclosed:




embedded image




    • wherein the variables Y, R3, R7, R8, R9a, R9b, R10 and R22 are described herein. In one embodiment, compounds of Formula VI are disclosed:







embedded image




    • wherein:

    • R41 is halogen (in particular F or Cl), OR3, N3, NH2 or CN; and

    • the variables Y, R3, R4, and R12 are described herein.





In one embodiment, compounds of Formula VII are disclosed:




embedded image


Wherein the variables Y, R3, R4, R12 and R41 are described herein.


The phosphorus in any of the Formulas above may be chiral and thus can be provided as an R or S enantiomer or mixture thereof, including a racemic mixture.


Compound 5 was separated into the enantiomer compounds 5-1 and 5-2. Compound 5-2 was also prepared by chiral synthesis and assigned compound 24.


In one embodiment, compounds, methods, and compositions are provided for the treatment of a host infected with or exposed to hepatitis C described herein. The compounds of the invention can be administered in an effective amount alone or in combination with another anti-HCV drug, to treat the infected host. In certain embodiments, it is useful to administer a combination of drugs that modulates the same or a different pathway or inhibits a different target in the virus. As the disclosed β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides are NS5B polymerase inhibitors, it may be useful to administer the compound to a host in combination with a protease inhibitor, such as an NS3/4A protease inhibitor (for example, telaprevir (Incivek®) boceprevir (Victrelis™) simeprevir (Olysio™), or paritaprevir, or an NS5A inhibitor (for example, Ombitasvir). The compounds of the invention can also be administered in combination with a structurally different NS5B polymerase inhibitor such as another compound described herein or below, including Gilead's Sovaldi®. The compounds of the invention can also be administered in combination with interferon alfa-2a, which may be pegylated or otherwise modified, and/or ribavirin.


The β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides of the invention are typically administered orally, for example in pill or tablet form, but may be administered via an other route which the attending physician considers appropriate, including via intravenous, transdermal, subcutaneous, topical, parenteral, or other suitable route.





BRIEF DESCRIPTION OF THE FIGURES


FIG. 1 is a sample chromatogram of a semi-prep run illustrating the separation of the stereoisomers of Compound 5 using a Phenominex Luna column as disclosed in Example 9. The y axis is shown in mAU and the x axis is measured in minutes.



FIG. 2 is a graph of the HCV replication inhibition curves for Compound 5-2 (Table 7) and Sofosbuvir. Compound 5-2 has an EC50=4 nM, a TC50 greater than one hundred micromolar and a therapeutic index of greater than 25,000. Sofosbuvir has an EC50=53 nM, a TC50 greater than one hundred micromolar and a therapeutic index greater than 1,920. The y-axis is the percent of virus control and the x-axis is the concentration of drug in M.



FIG. 3 is a graph of the HCV replication inhibition curves for Compound 25 (Table 7) and Sofosbuvir. As described in Example 27, Compound 25 has an EC50=4 nM, a TC50 of greater than 100 μM, and a therapeutic index of greater than 25,000. Sofosbuvir has an EC50=53 nM, a TC50 greater than one hundred micromolar and a therapeutic index greater than 1,920. The y-axis is the percent of virus control and the x-axis is the concentration of drug in μM.



FIG. 4 is an intra-assay comparison of the anti-HCV activity for Compounds 5-2, 25, 27 (Table 7) and Sofosbuvir. The y-axis is the percent of virus control and the x-axis is the concentration of drug in M. See, Example 27.



FIG. 5 is a graph that shows the stability of compounds 5-2; the N2-acetate of compound 5-2, the N2-butyrate of compound 5-2; the N2-methyl derivative of compound 5-2; and the N2-n-pentylcarbamate of compound 5-2 in human blood. The x axis is incubation time measured in minutes and the y axis is the measurement of the percent of the parent compound remaining.



FIG. 6 is a graph showing the in vitro time course dealkylation of 2′-deoxy-2′-α-fluoro-2′-β-methyl-N2-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate to 2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate in the presence of a human liver S9 fraction. The x axis is measured in minutes and the y axis is the measurement of the concentration of the compound remaining in nM.



FIG. 7 is a graph showing the stability of compounds 5-2; the N2-acetate of compound 5-2, the N2-butyrate of compound 5-2; the N2-methyl derivative of compound 5-2; and the N2-n-pentylcarbamate of compound 5-2 in the presence of a human liver S9 fraction. The x axis is measured in minutes and the y axis is the measurement of percent compound remaining.



FIG. 8 shows the predominant Compound 25 metabolites generated in human hepatocytes. The x axis is incubation time in hours. The y axis is intracellular concentration in μmol/106 cells. See Example 33.



FIG. 9 shows the predominant Compound 27 metabolites generated in human hepatocytes. The x axis is incubation time in hours. The y axis is intracellular concentration in μmol/106 cells. See Example 33.



FIG. 10 shows the predominant Compound 5-2 metabolites generated in human hepatocytes. The x axis is incubation time in hours. The y axis is intracellular concentration in μmol/106 cells. See Example 33.



FIG. 11 is a graph showing the activation pathways for Compounds 25, 27 and 5-2. As can be seen, Compounds 25, 27 and 5-2 are converted to their corresponding monophosphate analogs which are subsequently metabolized to a common MP analog; β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine monophosphate. The monophosphate is then stepwise phosphorylated to the active triphosphate: β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine triphosphate. See Example 33.





DETAILED DESCRIPTION OF THE INVENTION

The invention disclosed herein is a compound, method, and composition for the treatment of infections in or exposure to humans and other host animals of the HCV virus that includes the administration of an effective amount of a compound of Formula I-VII as described herein or a pharmaceutically acceptable salt or prodrug thereof, optionally in a pharmaceutically acceptable carrier. The compounds of this invention either possess antiviral activity, or are metabolized to a compound that exhibits such activity.


The compounds and compositions can also be used to treat conditions related to or occurring as a result of a HCV viral exposure. For example, the active compound can be used to treat HCV antibody positive and HCV antigen positive conditions, viral-based chronic liver inflammation, liver cancer resulting from advanced hepatitis C, cirrhosis, acute hepatitis C, fulminant hepatitis C, chronic persistent hepatitis C, and anti-HCV-based fatigue. In one embodiment, the compounds or formulations that include the compounds can also be used prophylactically to prevent or retard the progression of clinical illness in individuals who are HCV antibody or HCV antigen positive or who have been exposed to hepatitis C.


In particular, it has been discovered that a 5′-stabilized phosphate prodrug or derivative of β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diamino purine nucleotide, as well as β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diamino purine nucleotide, and other β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides as described below, are highly active against HCV. This is surprising because the activity of the parent nucleoside β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diamino purine in a replicon assay (EC50=15.7 micromolar) indicates that it is not suitable for use as a human drug due to insufficient activity, however, the stabilized phosphate prodrug (phosphoramidate) exhibits an EC50=26 nanomolar, in a replicon assay, which is at least an 870 fold increase in activity. Likewise, the activity of the parent nucleoside β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diaminopurine in a replicon assay (EC50=10.7 micromolar, “M”) indicates that it is also not suitable for use as a human drug due to insufficient activity, however, the stabilized phosphate prodrug (phosphoramidate) exhibits an EC50=12 nanomolar, (“nM”), in a replicon assay, which is more than a 1,300 fold increase in activity.


Despite the volume of antiviral nucleoside literature and patent filings, the 5′-stabilized phosphate derivative of 2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diamino purine nucleotide, 2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diamino purine nucleotide, and other β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides have not been specifically disclosed.


Unless otherwise specified, the compounds described herein are provided in the β-D-configuration. In an alternative embodiment, the compounds can be provided in a β-L-configuration. Likewise, any substituent group that exhibits chirality can be provided in racemic, enantiomeric, diastereomeric form or any mixture thereof. Where a phosphoramidate, thiophosphoramidate or other stabilized phosphorus prodrug in which the phosphorus exhibits chirality is used as the R4 stabilized phosphate prodrug, it can be provided as an R or S chiral phosphorus derivative or a mixture thereof, including a racemic mixture. The amino acid of the phosphoramidate or thiophosphoramidate can be in the D- or L-configuration, or a mixture thereof, including a racemic mixture. All of the combinations of these stereo configurations are included in the invention described herein.


The present invention includes the following features:

    • (a) a compound of Formula I-VII as described herein, and pharmaceutically acceptable salts and prodrugs thereof,
    • (b) Formulas I-VII as described herein, and pharmaceutically acceptable salts and prodrugs thereof for use in the treatment or prophylaxis of a hepatitis C virus infection;
    • (c) use of Formulas I-VII, and pharmaceutically acceptable salts and prodrugs thereof in the manufacture of a medicament for treatment of a hepatitis C virus infection;
    • (d) a method for manufacturing a medicament intended for the therapeutic use for treating a hepatitis C virus infection, characterized in that a Formulas I-VII as described herein is used in the manufacture;
    • (e) a pharmaceutical formulation comprising an effective host-treating amount of the Formulas I-VII or a pharmaceutically acceptable salt or prodrug thereof together with a pharmaceutically acceptable carrier or diluent;
    • (f) Formulas I-VII as described herein substantially in the absence of stereoisomers of the described compound, or substantially isolated from other chemical entities; and,
    • (g) processes for the preparation of therapeutic products that contain an effective amount of a Formulas I-VII, as described herein.


I. 2′-Deoxy-2′-α-Fluoro-2′-β-C-Substituted-2-Modified-N6-Substituted Purine Nucleotides of the Invention

The active compounds of the invention are those depicted, for example, in Formula I, which can be provided in a pharmaceutically acceptable composition, salt or prodrug thereof:




embedded image




    • wherein:

    • Y is NR1R2;

    • R1 is C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CH2F, CH2F, CF3, CH2CF3, CF2CH3 and CF2CF3), C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl), —OR25, —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28 each of which can be optionally substituted;

    • R2 is hydrogen, optionally substituted C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CH2F, CF3, CH2CF3 and CF2CF3), optionally substituted —(C0-C2alkyl)(C3-C6cycloalkyl), optionally substituted —(C0-C2alkyl)(heterocycle), optionally substituted —(C0-C2alkyl)(aryl), optionally substituted —(C0-C2alkyl)(heteroaryl), —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28; and


      wherein at least one of R1 and R2 is methyl, CH2F, CHF2 or CF3;

    • R3 is hydrogen,







embedded image




    • diphosphate, triphosphate, an optionally substituted carbonyl linked amino acid, or —C(O)R3C;

    • R3A can be selected from O, OH, an —O-optionally substituted aryl, an —O-optionally substituted heteroaryl, or an optionally substituted heterocyclyl;

    • R3B can be selected from O, OH, an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester;

    • R3C is alkyl, alkenyl, alkynyl, —(C0-C2)(cycloalkyl), —(C0-C2)(heterocyclo), —(C0-C2)(aryl), —(C0-C2)(heteroaryl), —O-alkyl, —O-alkenyl, —O-alkynyl, —O—(C0-C2)(cycloalkyl), —O—(C0-C2)(heterocyclo), —O—(C0-C2)(aryl), or —O—(C0-C2)(heteroaryl), each of which can be optionally substituted;

    • R4 is a monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug, including but not limited to a phosphoramidate, a thiophosphoramidate, or any other moiety that is metabolized to a monophosphate, diphosphate or triphosphate in vivo in the host human or animal; or

    • R3 and R4 together with the oxygens that they are bonded to can form a 3′,5′-cyclic prodrug, including but not limited to, a 3′,5′-cyclic phosphate prodrug;

    • R12 is CH3, CH2F, CHF2, CF3, or ethynyl.





A stabilized phosphate prodrug is any moiety that can deliver a mono, di, or triphosphate.


In another embodiment, compounds of Formula Ia are disclosed:




embedded image




    • wherein:

    • Y, R3 and R4 are as defined above.





In another embodiment, compounds of Formula Ib are disclosed:




embedded image




    • wherein:

    • Y, R3 and R4 are as defined above.





In another embodiment, the compound is according to Formula Ic:




embedded image




    • wherein:

    • R7 is hydrogen, C1-6alkyl; C3-7cycloalkyl; heteroaryl, heterocyclic, or aryl, which includes, but is not limited to, phenyl or naphthyl, where phenyl or naphthyl are optionally substituted with C1-6alkyl, C2-6alkenyl, C2-6 alkynyl, C1-6alkoxy, F, Cl, Br, I, nitro, cyano, C1-6haloalkyl, —N(R7′)2, C1-6acylamino, NHSO2C1-6alkyl, —SO2N(R7′)2, COR7″, and —SO2C1-6alkyl; (R7′ is independently hydrogen or C1-6alkyl; R7″ is —OR11 or —N(R7)2);

    • R8 is hydrogen, C1-6alkyl, or R9a or R9b and R8 together are (CH2)n so as to form a cyclic ring that includes the adjoining N and C atoms; where n is 2 to 4;

    • R9a and R9b are (i) independently selected from hydrogen, C1-6alkyl, cycloalkyl, —(CH2)c(NR9′)2, C1-6hydroxyalkyl, —CH2SH, —(CH2)2S(O)(Me, —(CH2)3NHC(═NH)NH2, (1H-indol-3-yl)methyl, (1H-imidazol-4-yl)methyl, —(CH2)cCOR9″, aryl and aryl(C1-3alkyl)-, the aryl groups can be optionally substituted with a group selected from hydroxyl, C1-6alkyl, C1-6alkoxy, halogen, nitro and cyano; (ii) R9a and R9b both are C1-6alkyl; (iii) R9a and R9b together are (CH2)r so as to form a spiro ring; (iv) R9a is hydrogen and R9b and R8 together are (CH2)n so as to form a cyclic ring that includes the adjoining N and C atoms (v) R9b is hydrogen and R9a and R8 together are (CH2)n so as to form a cyclic ring that includes the adjoining N and C atoms, where c is 1 to 6, n is 2 to 4, r is 2 to 5 and where R9′ is independently hydrogen or C1-6 alkyl and R9″ is —OR11 or —N(R11′)2); (vi) R9a is hydrogen and R9b is hydrogen, CH3, CH2CH3, CH(CH3)2, CH2CH(CH3)2, CH(CH3)CH2CH3, CH2Ph, CH2-indol-3-yl, —CH2CH2SCH3, CH2CO2H, CH2C(O)NH2, CH2CH2COOH, CH2CH2C(O)NH2, CH2CH2CH2CH2NH2, —CH2CH2CH2NHC(NH)NH2, CH2-imidazol-4-yl, CH2OH, CH(OH)CH3, CH2((4′-OH)-Ph), CH2SH, or lower cycloalkyl; or (vii) R9a is CH3, CH2CH3, CH(CH3)2, CH2CH(CH3)2, CH(CH3)CH2CH3, CH2Ph, CH2-indol-3-yl, —CH2CH2SCH3, CH2CO2H, CH2C(O)NH2, CH2CH2COOH, CH2CH2C(O)NH2, CH2CH2CH2CH2NH2, —CH2CH2CH2NHC(NH)NH2, CH2-imidazol-4-yl, CH2OH, CH(OH)CH3, CH2((4′-OH)-Ph), CH2SH, or lower cycloalkyl and R9b is hydrogen;

    • R10 is hydrogen, C1-6alkyl optionally substituted with an alkoxy, di(lower alkyl)-amino, or halogen, C1-6haloalkyl, C3-7cycloalkyl, heterocycloalkyl, aminoacyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl;

    • R11 is an optionally substituted C1-6alkyl, an optionally substituted cycloalkyl; an optionally substituted C2-6alkynyl, an optionally substituted C2-6alkenyl, or optionally substituted acyl, which includes but is not limited to C(O)(C1-6 alkyl); and

    • Y, R3 and R12 are as defined herein.





In one embodiment, compounds of Formula II are disclosed:




embedded image




    • wherein:

    • Y is NR1R2;

    • R1 is C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CH2F, CHF2, CF3, CH2CF3, CF2CH3 and CF2CF3), C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl), —OR25, —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28 each of which can be optionally substituted;

    • R2 is hydrogen, optionally substituted C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CHF2, CF3, CH2CF3 and CF2CF3), optionally substituted —(C0-C2alkyl)(C3-C6cycloalkyl), optionally substituted —(C0-C2alkyl)(heterocycle), optionally substituted —(C0-C2alkyl)(aryl), optionally substituted —(C0-C2alkyl)(heteroaryl), —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28; and wherein at least one of R1 and R2 is methyl, CH2F, CHF2 or CF3;

    • R3 is hydrogen,







embedded image



diphosphate, triphosphate, an optionally substituted carbonyl linked amino acid, or —C(O)R3C;

    • R3A can be selected from O, OH, an —O-optionally substituted aryl, an —O-optionally substituted heteroaryl, or an optionally substituted heterocyclyl;
    • R3B can be selected from O, OH, an optionally substituted N-linked amino acid or an optionally substituted N-linked amino acid ester;
    • R3C is alkyl, alkenyl, alkynyl, —(C0-C2)(cycloalkyl), —(C0-C2)(heterocyclo), —(C0-C2)(aryl), —(C0-C2)(heteroaryl), —O-alkyl, —O-alkenyl, —O-alkynyl, —O—(C0-C2)(cycloalkyl), —O—(C0-C2)(heterocyclo), —O—(C0-C2)(aryl), —O—(C0-C2)(heteroaryl), —S-alkyl, —S-alkenyl, —S-alkynyl, —S—(C0-C2)(cycloalkyl), —S—(C0-C2)(heterocycle), —S—(C0-C2)(aryl), or —S—(C0-C2)(heteroaryl) each of which can be optionally substituted;
    • R3D is alkyl, alkenyl, alkynyl, —(C0-C2)(cycloalkyl), —(C0-C2)(heterocyclo), —(C0-C2)(aryl), —(C0-C2)(heteroaryl), —O-alkyl, —O-alkenyl, —O-alkynyl, —O—(C0-C2)(cycloalkyl), —O—(C0-C2)(heterocyclo), —O—(C0-C2)(aryl), or —O—(C0-C2)(heteroaryl), each of which can be optionally substituted;
    • R4 is a monophosphate, diphosphate, triphosphate, or a stabilized phosphate prodrug, including but not limited to a phosphoramidate, a thiophosphoramidate, or any other moiety that is metabolized to a monophosphate, diphosphate or triphosphate in vivo in the host human or animal; or
    • R3 and R4 together with the oxygens that they are bonded to can form a 3′,5′-cyclic prodrug, including but not limited to, a 3′,5′-cyclic phosphate prodrug;
    • R5 is C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CHF2, CF3, CH2CF3 and CF2CF3), C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl), —OR25, —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H5, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28 each of which can be optionally substituted;
    • R6 is hydrogen, optionally substituted C1-C5alkyl (including methyl, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and pentyl), C1-C5haloalkyl (including CHF2, CH2F, CF3, CH2CF3 and CF2CF3), optionally substituted —(C0-C2alkyl)(C3-C6cycloalkyl), optionally substituted —(C0-C2alkyl)(heterocycle), optionally substituted —(C0-C2alkyl)(aryl), optionally substituted —(C0-C2alkyl)(heteroaryl), —C(O)R3C (including —C(O)CH3, —C(O)CH2CH3—C(O)CH(CH3)2, —C(O)OCH3, —C(O)OC2H, —C(O)OC3H7, —C(O)OC4H9, and —C(O)OC5H11), —C(S)R3D, or —SO2R28; or
    • R5 and R6 together with the nitrogen that they are bonded to can form a heterocyclic ring;
    • R12 is CH3, CH2F, CHF2, CF3, or ethynyl;
    • R22 is Cl, Br, F, CN, N3, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl), —(C0-C2alkyl)(heteroaryl); —ONHC(═O)OR23, —NHOR24, —OR25, —SR25, —NH(CH2)1-4N(R26)2, —NHNHR26, —N═NR27, —NHC(O)NHNHR27, —NHC(S)NHNHR27, —C(O)NHNHR27, —NR27SO2R28, —SO2NR27R29, —C(O)NR27R29, —CO2R29, —SO2R29,




embedded image



—P(O)H(OR29), —P(O)(OR29)(OR30), —P(O)(OR29)(NR29R30) or —NR5R6;

    • for example including but not limited to the following embodiments, chloro, bromo, fluoro, cyano, azido, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and n-pentyl, 1,1-dimethylpropyl, 2,2-dimtheylpropyl, 3-methylbutyl, 1-methylbutyl, 1-ethylpropyl, vinyl, allyl, 1-butynyl, 2-butynyl, acetylenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —(CH2)-cyclopropyl, —(CH2)-cyclobutyl, —(CH2)-cyclopentyl, —(CH2)-cyclohexyl, aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, tetrahydrofuran, thiolane, pyrazolidine, piperidine, oxane, thiane, —(CH2)-aziridine, —(CH2)-oxirane, —(CH2)-thiirane, —(CH2)-azetidine, —(CH2)-oxetane, —(CH2)-thietane, —(CH2)-pyrrolidine, —(CH2)-tetrahydrofuran, —(CH2)-thiolane, —(CH2)-pyrazolidine, —(CH2)-piperidine, —(CH2)-oxane, —(CH2)-thiane, phenyl, pyridyl, —ONHC(═O)OCH3, —ONHC(═O)OCH2CH3, —NHOH, NHOCH3, —OCH3, OC2H5, —OPh, OCH2Ph, —SCH3, —SC2H5, —SPh, SCH2Ph, —NH(CH2)2NH2, —NH(CH2)2N(CH3)2, —NHNH2, —NHNHCH3, —N═NH, —N═NCH3, —N═NCH2CH3, —NHC(O)NHNH2, —NHC(S)NHNH2, —C(O)NHNH2, —NHSO2CH3, —NHSO2CH2CH3, —SO2NHCH3, —SO2N(CH3)2, —C(O)NH2, —C(O)NHCH3, —C(O)N(CH3)2, —CO2CH3, —CO2CH2CH3, —CO2Ph, —CO2CH2Ph, —SO2CH3, —SO2CH2CH3, —SO2Ph, —SO2CH2Ph,




embedded image



—P(O)H(OH), —P(O)H(OCH3), —P(O)(OH)(OH), —P(O)(OH)(OCH3), —P(O)(OCH3)(OCH3), —P(O)(OH)(NH2), —P(O)(OH)(NHCH3), —P(O)(OH)N(CH3)2, —NHC(O)CH3, —NHC(O)CH2CH3, —NHC(O)CH(CH3)2, —NHC(O)OCH3, —NHC(O)OCH2CH3, —NHC(O)OCH(CH3)2, —NHC(O)OCH2CH2CH3, —NHC(O)OCH2CH2CH2CH3 and —NHC(O)OCH2CH2CH2CH2CH3;

    • R23 is C1-C5alkyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle)-(C0-2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) each of which can be optionally substituted;
    • R24 is hydrogen, C1-C6 alkyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C1-C2alkyl)(C3-C6heterocycle) —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted;
    • R25 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted;
    • R26 is independently selected from hydrogen, C1-C6alkyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(heterocycle), —(C0-C2alkyl)(aryl), or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted;
    • R27 hydrogen or optionally substituted C1-C6 alkyl;
    • R28 is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) each of which can be optionally substituted;
    • R29 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3-C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted; or
    • R27 and R29 together with the nitrogen that they are bonded to can form a heterocyclic ring;
    • R30 is hydrogen, C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, —(C0-C2alkyl)(C3C6cycloalkyl), —(C0-C2alkyl)(C3-C6heterocycle), —(C0-C2alkyl)(aryl) or —(C0-C2alkyl)(heteroaryl) wherein except for the hydrogen each of which can be optionally substituted; or
    • R29 and R30 can be bonded together to form a heterocyclic ring;
    • x is 1, 2 or 3.


In another embodiment, compounds of Formula IIa are disclosed:




embedded image




    • wherein:

    • Y, R3, R4 and R22 are as defined above.





In another embodiment, compounds of Formula IIb are disclosed:




embedded image




    • wherein:

    • Y, R3, R4 and R22 are as defined above.





In a typical embodiment, the compound is a β-D isomer with reference to the corresponding nucleoside (i.e., in the naturally occurring configuration). In an alternative configuration, the compound is provided as a β-L isomer. The compound is typically at least 90% free of the opposite enantiomer, and can be at least 98%, 99% or even 100% free of the opposite enantiomer. Unless described otherwise, the compound is at least 90% free of the opposite enantiomer.


In another embodiment, the compound is according to Formula III:




embedded image




    • wherein:

    • R7 is hydrogen, C1-6alkyl; C3-7cycloalkyl; heteroaryl, heterocyclic, or aryl, which includes, but is not limited to, phenyl or naphthyl, where phenyl or naphthyl are optionally substituted with C1-6alkyl, C2-6alkenyl, C2-6 alkynyl, C1-6alkoxy, F, Cl, Br, I, nitro, cyano, C1-6haloalkyl, —N(R7′)2, C1-6acylamino, NHSO2C1-6alkyl, —SO2N(R7′)2, COR7″, and —S2C1-6alkyl; (R7′ is independently hydrogen or C1-6alkyl; R7″ is —OR11 or —N(R7)2);

    • R8 is hydrogen, C1-6alkyl, or R9a or R9b and R8 together are (CH2)n so as to form a cyclic ring that includes the adjoining N and C atoms; where n is 2 to 4;

    • R9a and R9b are (i) independently selected from hydrogen, C1-6alkyl, cycloalkyl, —(CH2)c(NR9′)2, C1-6hydroxyalkyl, —CH2SH, —(CH2)2S(O)(Me, —(CH2)3NHC(═NH)NH2, (1H-indol-3-yl)methyl, (1H-imidazol-4-yl)methyl, —(CH2)cCOR9″, aryl and aryl(C1-3alkyl)-, the aryl groups can be optionally substituted with a group selected from hydroxyl, C1-6alkyl, C1-6alkoxy, halogen, nitro and cyano; (ii) R9a and R9b both are C1-6alkyl; (iii) R9a and R9b together are (CH2)r so as to form a spiro ring; (iv) R9a is hydrogen and R9b and R8 together are (CH2)n so as to form a cyclic ring that includes the adjoining N and C atoms (v) R9b is hydrogen and R9a and R8 together are (CH2)n so as to form a cyclic ring that includes the adjoining N and C atoms, where c is 1 to 6, n is 2 to 4, r is 2 to 5 and where R9′ is independently hydrogen or C1-6 alkyl and R9″ is —OR11 or —N(R11′)2); (vi) R9a is hydrogen and R9b is hydrogen, CH3, CH2CH3, CH(CH3)2, CH2CH(CH3)2, CH(CH3)CH2CH3, CH2Ph, CH2-indol-3-yl, —CH2CH2SCH3, CH2CO2H, CH2C(O)NH2, CH2CH2COOH, CH2CH2C(O)NH2, CH2CH2CH2CH2NH2, —CH2CH2CH2NHC(NH)NH2, CH2-imidazol-4-yl, CH2OH, CH(OH)CH3, CH2((4′-OH)-Ph), CH2SH, or lower cycloalkyl; or (vii) R9a is CH3, CH2CH3, CH(CH3)2, CH2CH(CH3)2, CH(CH3)CH2CH3, CH2Ph, CH2-indol-3-yl, —CH2CH2SCH3, CH2CO2H, CH2C(O)NH2, CH2CH2COOH, CH2CH2C(O)NH2, CH2CH2CH2CH2NH2, —CH2CH2CH2NHC(NH)NH2, CH2-imidazol-4-yl, CH2OH, CH(OH)CH3, CH2((4′-OH)-Ph), CH2SH, or lower cycloalkyl and R9b is hydrogen;

    • R10 is hydrogen, C1-6alkyl optionally substituted with an alkoxy, di(lower alkyl)-amino, or halogen, C1-6haloalkyl, C3-7cycloalkyl, heterocycloalkyl, aminoacyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl;

    • R11 is an optionally substituted C1-6alkyl, an optionally substituted cycloalkyl; an optionally substituted C2-6alkynyl, an optionally substituted C2-6alkenyl, or optionally substituted acyl, which includes but is not limited to C(O)(C1-6 alkyl); and

    • Y, R3 R12 and R22 are as defined above.





In one embodiment, compounds of Formula IV are disclosed:




embedded image




    • wherein the variables Y, R3, R7, R1, R9a, R9b, R10 and R22 are described herein.





In one embodiment, compounds of Formula V are disclosed:




embedded image




    • wherein the variables Y, R3, R7, R1, R9a, R9b, R10 and R22 are described herein.





In an alternative embodiment, compounds, methods, and compositions are provided for the treatment of a host infected with or exposed to hepatitis C.


In one embodiment, compounds of Formula VI are disclosed:




embedded image




    • wherein:

    • R41 is halogen (in particular F or Cl), OR3 (including OH), N3, NH2 or CN; and

    • the variables Y, R3, R4, and R12 are described herein.





In one embodiment, compounds of Formula VII are disclosed:




embedded image


Wherein the variables Y, R3, R4, R12 and R41 are described herein.


Metabolism of β-D-2′-deoxy-2′-α-fluoro-2′-β-C-substituted-N6-substituted-2,6-diaminopurine nucleotides

The metabolism of the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate involves the production of a 5′-monophosphate and the subsequent anabolism of the N6-methyl-2,6-diaminopurine base to generate the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine nucleoside as the 5′-monophosphate. The monophosphate is then further anabolized to the active species; the 5′-triphosphate. The β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine triphosphate has an IC50=0.15 μM against the HCV genotype 1b NS5B polymerase. The metabolic pathway for the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate is illustrated in Scheme 1 below.




embedded image


The metabolism of the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diaminopurine nucleotide involves both the formation of the β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-dimethyl-2,6-diaminopurine nucleoside triphosphate as well as the generation of the corresponding guanine nucleoside triphosphate. These metabolic pathways are illustrated in Schemes 2 and 3 below.




embedded image




embedded image



Stabilized Phosphate Prodrugs


Stabilized phosphate prodrugs are moieties that can deliver a mono, di, or triphosphate in vivo. For example, McGuigan has disclosed phosphoramidates in U.S. Pat. Nos. 8,933,053; 8,759,318; 8,658,616; 8,263,575; 8,119,779; 7,951,787 and 7,115,590. Alios has disclosed thiophosphoramidates in U.S. Pat. Nos. 8,895,723 and 8,871,737 incorporated by reference herein. Alios has also disclosed cyclic nucleotides in U.S. Pat. No. 8,772,474 incorporated by reference herein. Idenix has disclosed cyclic phosphoramidates and phosphoramidate/SATE derivatives in WO 2013/177219 incorporated by reference herein. Idenix has also disclosed substituted carbonyloxymethylphosphoramidate compounds in WO 2013/039920 incorporated by reference herein. Hostetler has disclosed lipid phosphate prodrugs, see, for example, U.S. Pat. No. 7,517,858. Hostetler has also disclosed lipid conjugates of phosphonate prodrugs, see, for example, U.S. Pat. Nos. 8,889,658; 8,846,643; 8,710,030; 8,309,565; 8,008,308; and 7,790,703. Emory University has disclosed nucleotide sphingoid and lipid derivatives in WO 2014/124430. RFS Pharma has disclosed purine nucleoside monophosphate prodrugs in WO 2010/091386. Cocrystal Pharma Inc. has also disclosed purine nucleoside monophosphate prodrugs in U.S. Pat. No. 9,173,893 incorporated by reference herein. HepDirect™ technology is disclosed in the article “Design, Synthesis, and Characterization of a Series of Cytochrome P(450) 3A-Activated Prodrugs (HepDirect Prodrugs) Useful for Targeting Phosph(on)ate-Based Drugs to the Liver,” (J. Am. Chem. Soc. 126, 5154-5163 (2004). Additional phosphate prodrugs include, but are not limited to phosphate esters, 3′,5′-cyclic phosphates including CycloSAL, SATE derivatives (S-acyl-2thioesters) and DTE (dithiodiethyl) prodrugs. For literature reviews that disclose non-limiting examples see: A. Ray and K. Hostetler, “Application of kinase bypass strategies to nucleoside antivirals,” Antiviral Research (2011) 277-291; M. Sofia, “Nucleotide prodrugs for HCV therapy,” Antiviral Chemistry and Chemotherapy 2011; 22-23-49; and S. Peyrottes et al., “SATE Pronucleotide Approaches: An Overview,” Mini Reviews in Medicinal Chemistry 2004, 4, 395. In one embodiment, a 5′-prodrug described in any of these patent filings or literature can be used in the R4 position of the presented compounds.


In one alternative embodiment, the stabilized phosphate prodrugs, include, but are not limited to those described in U.S. Pat. Nos. 9,173,893 and 8,609,627, incorporated by reference herein, including for processes of preparation. For example, 5′-prodrugs of Formula I-V can be represented by the group:




embedded image


In an alternate embodiment, 3′,5′-prodrugs of Formula I-V can be represented by the group:




embedded image



wherein:


when chirality exists at the phosphorous center it may be wholly or partially Rp or Sp or any mixture thereof.

    • Z is O or S;
    • R33 is selected from OR34,




embedded image



fatty alcohol derived (for example but not limited to:




embedded image



wherein R34, R35, and R36 are as defined below;


R31 and R32, when administered in vivo, are capable of providing the nucleoside monophosphate or thiomonophosphate, which may or may not be partially or fully resistant to 6-NH2 deamination in a biological system. Representative R31 and R32 are independently selected from: (a) OR34 where R34 is selected from H, Li, Na, K, phenyl and pyridinyl; phenyl and pyridinyl are substituted with one to three substituents independently selected from the group consisting of (CH2)0-6CO2R37 and (CH2)0-6CON(R37)2;


R37 is independently H, C-2 alkyl, the carbon chain derived from a fatty alcohol (such as oleyl alcohol, octacosanol, triacontanol, linoleyl alcohol, and etc) or C1-20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C1-5 alkyl, or C1-5 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, or cycloalkyl;


(b)




embedded image



or


(c) the ester of a D-amino acid or L-amino acid




embedded image



where R36 is restricted to those sidechains occurring in natural L-amino acids, and


R35 is H, C1-20 alkyl, the carbon chain derived from a fatty alcohol (such as oleyl alcohol, octacosanol, triacontanol, linoleyl alcohol, and etc) or C1-20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C1-5 alkyl, or C1-5 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, or cycloalkyl;


(d) R31 and R32 can come together to form a ring




embedded image



where R38 is H, C1-20 alkyl, C1-20 alkenyl, the carbon chain derived from a fatty alcohol (such as oleyl alcohol, octacosanol, triacontanol, linoleyl alcohol, etc) or C1-20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as, pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C1-5 alkyl, or C1-5 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, or cycloalkyl;


(e) R31 and R32 can come together to form a ring selected from




embedded image



where R39 is O or NH and


R40 is selected from H, C1-20 alkyl, C1-20 alkenyl, the carbon chain derived from a fatty acid (such as oleic acid, linoleic acid, and the like), and C1-20 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, cycloalkyl alkyl, cycloheteroalkyl, aryl, such as phenyl, heteroaryl, such as pyridinyl, substituted aryl, or substituted heteroaryl; wherein the substituents are C1-5 alkyl, or C1-5 alkyl substituted with a lower alkyl, alkoxy, di(lower alkyl)-amino, fluoro, C3-10 cycloalkyl, or cycloalkyl.


The compounds can be prepared, for example, by preparing the 5′-OH analogs, then converting these to the monophosphate analogs.


Embodiments

In particular embodiments:

    • (i) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (ii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (iii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a phosphoramidate;
    • (iv) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (v) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a monophosphate;
    • (vi) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a diphosphate;
    • (vii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a triphosphate;
    • (viii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (ix) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (x) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a phosphoramidate;
    • (xi) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (xii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a monophosphate;
    • (xiii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a diphosphate;
    • (xiv) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a triphosphate;
    • (xv) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (xvi) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (xvii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a phosphoramidate;
    • (xviii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (xix) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a monophosphate;
    • (xx) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is methyl, and R4 is a diphosphate;
    • (xxi) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a triphosphate;
    • (xxii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is propyl, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (xxiii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is propyl, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (xxiv) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is propyl, R3 is hydrogen, and R4 is a phosphoramidate;
    • (xxv) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is propyl, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (xxvi) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is propyl, R3 is hydrogen, and R4 is a monophosphate;
    • (xxvii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is propyl, R3 is hydrogen, and R4 is a diphosphate;
    • (xxviii) in Formula Ia, Y is NR1R2, Y is NR1R2, R1 is methyl, R2 is propyl, R3 is hydrogen, and R4 is a triphosphate;
    • (xxix) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is ethyl, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (xxx) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is ethyl, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (xxxi) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is ethyl, R3 is hydrogen, and R4 is a phosphoramidate;
    • (xxxii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is ethyl, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (xxxiii) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is ethyl, R3 is hydrogen, and R4 is a monophosphate;
    • (xxxiv) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is ethyl, R3 is hydrogen, and R4 is a diphosphate;
    • (xxxv) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is ethyl, R3 is hydrogen, and R4 is a triphosphate;
    • (xxxvi) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (xxxvii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (xxxviii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a phosphoramidate;
    • (xxxix) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (xl) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a monophosphate;
    • (xli) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a diphosphate;
    • (xlii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is methyl, R3 is hydrogen, and R4 is a triphosphate;
    • (xliii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (xliv) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (xlv) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a phosphoramidate;
    • (xlvi) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (xlvii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a monophosphate;
    • (xlviii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a diphosphate;
    • (xlix) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is hydrogen, R3 is hydrogen, and R4 is a triphosphate;
    • (1) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, R4 is a stabilized phosphate prodrug;
    • (li) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a stabilized thiophosphate prodrug;
    • (lii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a phosphoramidate;
    • (liii) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a thiophosphoramidate:
    • (liv) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a monophosphate;
    • (lv) in Formula Ib, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is methyl, and R4 is a diphosphate;
    • (lvi) in Formula Ia, Y is NR1R2, R1 is methyl, R2 is cyclopropyl, R3 is hydrogen, and R4 is a triphosphate.


In alternative embodiments of any of the above, the compound has an R22 substituent. In some of these specific embodiments, the R22 is F, amide or carbamate. In other specific aspects of the embodiments above, R22 is chloro, bromo, cyano, azido, ethyl, n-propyl, iso-propyl, n-butyl, iso-butyl, sec-butyl, tert-butyl and n-pentyl, 1,1-dimethylpropyl, 2,2-dimtheylpropyl, 3-methylbutyl, 1-methylbutyl, 1-ethylpropyl, vinyl, allyl, 1-butynyl, 2-butynyl, acetylenyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, —(CH2)-cyclopropyl, —(CH2)-cyclobutyl, —(CH2)-cyclopentyl, —(CH2)-cyclohexyl, aziridine, oxirane, thiirane, azetidine, oxetane, thietane, pyrrolidine, tetrahydrofuran, thiolane, pyrazolidine, piperidine, oxane, thiane, —(CH2)-aziridine, —(CH2)-oxirane, —(CH2)-thiirane, —(CH2)-azetidine, —(CH2)-oxetane, —(CH2)-thietane, —(CH2)-pyrrolidine, —(CH2)-tetrahydrofuran, —(CH2)-thiolane, —(CH2)-pyrazolidine, —(CH2)-piperidine, —(CH2)-oxane, —(CH2)-thiane, phenyl, pyridyl, —ONHC(═O)OCH3, —ONHC(═O)OCH2CH3, —NHOH, NHOCH3, —OCH3, OC2H5, —OPh, OCH2Ph, —SCH3, —SC2H5, —SPh, SCH2Ph, —NH(CH2)2NH2, —NH(CH2)2N(CH3)2, —NHNH2, —NHNHCH3, —N═NH, —N═NCH3, —N═NCH2CH3, —NHC(O)NHNH2, —NHC(S)NHNH2, —C(O)NHNH2, —NHSO2CH3, —NHSO2CH2CH3, —SO2NHCH3, —SO2N(CH3)2, —C(O)NH2, —C(O)NHCH3, —C(O)N(CH3)2, —CO2CH3, —CO2CH2CH3, —CO2Ph, CO2CH2Ph, —SO2CH3, —SO2CH2CH3, —SO2Ph, —SO2CH2Ph,




embedded image



—P(O)H(OH), —P(O)H(OCH3), —P(O)(OH)(OH), —P(O)(OH)(OCH3), —P(O)(OCH3)(OCH3), —P(O)(OH)(NH2), —P(O)(OH)(NHCH3), —P(O)(OH)N(CH3)2, —NHC(O)CH3, —NHC(O)CH2CH3, —NHC(O)CH(CH3)2, —NHC(O)OCH3, —NHC(O)OCH2CH3, —NHC(O)OCH(CH3)2, —NHC(O)OCH2CH2CH3, —NHC(O)OCH2CH2CH2CH3 and —NHC(O)OCH2CH2CH2CH2CH3;


In alternative embodiments of compounds (i) through (lvi), an L-nucleoside is used in Formula I-VII.


In an alternate embodiment, the Formula I R12 variable is CH2F.


In an alternate embodiment, the Formula I R12 variable is CHF2.


In an alternate embodiment, the Formula I R12 variable is CF3.


In one embodiment, a compound of Formula Ia is provided. Non-limiting examples of compounds of Formula Ia include:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment, a thiophosphoramidate of Formula Ia is provided. Non-limiting examples of thiophosphoramidates of Formula Ia include, but are not limited to:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment, a stabilized phosphate prodrug of Formula Ia is provided. Non-limiting examples of stabilized phosphate prodrugs of Formula Ia are illustrated below:




embedded image


embedded image


In another embodiment, a compound of Formula Ia is provided. Non-limiting examples of compounds of Formula Ia include:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment, a thiophosphoramidate of Formula Ia is provided. Non-limiting examples of thiophosphoramidates of Formula Ia include, but are not limited to:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment, a stabilized phosphate prodrug of Formula Ia is provided. Non-limiting examples of stabilized phosphate prodrugs of Formula Ia are illustrated below:




embedded image


embedded image


In one embodiment, a compound of Formula II is provided. Non-limiting examples of compounds of Formula II include:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In one embodiment, a compound of Formula I is provided. Non-limiting examples of compounds of Formula I include:




embedded image


embedded image


embedded image


In one embodiment, a compound of Formula II is provided. Non-limiting examples of compounds of Formula II include.




embedded image


embedded image


In one embodiment, and R4 is




embedded image


In one embodiment, a compound of Formula II is provided. Non-limiting examples of compounds of Formula II include:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In some embodiments, R3 is H and R4 is




embedded image


In some embodiments, R3 is H and R4 is




embedded image


In some embodiments, R3 is H and R4 is




embedded image


In one embodiment, a compound of Formula II is provided. Non-limiting examples of compounds of Formula II include:




embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


embedded image


In some embodiments, R3 is H and R4 is




embedded image


In some embodiments, R3 is H and R4 is




embedded image


In some embodiments, R3 is H and R4 is




embedded image


In some embodiments, R1 is CH3, R2 is H, R3 is H and R4 is




embedded image


In some embodiments, R1 is CH3, R2 is H, R3 is H and R4 is




embedded image


In some embodiments, R1 is CH3, R2 is H, R3 is H and R4 is




embedded image


In some embodiments, R1 is CH3, R2 is CH3, R3 is H and R4 is




embedded image


In some embodiments, R1 is CH3, R2 is CH3, R3 is H and R4 is




embedded image


In some embodiments, R1 is CH3, R2 is CH3, R3 is H and R4 is




embedded image


In some embodiments, R1 is cyclopropyl, R2 is CH3, R3 is H and R4 is




embedded image


In some embodiments, R1 is cyclopropyl, R2 is CH3, R3 is H and R4 is




embedded image


In some embodiments, R1 is cyclopropyl, R2 is CH3, R3 is H and R4 is




embedded image


II. Definitions

The following terms are used to describe the present invention. In instances where a term is not specifically defined herein, that term is given an art-recognized meaning by those of ordinary skill applying that term in context to its use in describing the present invention.


The term “alkyl” shall mean within its context, a linear, or branch-chained fully saturated hydrocarbon radical or alkyl group which can be optionally substituted (for example, with halogen, including F). For example, an alkyl group can have 1, 2, 3, 4, 5, 6, 7 or 8 carbon atoms (i.e., C1-C8 alkyl), 1, 2, 3, 4, 5 or 6 carbon atoms (i.e., C1-C6 alkyl) or 1 to 4 carbon atoms (i.e., C1-C4 alkyl). Examples of suitable alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl, isobutyl, sec-butyl, tert-butyl, pentyl, isopentyl, tert-pentyl, neopentyl, hexyl, 2-methylpentyl, 3-methylpentyl, 2,2-dimethylbutyl and 2,3-dimethylbutyl.


The term “alkenyl” refers to a non-aromatic hydrocarbon group which contains at least one double bond between adjacent carbon atoms and a similar structure to an alkyl group as otherwise described herein. For example, an alkenyl group can have 2 to 8 carbon atoms (i.e., C2-C8 alkenyl), or 2 to 4 carbon atoms (i.e., C2-C4 alkenyl). Examples of suitable alkenyl groups include, but are not limited to, ethenyl or vinyl (—CH═CH2), allyl (—CH2CH═CH2), 1-butenyl (—C═CH—CH2CH3) and 2-butenyl (—CH2CH═CHCH2). The alkenyl group can be optionally substituted as described herein.


The term “alkynyl” refers to a non-aromatic hydrocarbon group containing at least one triple bond between adjacent carbon atoms and a similar structure to an alkyl group as otherwise described herein. For example, an alkynyl group can have 2 to 8 carbon atoms (i.e., C2-C8 alkyne), or 2 to 4 carbon atoms (i.e., C2-C4 alkynyl). Examples of alkynyl groups include, but are not limited to, acetylenic or ethynyl and propargyl. The alkynyl group can be optionally substituted as described herein.


The term “acyl” refers to the moiety —C(O)R in which the carbonyl moiety is bonded to R, for example, —C(O)alkyl. R can be selected from alkoxy, alkyl, cycloalkyl, lower alkyl (i.e., C1-C4); alkoxyalkyl, including methoxymethyl; aralkyl- including benzyl, aryloxyalkyl- such as phenoxymethyl; aryl including phenyl optionally substituted with halogen, C1 to C4 alkyl or C1 to C4 alkoxy. In one embodiment, the term “acyl” refers to a mono, di or triphosphate.


The term “lower acyl” refers to an acyl group in which the carbonyl moiety is lower alkyl (i.e., C1-C4).


The term “alkoxy” refers to the group —OR′ where —OR′ is —O-alkyl, —O-alkenyl, —O— alkynyl, —O—(C0-C2)(cycloalkyl), —O—(C0-C2)(heterocyclo), —O—(C0-C2)(aryl), or —O—(C0-C2)(heteroaryl), each of which can be optionally substituted.


The term “amino” refers to the group —NH2.


The term “amino acid” or “amino acid residue” refers to a D- or L-natural or non-naturally occurring amino acid. Representative amino acids include, but are not limited to, alanine, β-alanine, arginine, asparagine, aspartic acid, cysteine, cysteine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine, among others.


The term “azido” refers to the group —N3.


The term “aryl” or “aromatic”, in context, refers to a substituted (as otherwise described herein) or unsubstituted monovalent aromatic radical having a single ring (e.g., phenyl or benzyl) or condensed rings (e.g., naphthyl, anthracenyl, phenanthrenyl, etc.) and can be bound to the compound according to the present invention at any available stable position on the ring(s) or as otherwise indicated in the chemical structure presented. The aryl group can be optionally substituted as described herein.


“Cycloalkyl”, “carbocycle”, or “carbocyclyl” refers to a saturated (i.e., cycloalkyl) or partially unsaturated (e.g., cycloakenyl, cycloalkadienyl, etc.) ring having 3 to 7 carbon atoms as a monocycle. Monocyclic carbocycles have 3 to 7 ring atoms, still more typically 5 or 6 ring atoms. Non-limiting examples of cycloalkyl groups include cyclopropyl, cyclobutyl, cyclopentyl, 1-cyclopent-1-enyl, 1-cyclopent-2-enyl, 1-cyclopent-3-enyl, cyclohexyl, 1-cyclohex-1-enyl, 1-cyclohex-2-enyl, and 1-cyclo-hex-3-enyl.


The term “cyano” refers to the group —CN.


The term “halogen” or “halo” refers to chloro, bromo, fluoro or iodo.


A heteroaryl ring system is a saturated or unsaturated ring with one or more nitrogen, oxygen, or sulfur atoms in the ring (monocyclic) including but not limited to imidazole, furyl, pyrrole, furanyl, thiene, thiazole, pyridine, pyrimidine, purine, pyrazine, triazole, oxazole, or fused ring systems such as indole, quinoline, etc., among others, which may be optionally substituted as described above. Heteroaryl groups include nitrogen-containing heteroaryl groups such as pyrrole, pyridine, pyridone, pyridazine, pyrimidine, pyrazine, pyrazole, imidazole, triazole, triazine, tetrazole, indole, isoindole, indolizine, purine, indazole, quinoline, isoquinoline, quinolizine, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, imidazopyridine, imidazotriazine, pyrazinopyridazine, acridine, phenanthridine, carbazole, carbazoline, perimidine, phenanthroline, phenacene, oxadiazole, benzimidazole, pyrrolopyridine, pyrrolopyrimidine and pyridopyrimidine; sulfur-containing aromatic heterocycles such as thiophene and benzothiophene; oxygen-containing aromatic heterocycles such as furan, pyran, cyclopentapyran, benzofuran and isobenzofuran; and aromatic heterocycles comprising two or more hetero atoms selected from among nitrogen, sulfur and oxygen, such as thiazole, thiadizole, isothiazole, benzoxazole, benzothiazole, benzothiadiazole, phenothiazine, isoxazole, furazan, phenoxazine, pyrazoloxazole, imidazothiazole, thienofuran, furopyrrole, pyridoxazine, furopyridine, furopyrimidine, thienopyrimidine and oxazole, among others, all of which may be optionally substituted.


The term “heterocycle” or “heterocyclo” refers to a cyclic group which contains at least one heteroatom, i.e., O, N, or S, and may be aromatic (heteroaryl) or non-aromatic. Exemplary non-aromatic heterocyclic groups for use in the present invention include, for example, pyrrolidinyl, piperidinyl, piperazinyl, N-methylpiperazinyl, imidazolinyl, pyrazolidinyl, imidazolidinyl, morpholinyl, tetrahydropyranyl, azetidinyl, oxetanyl, oxathiolanyl, pyridone, 2-pyrrolidone, ethyleneurea, 1,3-dioxolane, 1,3-dioxane, 1,4-dioxane, phthalimide, and succinimide, among others, all of which may be optionally substituted.


The term “hydroxyl” refers to the group —OH.


The term “nitro” refers to the group —NO2.


The term “pharmaceutically acceptable salt” or prodrug” is used throughout the specification to describe any pharmaceutically acceptable form (such as an ester, phosphoramidate, thiophosphoramidate, phosphate ester, salt of an ester, or a related group) of a β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide which, upon administration to a patient, provides the desired active compound. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids, which form a physiological acceptable anion, for example, tosylate, methanesulfonate, acetate, citrate, malonate, tartrate, succinate, benzoate, ascorbate, α-ketoglutarate, and α-glycerophosphate. Suitable inorganic salts may also be formed, including sulfate, nitrate, bicarbonate, and carbonate salts. Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid affording a physiologically acceptable anion. Alkali metal (for example, sodium, potassium, or lithium) or alkaline earth metal (for example calcium) salts of carboxylic acids can also be made.


“Pharmaceutically acceptable prodrug” refers to a compound that is metabolized, for example hydrolyzed or oxidized, in the host to form the compound of the present invention. Typical examples of prodrugs include compounds that have biologically labile protecting groups on a functional moiety of the active compound. Prodrugs include compounds that can be oxidized, reduced, aminated, deaminated, hydroxylated, dehydroxylated, hydrolyzed, dehydrolyzed, alkylated, dealkylated, acylated, deacylated, phosphorylated, dephosphorylated, thiophoshoramidated, dethiophoshoramidated, phoshoramidated or dephosphoramidated to produce the active compound. The compounds of this invention possess antiviral activity against HCV, or are metabolized to a compound that exhibits such activity. The β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleoside can also be administered as a 5′-phosphoether lipid, a bisphosphoramidate, a 3′,5′-cyclic phosphoramidate, a 3′,5′-cyclic thiophosphoramidate, a DTE conjugate, a mixed phosphoramidate-SATE derivative or a “SATE” derivative.


The term “phosphonic acid” refers to the group —P(O)(OH)2.


In one embodiment, the term purine or pyrimidine base includes, but is not limited to, adenine, N6-alkylpurines, N6-acylpurines (wherein acyl is —C(O)alkyl, —C(O)(aryl)C0-C4alkyl, or —C(O)(C0-C4alkyl)aryl), N6-benzylpurine, N6-halopurine, N6-vinylpurine, N6-acetylenic purine, N6-acyl purine, N6-hydroxyalkyl purine, N6-thioalkyl purine, N2-alkylpurines, N2-alkyl-6-thiopurines, thymine, cytosine, 5-fluorocytosine, 5-methylcytosine, 6-azapyrimidine, including 6-azacytosine, 2- and/or 4-mercaptopyrmidine, uracil, 5-halouracil, including 5-fluorouracil, C5-alkylpyrimidines, C5-benzylpyrimidines, C5-halopyrimidines, C5-vinylpyrimidine, C5-acetylenic pyrimidine, C5-acyl pyrimidine, C5-hydroxyalkyl purine, C5-amidopyrimidine, C5-cyanopyrimidine, C5-nitropyrimidine, C5-aminopyrimidine, N2-alkylpurines, N2-alkyl-6-thiopurines, 5-azacytidinyl, 5-azauracilyl, triazolopyridinyl, imidazolopyridinyl, pyrrolopyrimidinyl, and pyrazolo-pyrimidinyl. Purine bases include, but are not limited to, guanine, adenine, hypoxanthine, 2,6-diaminopurine, and 6-chloropurine. Functional oxygen and nitrogen groups on the base can be protected as necessary or desired. Suitable protecting groups are well known to those skilled in the art, and include benzyl, trimethylsilyl, dimethylhexylsilyl, t-butyldimethylsilyl, t-butyldiphenylsilyl, trityl, alkyl groups, and acyl groups such as acetyl and propionyl; methanesulfonyl, and p-toluenesulfonyl. Alternatively, the purine or pyrimidine base can optionally be substituted such that it forms a viable prodrug, which can be cleaved in vivo. Examples of appropriate substituents include an acyl moiety.


The term “substituted” or “optionally substituted” indicates that the moiety can have at least one additional substituent including, but not limited to, halogen (F, Cl, Br, I), OH, phenyl, benzyl, N3, CN, acyl, alkyl, including methyl; alkenyl, alkynyl, alkoxy, haloalkyl; including CHF2, CH2F and CF3; etc. In one embodiment, the term “substituted” or “optionally substituted” indicates that the moiety can have at least one additional substituent including, but not limited to, azido, cyano, halogen (fluoro, chloro, bromo, or iodo), alkyl, alkenyl, alkynyl, cycloalkyl, heterocycle, aryl, heteroaryl, haloalkyl, hydroxyl, alkoxy, amino, —NH(C1-C6 unsubstituted alkyl), —NH(C1-C6 substituted alkyl), —NH—(C0-C2alkyl)(C3-C8cycloalkyl), —NH—(C0-C2alkyl)(C3-C8heterocycle), —NH—(C0-C2alkyl)(aryl), —N(C1-C6 unsubstituted alkyl)2, —N(C1-C6 unsubstituted alkyl)(C1-C6 substituted alkyl), —N(C1-C6 substituted alkyl)2, —NH—(C0-C2alkyl)(C3-C8cycloalkyl), —NH—(C0-C2alkyl)(C3-C8heterocycle), —NH—(C0-C2alkyl)(aryl), acyl, nitro, sulfonic acid, sulfate, phosphonic acid, phosphate, phosphonate, or thiol.


The term “sulfonate esters”, represented by the formula, R14S(O)2OR15, comprise R14 wherein R14 is alkyl, haloalkyl, aralkyl or aryl. R15 is alkyl, aryl or aralkyl.


The term “sulfonic acid” refers to the group —SO2OH.


The term “thiol” refers to the group —SH.


The term “nitrogen-protecting group” as used herein refers to a moiety that is covalently attached to nitrogen and which can be removed, and typically replaced with hydrogen, when appropriate. For example, a nitrogen-protecting group may be a group that is removed in vivo after administration to a host, in vitro by a cell, or it may be removed during a manufacturing process. Suitable nitrogen-protecting groups useful in the present invention are described by Greene and Wuts in Protective Groups in Organic Synthesis (1991) New York, John Wiley and Sons, Inc.


The term “oxygen-protecting group” as used herein refers to a moiety that is covalently attached to oxygen and which can be removed, and typically replaced with hydrogen, when appropriate. For example, an oxygen-protecting group may be a group that is removed in vivo after administration to a host, in vitro by a cell, or it may be removed during a manufacturing process. Suitable oxygen-protecting groups useful in the present invention are described by Greene and Wuts in Protective Groups in Organic Synthesis (1991) New York, John Wiley and Sons, Inc.


“Phosphate” refers to the group —OP(O)(OH)2.


“Phosphate ester” refers to mono, di, and tri phosphates unless otherwise indicated.


The term “phosphoamidate”, “phosphoramidate”, or “phosphoroamidate” is a moiety that has a phosphorus bound to three oxygen groups and an amine (which may optionally be substituted). Suitable phosphoramidates useful in the present invention are described by Madela, Karolina and McGuigan in 2012, “Progress in the development of anti-hepatitis C virus nucleoside and nucleotide prodrugs”, Future Medicinal Chemistry 4(5), pages 625-650 10:1021/jm300074y and Dominique, McGuigan and Balzarini in 2004, “Aryloxy Phosphoramidate Triesters as Pro-Tides”, Mini Reviews in Medicinal Chemistry 4(4), pages 371-381. Additional phosphoramidates useful in the present invention are described in U.S. Pat. Nos. 5,233,031, 7,115,590, 7,547,704, 7,879,815, 7,888,330, 7,902,202, 7,951,789, 7,964,580, 8,071,568; 8,148,349, 8,263,575, 8,324,179, 8,334,270, 8,552,021, 8,563,530, 8,580,765, 8,735,372, 8,759,318; EP 2120565; EP 1143995; U.S. Pat. Nos. 6,455,513; and 8,334,270. Other phosphoramidates are described in the nucleoside patents described in the Background of the Invention.


Phosphoramidate groups for use in the present invention include those of the structures:




embedded image


embedded image


Other phosphoramidates for use in the present invention include those of the structure:




embedded image




    • wherein:

    • RP1 is an optionally substituted linear, branched, or cyclic alkyl group, or an optionally substituted aryl, heteroaryl or heterocyclic group or a linked combination thereof; and

    • RP2 is a —NRN1RN2 group or a B′ group;

    • wherein:

    • RN1 and RN2 are each independently H, C1-8alkyl, (C3-C7cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocycle)C0-C4alkyl-, or (heteroaryl)C0-C4alkyl-; which may be optionally substituted; or

    • RN1 and RN2 along with the nitrogen atom to which that are attached, join to form a 3 to 7 membered heterocyclic ring;

    • B is a







embedded image



group;

    • wherein:
    • R16 is hydrogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, (heteroaryl)C0-C4alkyl-, or the sidechain of an amino acid, for example a sidechain of an amino acid (as otherwise described herein) often selected from the group consisting of alanine, β-alanine, arginine, asparagine, aspartic acid, cysteine, cysteine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine (often R16 is hydrogen, methyl, isopropyl, or isobutyl);
    • R17 is hydrogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, (heteroaryl)C0-C4alkyl-, or the sidechain of an amino acid, for example a sidechain of an amino acid (as otherwise described herein) often selected from the group consisting of alanine, β-alanine, arginine, asparagine, aspartic acid, cysteine, cysteine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine (often R17 is hydrogen, methyl, isopropyl, or isobutyl);
    • R18 is hydrogen or C1-C3alkyl; or
    • R16 and R17 can form a (C3-C7)cycloalkyl or (C3-C7)heterocyclic group; or
    • R18 and R16 or R17 can form (C3-C6)heterocyclic group; and
    • R19 is hydrogen, (C1-C6)alkyl, (C3-C6)alkenyl, (C3-C6)alkynyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, (heteroaryl)C0-C4alkyl-; or
    • B′ is a




embedded image



group;

    • wherein:
    • R20 is hydrogen, (C1-C3)alkyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, or (heteroaryl)C0-C4alkyl-;
    • R21 is hydrogen, (C1-C3)alkyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, or (heteroaryl)C0-C4alkyl-; and
    • R18 and R19 are as defined above.


Preferred RP1 groups include optionally substituted phenyl, naphthyl, and monocyclic heteroaryl groups, especially those groups (particularly lipophilic groups) which enhance bioavailability of the compounds in the cells of the patient and which exhibit reduced toxicity, enhanced therapeutic index and enhanced pharmacokinetics (the compounds are metabolized and excreted more slowly).


The term phosphoramidate is used throughout the specification to describe a group that is found at the 5′ or 3′ position of the furanose ring of the nucleoside compound and forms a prodrug form of the nucleoside compound. In one embodiment, phosphoramidates can be found at both the 5′ and 3′ position of the furanose ring of the nucleoside compound and form a prodrug form of the nucleoside compound. In another embodiment, the phosphoramidate found at the 5′ position of the furanose ring of the nucleoside can form a cyclic phosphoramidate compound by forming a bond with the 3′-hydroxyl substituent at the 3′ position of the furanose ring of the nucleoside compound and form a prodrug form of the nucleoside compound.


The term “thiophosphoamidate”, “thiophosphoramidate”, or “thiophosphoroamidate” is a moiety that has a phosphorus bound to sulfur, two oxygen groups and an amine (which may optionally be substituted). Thiophosphoramidates useful in the present invention are described in U.S. Pat. No. 8,772,474 and WO 2012/040124.


Thiophosphoramidate groups for use in the present invention include those of the structures:




embedded image


embedded image


Other thiophosphoramidates include those of the structure:




embedded image




    • wherein:

    • RP1 is an optionally substituted linear, branched, or cyclic alkyl group, or an optionally substituted aryl, heteroaryl or heterocyclic group or a linked combination thereof; and

    • RP2 is a —RN1RN2 group or a B′ group;

    • wherein:

    • RN1 and RN2 are each independently H, C1-C8 alkyl, (C3-C7cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, or (heteroaryl)C0-C4alkyl-; or

    • RN1 and RN2 along with the nitrogen atom to which that are attached, join to form a 3 to 7 membered heterocyclic ring;

    • B′ is a







embedded image



group;

    • wherein:
    • R16 is hydrogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, (heteroaryl)C0-C4alkyl-, or the sidechain of an amino acid, for example a sidechain of an amino acid (as otherwise described herein) often selected from the group consisting of alanine, β-alanine, arginine, asparagine, aspartic acid, cysteine, cysteine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine (often R16 is hydrogen, methyl, isopropyl, or isobutyl);
    • R17 is hydrogen, (C1-C8)alkyl, (C2-C8)alkenyl, (C2-C8)alkynyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, (heteroaryl)C0-C4alkyl-, or the sidechain of an amino acid, for example a sidechain of an amino acid (as otherwise described herein) often selected from the group consisting of alanine, β-alanine, arginine, asparagine, aspartic acid, cysteine, cysteine, glutamic acid, glutamine, glycine, phenylalanine, histidine, isoleucine, lysine, leucine, methionine, proline, serine, threonine, valine, tryptophan, or tyrosine (often R17 is hydrogen, methyl, isopropyl, or isobutyl);
    • R18 is hydrogen or C1-C3alkyl; or
    • R16 and R17 can form a (C3-C7)cycloalkyl or (C3-C7)heterocyclic group; or
    • R18 and R16 or R17 can form (C3-C6) heterocyclic group; and
    • R19 is hydrogen, (C1-C6)alkyl, (C3-C6)alkenyl, (C3-C6)alkynyl, (C3-C8cycloalkyl)C0-C4alkyl-, (aryl)C0-C4alkyl-, (C3-C6heterocyclo)C0-C4alkyl-, (heteroaryl)C0-C4alkyl-; or
    • B′ is a




embedded image



group; and

    • R18, R19, R20 and R21 are as defined above.


Preferred RP1 groups include optionally substituted phenyl, naphthyl, and monocyclic heteroaryl groups, especially those groups (particularly lipophilic groups) which enhance bioavailability of the compounds into the cells of the patient and which exhibit reduced toxicity, enhanced therapeutic index and enhanced pharmacokinetics (the compounds are metabolized and excreted more slowly).


The thiophosphoramidate can be at the 5′ or 3′ position of the furanose ring of the nucleoside compound to form a prodrug form of the nucleoside compound. In one embodiment, thiophosphoramidates can be found at both the 5′ and 3′ position of the furanose ring of the nucleoside compound and form a prodrug form of the nucleoside compound. In another embodiment, the thiophosphoramidate found at the 5′ position of the furanose ring of the nucleoside can form a cyclic thiophosphoramidate compound by forming a bond with the 3′-hydroxyl substituent at the 3′ position of the furanose ring of the nucleoside compound and form a prodrug form of the nucleoside compound.


The term “D-configuration” as used in the context of the present invention refers to the principle configuration which mimics the natural configuration of sugar moieties as opposed to the unnatural occurring nucleosides or “L” configuration. The term “β” or “β anomer” is used with reference to nucleoside analogs in which the nucleoside base is configured (disposed) above the plane of the furanose moiety in the nucleoside analog.


The terms “coadminister” and “coadministration” or combination therapy are used to describe the administration of at least one of the 2′-deoxy-2′-α-fluoro-2′-β-C-nucleoside compounds according to the present invention in combination with at least one other active agent, for example where appropriate at least one additional anti-HCV agent, including other 2′-deoxy-2′-α-fluoro-2′-β-C-nucleoside agents which are disclosed herein. The timing of the coadministration is best determined by the medical specialist treating the patient. It is sometimes preferred that the agents be administered at the same time. Alternatively, the drugs selected for combination therapy may be administered at different times to the patient. Of course, when more than one viral or other infection or other condition is present, the present compounds may be combined with other agents to treat that other infection or condition as required.


The term “host”, as used herein, refers to a unicellular or multicellular organism in which a HCV virus can replicate, including cell lines and animals, and typically a human. The term host specifically refers to infected cells, cells transfected with all or part of a HCV genome, and animals, in particular, primates (including chimpanzees) and humans. In most animal applications of the present invention, the host is a human patient. Veterinary applications, in certain indications, however, are clearly anticipated by the present invention (such as chimpanzees). The host can be for example, bovine, equine, avian, canine, feline, etc.


Isotopic Substitution


The present invention includes compounds and the use of compounds with desired isotopic substitutions of atoms, at amounts above the natural abundance of the isotope, i.e., enriched. Isotopes are atoms having the same atomic number but different mass numbers, i.e., the same number of protons but a different number of neutrons. By way of general example and without limitation, isotopes of hydrogen, for example, deuterium (2H) and tritium (3H) may be used anywhere in described structures. Alternatively or in addition, isotopes of carbon, e.g., 13C and 14C, may be used. A preferred isotopic substitution is deuterium for hydrogen at one or more locations on the molecule to improve the performance of the drug. The deuterium can be bound in a location of bond breakage during metabolism (an α-deuterium kinetic isotope effect) or next to or near the site of bond breakage (a β-deuterium kinetic isotope effect). Achillion Pharmaceuticals, Inc. (WO/2014/169278 and WO/2014/169280) describes deuteration of nucleotides to improve their pharmacokinetics or pharmacodynamics, including at the 5-position of the molecule.


Substitution with isotopes such as deuterium can afford certain therapeutic advantages resulting from greater metabolic stability, such as, for example, increased in vivo half-life or reduced dosage requirements. Substitution of deuterium for hydrogen at a site of metabolic break down can reduce the rate of or eliminate the metabolism at that bond. At any position of the compound that a hydrogen atom may be present, the hydrogen atom can be any isotope of hydrogen, including protium (1H), deuterium (2H) and tritium (3H). Thus, reference herein to a compound encompasses all potential isotopic forms unless the context clearly dictates otherwise.


The term “isotopically-labeled” analog refers to an analog that is a “deuterated analog”, a “13C-labeled analog,” or a “deuterated/13C-labeled analog.” The term “deuterated analog” means a compound described herein, whereby a H-isotope, i.e., hydrogen/protium (1H), is substituted by a H-isotope, i.e., deuterium (2H). Deuterium substitution can be partial or complete. Partial deuterium substitution means that at least one hydrogen is substituted by at least one deuterium. In certain embodiments, the isotope is 90, 95 or 99% or more enriched in an isotope at any location of interest. In some embodiments it is deuterium that is 90, 95 or 99% enriched at a desired location. Unless indicated to the contrary, the deuteration is at least 80% at the selected location. Deuteration of the nucleoside can occur at any replaceable hydrogen that provides the desired results.


III. Methods of Treatment or Prophylaxis

Treatment, as used herein, refers to the administration of an active compound to a host that is infected with a HCV virus.


The term “prophylactic” or preventative, when used, refers to the administration of an active compound to prevent or reduce the likelihood of an occurrence of the viral disorder. The present invention includes both treatment and prophylactic or preventative therapies. In one embodiment, the active compound is administered to a host who has been exposed to and thus at risk of infection by a hepatitis C virus infection.


The invention is directed to a method of treatment or prophylaxis of a hepatitis C virus, including drug resistant and multidrug resistant forms of HCV and related disease states, conditions, or complications of an HCV infection, including cirrhosis and related hepatotoxicities, as well as other conditions that are secondary to a HCV infection, such as weakness, loss of appetite, weight loss, breast enlargement (especially in men), rash (especially on the palms), difficulty with clotting of blood, spider-like blood vessels on the skin, confusion, coma (encephalopathy), buildup of fluid in the abdominal cavity (ascites), esophageal varices, portal hypertension, kidney failure, enlarged spleen, decrease in blood cells, anemia, thrombocytopenia, jaundice, and hepatocellular cancer, among others. The method comprises administering to a host in need thereof an effective amount of at least one 0-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide as described herein, optionally in combination with at least one additional bioactive agent, for example, an additional anti-HCV agent, further in combination with a pharmaceutically acceptable carrier additive and/or excipient.


In yet another aspect, the present invention is a method for prevention or prophylaxis of a an HCV infection or a disease state or related or follow-on disease state, condition or complication of an HCV infection, including cirrhosis and related hepatotoxicities, weakness, loss of appetite, weight loss, breast enlargement (especially in men), rash (especially on the palms), difficulty with clotting of blood, spider-like blood vessels on the skin, confusion, coma (encephalopathy), buildup of fluid in the abdominal cavity (ascites), esophageal varices, portal hypertension, kidney failure, enlarged spleen, decrease in blood cells, anemia, thrombocytopenia, jaundice, and hepatocellular (liver) cancer, among others, said method comprising administering to a patient at risk with an effective amount of at least one compound according to the present invention as described above in combination with a pharmaceutically acceptable carrier, additive, or excipient, optionally in combination with another anti-HCV agent. In another embodiment, the active compounds of the invention can be administered to a patient after a hepatitis-related liver transplantation to protect the new organ.


The 5′-stabilized β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide can be administered if desired as any salt or prodrug that upon administration to the recipient is capable of providing directly or indirectly the parent compound, or that exhibits activity itself. Nonlimiting examples are the pharmaceutically acceptable salts and a compound, which has been modified at a function group, such as a hydroxyl or amine function, to modify the biological activity, pharmacokinetics, half-life, controlled delivery, lipophilicity, absorption kinetics, ease of phosphorylation to the active 5′-triphosphate or efficiency of delivery using a desired route of administration of the compound. Methods to modify the properties of an active compound to achieve target properties are known to those of skill in the art or can easily be assessed by standard methods, for example, acylation, phosphorylation, thiophosphoramidation, phosphoramidation, phosphonation, alkylation, or pegylation.


IV. Pharmaceutical Compositions

In an aspect of the invention, pharmaceutical compositions according to the present invention comprise an anti-HCV virus effective amount of at least one of the 5′-stabilized β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide compounds described herein, optionally in combination with a pharmaceutically acceptable carrier, additive, or excipient, further optionally in combination or alternation with at least one other active compound.


In an aspect of the invention, pharmaceutical compositions according to the present invention comprise an anti-HCV effective amount of at least one of the active β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide compounds described herein, optionally in combination with a pharmaceutically acceptable carrier, additive, or excipient, further optionally in combination with at least one other antiviral, such as an anti-HCV agent.


The invention includes pharmaceutical compositions that include an effective amount to treat a hepatitis C virus infection, of one of the β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide compounds of the present invention or its salt or prodrug, in a pharmaceutically acceptable carrier or excipient. In an alternative embodiment, the invention includes pharmaceutical compositions that include an effective amount to prevent a hepatitis C virus infection, of one of the β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide compounds of the present invention or its salt or prodrug, in a pharmaceutically acceptable carrier or excipient.


One of ordinary skill in the art will recognize that a therapeutically effective amount will vary with the infection or condition to be treated, its severity, the treatment regimen to be employed, the pharmacokinetics of the agent used, as well as the patient or subject (animal or human) to be treated, and such therapeutic amount can be determined by the attending physician or specialist.


The 5′-stabilized β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified —N6-substituted purine nucleotide compounds according to the present invention can be formulated in an admixture with a pharmaceutically acceptable carrier. In general, it is preferable to administer the pharmaceutical composition in orally-administrable form, but certain formulations may be administered via a parenteral, intravenous, intramuscular, topical, transdermal, buccal, subcutaneous, suppository, or other route, including intranasal spray. Intravenous and intramuscular formulations are often administered in sterile saline. One of ordinary skill in the art may modify the formulations to render them more soluble in water or other vehicle, for example, this can be easily accomplished by minor modifications (salt formulation, esterification, etc.) which are well within the ordinary skill in the art. It is also well within the routineers' skill to modify the route of administration and dosage regimen of a particular compound in order to manage the pharmacokinetics of the present compounds for maximum beneficial effect in patients.


In certain pharmaceutical dosage forms, the prodrug form of the compounds, especially including acylated (acetylated or other), and ether (alkyl and related) derivatives, phosphate esters, thiophosphoramidates, phosphoramidates, and various salt forms of the present compounds, are preferred. One of ordinary skill in the art will recognize how to readily modify the present compounds to prodrug forms to facilitate delivery of active compounds to a targeted site within the host organism or patient. The routineer also will take advantage of favorable pharmacokinetic parameters of the prodrug forms, where applicable, in delivering the present compounds to a targeted site within the host organism or patient to maximize the intended effect of the compound.


The amount of compound included within therapeutically active formulations according to the present invention is an effective amount for treating the HCV infection, reducing the likelihood of a HCV infection or the inhibition, reduction, and/or abolition of HCV or its secondary effects, including disease states, conditions, and/or complications which occur secondary to HCV. In general, a therapeutically effective amount of the present compound in pharmaceutical dosage form usually ranges from about 0.001 mg/kg to about 100 mg/kg per day or more, more often, slightly less than about 0.1 mg/kg to more than about 25 mg/kg per day of the patient or considerably more, depending upon the compound used, the condition or infection treated and the route of administration. The active nucleoside compound according to the present invention is often administered in amounts ranging from about 0.1 mg/kg to about 15 mg/kg per day of the patient, depending upon the pharmacokinetics of the agent in the patient. This dosage range generally produces effective blood level concentrations of active compound which may range from about 0.001 to about 100, about 0.05 to about 100 micrograms/cc of blood in the patient.


Often, to treat, prevent or delay the onset of these infections and/or to reduce the likelihood of an HCV virus infection, or a secondary disease state, condition or complication of HCV, the compositions will be administered in oral dosage form in amounts ranging from about 250 micrograms up to about 500 mg or more at least once a day, for example, at least 25, 50, 100, 150, 250 or 500 milligrams, up to four times a day. The present compounds are often administered orally, but may be administered parenterally, topically, or in suppository form, as well as intranasally, as a nasal spray or as otherwise described herein.


In the case of the co-administration of the present compounds in combination with another anti-HCV compound as otherwise described herein, the amount of the compound according to the present invention to be administered ranges from about 0.01 mg/kg of the patient to about 500 mg/kg. or more of the patient or considerably more, depending upon the second agent to be co-administered and its potency against the virus, the condition of the patient and severity of the disease or infection to be treated and the route of administration. The other anti-HCV agent may for example be administered in amounts ranging from about 0.01 mg/kg to about 500 mg/kg. In certain preferred embodiments, these compounds may be often administered in an amount ranging from about 0.5 mg/kg to about 50 mg/kg or more (usually up to about 100 mg/kg), generally depending upon the pharmacokinetics of the two agents in the patient. These dosage ranges generally produce effective blood level concentrations of active compound in the patient.


For purposes of the present invention, a prophylactically or preventive effective amount of the compositions according to the present invention falls within the same concentration range as set forth above for therapeutically effective amount and is usually the same as a therapeutically effective amount.


Administration of the active compound may range from continuous (intravenous drip) to several oral or intranasal administrations per day (for example, Q.I.D.) or transdermal administration and may include oral, topical, parenteral, intramuscular, intravenous, sub-cutaneous, transdermal (which may include a penetration enhancement agent), buccal, and suppository administration, among other routes of administration. Enteric coated oral tablets may also be used to enhance bioavailability of the compounds for an oral route of administration. The most effective dosage form will depend upon the bioavailability/pharmacokinetics of the particular agent chosen as well as the severity of disease in the patient. Oral dosage forms are particularly preferred, because of ease of administration and prospective favorable patient compliance.


To prepare the pharmaceutical compositions according to the present invention, a therapeutically effective amount of one or more of the compounds according to the present invention is often intimately admixed with a pharmaceutically acceptable carrier according to conventional pharmaceutical compounding techniques to produce a dose. A carrier may take a wide variety of forms depending on the form of preparation desired for administration, e.g., oral or parenteral. In preparing pharmaceutical compositions in oral dosage form, any of the usual pharmaceutical media may be used. Thus, for liquid oral preparations such as suspensions, elixirs, and solutions, suitable carriers and additives including water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents, and the like may be used. For solid oral preparations such as powders, tablets, capsules, and for solid preparations such as suppositories, suitable carriers and additives including starches, sugar carriers, such as dextrose, manifold, lactose, and related carriers, diluents, granulating agents, lubricants, binders, disintegrating agents, and the like may be used. If desired, the tablets or capsules may be enteric-coated or sustained release by standard techniques. The use of these dosage forms may significantly enhance the bioavailability of the compounds in the patient.


For parenteral formulations, the carrier will usually comprise sterile water or aqueous sodium chloride solution, though other ingredients, including those which aid dispersion, also may be included. Of course, where sterile water is to be used and maintained as sterile, the compositions and carriers must also be sterilized. Injectable suspensions may also be prepared, in which case appropriate liquid carriers, suspending agents, and the like may be employed.


Liposomal suspensions (including liposomes targeted to viral antigens) may also be prepared by conventional methods to produce pharmaceutically acceptable carriers. This may be appropriate for the delivery of free nucleosides, acyl/alkyl nucleosides or phosphate ester prodrug forms of the nucleoside compounds according to the present invention.


In typical embodiments according to the present invention, the compounds and compositions are used to treat, prevent or delay a HCV infection or a secondary disease state, condition or complication of HCV.


V. Combination and Alternation Therapy

It is well recognized that drug-resistant variants of viruses can emerge after prolonged treatment with an antiviral agent. Drug resistance most typically occurs by mutation of a gene that encodes for an enzyme used in viral replication. The efficacy of a drug against an HCV infection, can be prolonged, augmented, or restored by administering the compound in combination or alternation with another, and perhaps even two or three other, antiviral compounds that induce a different mutation or act through a different pathway, from that of the principle drug. Alternatively, the pharmacokinetics, bio distribution, half-life, or other parameter of the drug can be altered by such combination therapy (which may include alternation therapy if considered concerted). Since the disclosed β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides are NS5B polymerase inhibitors, it may be useful to administer the compound to a host in combination with, for example a:

    • (1) Protease inhibitor, such as an NS3/4A protease inhibitor;
    • (2) NS5A inhibitor;
    • (3) Another NS5B polymerase inhibitor;
    • (4) NS5B non-substrate inhibitor;
    • (5) Interferon alfa-2a, which may be pegylated or otherwise modified, and/or ribavirin;
    • (6) Non-substrate-based inhibitor;
    • (7) Helicase inhibitor;
    • (8) Antisense oligodeoxynucleotide (S-ODN);
    • (9) Aptamer;
    • (10) Nuclease-resistant ribozyme;
    • (11) iRNA, including microRNA and SiRNA;
    • (12) Antibody, partial antibody or domain antibody to the virus, or
    • (13) Viral antigen or partial antigen that induces a host antibody response.


Non limiting examples of anti-HCV agents that can be administered in combination with the β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides of the invention are:

    • (i) protease inhibitors such as telaprevir (Incivek®), boceprevir (Victrelis™), simeprevir (Olysio™), paritaprevir (ABT-450), ACH-2684; AZD-7295; BMS-791325; danoprevir; Filibuvir; GS-9256; GS-9451; MK-5172; Setrobuvir; Sovaprevir; Tegobuvir; VX-135; VX-222 and ALS-220;
    • (ii) NS5A inhibitor such as ACH-2928, ACH-3102, IDX-719, daclatasvir, ledispasvir and Ombitasvir (ABT-267);
    • (iii) NS5B inhibitors such as ACH-3422; AZD-7295; Clemizole; ITX-5061; PPI-461; PPI-688, Sovaldi®, MK-3682, and mericitabine;
    • (iv) NS5B inhibitors such as ABT-333, MBX-700; and,
    • (v) Antibody such as GS-6624.


If the β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotide is administered to treat advanced hepatitis C virus leading to liver cancer or cirrhosis, in one embodiment, the compound can be administered in combination or alternation with another drug that is typically used to treat hepatocellular carcinoma (HCC), for example, as described by Andrew Zhu in “New Agents on the Horizon in Hepatocellular Carcinoma” Therapeutic Advances in Medical Oncology, V 5(1), January 2013, 41-50. Examples of suitable compounds for combination therapy where the host has or is at risk of HCC include anti-angiogenic agents, sunitinib, brivanib, linifanib, ramucirumab, bevacizumab, cediranib, pazopanib, TSU-68, lenvatinib, antibodies against EGFR, mTor inhibitors, MEK inhibitors, and histone decetylace inhibitors.


Drugs that are currently approved for influenza are Amantadine, Rimantadine and Oseltamivir. Any of these drugs can be used in combination or alternation with an active compound provided herein to treat a viral infection susceptible to such. Ribavirin is used to treat measles, Influenza A, influenza B, parainfluenza, severe RSV bronchiolitis and SARS as well as other viral infections, and therefore is particularly useful in combination with the present compound for treatment of the host infected with a single stranded RNA virus. Palivizumab is approved for use in infants with high risk for RSV infection.


Currently, there are no approved drugs for West Nile virus. Physicians are recommended to provide intensive support therapy, which may involve hospitalization, intravenous fluids, use of a ventilator to assist breathing, medications to control seizures, brain swelling, nausea and vomiting, and the use of antibiotics to prevent bacterial infections for making the disease even worse. This highlights the importance of the present compounds for viral medical therapy.


VI. Process of Preparation of β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-Substituted Purine Nucleotides of the Invention

General methods for providing the compounds of the present invention are known in the art or described herein. The synthesis of 2′-chloro nucleotides is described in US 20150366888, WO 2014058801; WO 2015/066370 and WO 2015200219.


The following abbreviations are used in the synthetic schemes.

    • CBr4: Carbon tetrabromide
    • DBU: 1,8-Diazabicyclo[5.4.0]undec-7-ene
    • DCM: Dichloromethane
    • THF: Tetrahydrofuran (THF), anhydrous
    • EtOAc: Ethyl acetate
    • EtOH: Ethanol
    • Li(OtBu)3AlH: Lithium tri-tert-butoxyaluminum hydride
    • Na2SO4: Sodium sulphate (anhydrous)
    • MeCN: Acetonitrile
    • MeNH2: Methylamine
    • MeOH: Methanol
    • Na2SO4: Sodium sulfate
    • NaHCO3: Sodium bicarbonate
    • NH4Cl: Ammonium chloride
    • NH4OH: Ammonium hydroxide
    • PE: Petroleum ether
    • Ph3P: Triphenylphosphine
    • Silica gel (230 to 400 mesh, Sorbent)
    • t-BuMgCl: t-Butyl magnesium chloride
    • t-BuOK: Sodium tert-butoxide
    • t-BuOH: Tert-butanol


EXAMPLES

General Methods



1H, 19F and 31P NMR spectra were recorded on a 300 MHz Fourier transform Brücker spectrometer. Spectra were obtained from samples prepared in 5 mm diameter tubes in CDCl3, CD3OD or DMSO-d6. The spin multiplicities are indicated by the symbols s (singlet), d (doublet), t (triplet), m (multiplet) and, br (broad). Coupling constants (J) are reported in Hz. MS spectra were obtained using electrospray ionization (ESI) on an Agilent Technologies 6120 quadrupole MS apparatus. The reactions were generally carried out under a dry nitrogen atmosphere using Sigma-Aldrich anhydrous solvents. All common chemicals were purchased from commercial sources.




embedded image


Example 1. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate
Step 1. Preparation of ((2R,3R,4R,5R)-3-(benzoyloxy)-5-bromo-4-fluoro-4-methyltetrahydrofuran-2-yl)methylbenzoate (2)

To a solution of (2R)-3,5-di-1-benzoyl-2-fluoro-2-C-methyl-D-ribono-γ-lactone (24.8 g, 66.6 mmol) in dry THE (333 mL), under a nitrogen atmosphere and cooled to −30° C., was added lithium tri-tert-butoxyaluminum hydride (1.0 M in THF, 22.6 mL, 22.6 mmol) dropwise. After completion of the addition the reaction mixture was slowly warmed up to −15° C. over 90 min then EtOAc was added (300 mL) and the mixture was quenched with a saturated aq. NH4Cl solution (200 mL). The resulting solution was filtered on Celite® and the filtrate was extracted twice with EtOAc. The combined organics were dried (Na2SO4), filtered and concentrated. The residue was taken up in dry DCM (225 mL) under a nitrogen atmosphere, cooled to −20° C., then PPh3 (19.1 g, 72.8 mmol) was added. After 10 min of stirring at −20° C., CBr4 (26.0 g, 78.4 mmol) was added and the reaction mixture was allowed to slowly warm up to 0° C. over 2 h. The resulting mixture was poured onto a silica gel column and eluted with PE/EtOAc (gradient 100:0 to 80:20). The fractions containing the α-bromofuranoside were collected and concentrated to afford the product 2 (18.1 g, 41.3 mmol, 62% over two steps) as a thick colorless oil.



1H NMR (300 MHz, CDCl3) δ 8.15-8.11 (m, 2H), 8.04-8.01 (m, 2H), 7.64-7.55 (m, 2H), 7.51-7.41 (m, 4H), 6.34 (d, J=1.6 Hz, 1H), 5.29 (dd, J=5.5, 3.1 Hz, 1H), 4.89-4.85 (m, 1H), 4.78 (dd, J=12.5, 3.2 Hz, 1H), 4.63 (dd, J=12.5, 4.5 Hz, 1H), 1.72 (d, J=21.6 Hz, 3H). 19F NMR (282 MHz, CDCl3) δ −150.0.


Step 2. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(benzoyloxymethyl)-4-fluoro-4-methyltetrahydrofuran-3-ylbenzoate (3)

2-Amino-6-chloropurine (2.63 g, 15.5 mmol) was suspended in t-BuOH (54 mL) under a nitrogen atmosphere. The reaction mixture was heated to 30° C. then potassium tert-butoxide (1.69 g, 15.1 mmol) was added. After 45 min a solution of bromofuranoside 2 (2.24 g, 5.12 mmol) dissolved in anhydrous MeCN (6 mL) was added, the reaction mixture was heated to 65° C. for 16 h then cooled down to room temperature. A saturated aq. NH4Cl solution (70 mL) was added and the resulting solution was extracted with EtOAc (3×60 mL). The combined organics were dried (Na2SO4), filtered and concentrated. The residue was purified twice by column chromatography (gradient PE/EtOAc 80:20 to 0:100 then 60:40 to 20:80) to afford the product 3 (1.56 g, 2.96 mmol, 57%) as a white solid.



1H NMR (300 MHz, CDCl3) δ 8.05-8.02 (m, 2H), 7.95-7.92 (m, 2H), 7.88 (s, 1H), 7.63-7.57 (m, 1H), 7.53-7.41 (m, 3H), 7.35-7.30 (m, 2H), 6.43 (dd, J=22.6, 9.1 Hz, 1H), 6.12 (d, J=18.3 Hz, 1H), 5.34 (br s, 2H), 5.00 (dd, J=11.9, 4.5 Hz, 1H), 4.79-4.73 (m, 1H), 4.60 (dd, J=11.9, 5.3 Hz, 1H), 1.34 (d, J=22.6 Hz, 3H). 19F NMR (282 MHz, CDCl3) δ −157.0. MS (ESI) m/z calcd. for C25H22FN5O5 [M+H]+ 526.9; found 527.0.


Step 3. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (4)

To a solution of compound 3 (575 mg, 1.09 mmol) in MeOH (9 mL) was added methylamine (33% in absolute EtOH, 1.7 mL, 1.81 mmol). The reaction mixture was heated to 85° C. in a sealed tube for 16 h, cooled down to room temperature and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 85:15) then reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford the product 4 (286 mg, 0.91 mmol, 84%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 8.06 (s, 1H), 6.11 (d, J=18.1 Hz, 1H), 4.41 (dd, J=24.4, 9.1 Hz, 1H), 4.07-4.01 (m, 2H), 3.86 (dd, J=12.9, 3.3 Hz, 1H), 3.04 (br s, 3H), 1.16 (d, J=22.3 Hz, 3H). 19F NMR (282 MHz, CD3OD) δ −163.7. MS (ESI) m/z calcd. for C12H19FN6O3 [M+H]+ 313.1; found 313.2.


Step 4. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (5)

To a solution of compound 4 (114 mg, 365 μmol) in dry THE (4 mL), under a nitrogen atmosphere and cooled to 0° C. was added t-butyl magnesium chloride (1.0 M in THF, 0.66 mL, 660 μmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. then a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate, Ross, B. S., Reddy, P. G., Zhang, H. R., Rachakonda, S., and Sofia, M. J., J. Org, Chem., (2011), (253 mg, 558 μmol) dissolved in dry THE (1 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min followed by 18 h at room temperature then quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried, filtered (Na2SO4) and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) then reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford product 5 (a mixture of diastereomers, 101 mg, 174 μmol, 48%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.83 (s, 0.55H), 7.82 (s, 0.45H), 7.38-7.16 (m, 5H), 6.15 (d, J=18.5 Hz, 0.45H), (d, J=18.8 Hz, 0.55H), 4.99-4.88 (overlapped with H2O, m, 1H), 4.65-4.36 (m, 3H), 4.25-4.17 (m, 1H), 3.97-3.85 (m, 1H), 3.05 (br s, 3H), 1.32-1.28 (m, 3H), 1.25-1.15 (m, 9H). 19F NMR (282 MHz, CD3OD) δ −162.8 (s), −163.3 (s). 3P NMR (121 MHz, CD3OD) δ 4.10 (s), 3.99 (s). MS (ESI) m/z calcd. for C24H34FN7O7P [M+H]+ 582.2; found 582.2.




embedded image


Example 2. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-Amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (7)
Step 1. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (6)

To a solution of compound 3, from Example 1, (500 mg, 0.95 mmol) in MeOH (6 mL) was added dimethylamine hydrochloride (783 mg, 9.6 mmol) and 1,8-diazabicyclo[5.4.0]undec-7-ene (1.43 mL, 9.6 mmol). The reaction mixture was heated at 85° C. in a sealed tube for 6 h, cooled down to room temperature and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 85:15) then by reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford product 6 (200 mg, 0.61 mmol, 64%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 8.07 (s, 1H), 6.14 (d, J=18.1 Hz, 1H), 4.41 (dd, J=24.4, 9.2 Hz, 1H), 4.08-4.02 (m, 2H), 3.87 (dd, J=12.8, 2.9 Hz, 1H), 3.42 (br s, 6H), 1.16 (d, J=22.0 Hz, 3H). 19F NMR (282 MHz, CD3OD) δ −163.8. MS (ESI) m/z calcd. for C13H2OFN6O3 [M+H]+ 327.2; found 327.2.


Step 2. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (7)

To a solution of compound 6 (80 mg, 245 μmol) in dry THE (4 mL), under a nitrogen atmosphere and cooled to 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 0.64 mL, 640 μmol) drop-wise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. then a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (167 mg, 367 μmol) dissolved in dry THE (4 mL) was added drop-wise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried, filtered (Na2SO4) and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) and then by reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford the product 7 (mixture of diastereomers, 35 mg, 58 μmol, 24%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.83 (s, 0.5H), 7.82 (s, 0.5H), 7.34-7.16 (m, 5H), 6.15 (d, J=18.7 Hz, 0.5H), 6.13 (d, J=18.8 Hz, 0.5H), 4.99-4.85 (overlapped with H2O, m, 1H), 4.65-4.26 (m, 3H), 4.27-4.12 (m, 1H), 3.99-3.81 (m, 1H), 3.42, 3.41 (2br s, 6H), 1.36-1.25 (m, 3H), 1.24-1.11 (m, 9H). 19F NMR (282 MHz, CD3OD) δ −162.7 (s), −163.2 (s). 31P NMR (121 MHz, CD3OD) δ 4.08 (s), 4.00 (s). MS (ESI) m/z calcd. for C25H36FN7O7P [M+H]+ 596.5; found 596.2.




embedded image


Example 3. Preparation of Isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-cyclopropylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (9)
Step 1. Preparation of (2R,3R,4R,5R)-5-(2-Amino-6-(N-methyl-cyclopropylamino)-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (8)

To a solution of compound 3 (600 mg, 1.14 mmol) in MeOH (10 mL) was added N-methylcyclopropylamine hydrochloride (366 mg, 3.40 mmol) and triethylamine (470 μL, 3.40 mmol). The reaction mixture was heated at 100° C. in a sealed tube for 15 h and cooled down to room temperature. An aqueous solution containing 30% NH4OH (4 mL) was added and the reaction mixture was heated at 100° C. in a sealed tube for 2 h, cooled down and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford product 8 (351 mg, 0.99 mmol, 87%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 8.13 (s, 1H), 6.15 (d, J=18.0 Hz, 1H), 4.40 (dd, J=24.3, 9.0 Hz, 1H), 4.06-4.02 (m, 2H), 3.89-3.83 (m, 1H), 3.32 (m, 3H), 3.18-3.11 (m, 1H), 1.16 (d, J=22.2 Hz, 3H), 0.96-0.89 (m, 2H), 0.74-0.69 (m, 2H). 19F NMR (282 MHz, CD3OD) δ −163.8. MS (ESI) m/z calcd. for C15H22FN6O3 [M+H]+ 353.2; found 353.2.


Step 2. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-cyclopropylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (9)

To a solution of compound 8 (200 mg, 0.57 mmol) in dry THE (15 mL) at 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 680 μL, 0.68 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (283 mg, 0.62 mmol) dissolved in dry THE (4 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) and then by reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford product 9 (mixture of 2 diastereoisomers, 160 mg, 0.26 mmol, 45%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.85 (m, 1H), 7.38-7.16 (m, 5H), 6.18 (d, J=18.6 Hz) and 6.16 (d, J=18.9 Hz, 1H), 4.95-4.90 (overlapped with H2O, m, 1H), 4.58-4.47 (m, 3H), 4.22-4.19 (m, 1H), 3.95-3.87 (m, 1H), 3.36-3.34 (overlapped with MeOH, m, 3H), 3.19-3.12 (m, 1H), 1.32-1.22 (m, 12H), 0.96-0.89 (m, 2H), 0.74-0.69 (m, 2H). 31P NMR (121 MHz, CD3OD) δ 4.11 (s), 4.02 (s). MS (ESI) m/z calcd. for C27H38FN7O7P [M+H]+ 622.2; found 622.2.




embedded image


Example 4. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2,6-bis-methylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (12)
Step 1. Preparation of (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(benzoyloxymethyl)-4-fluoro-4-methyltetrahydrofuran-3-yl benzoate (10)

The compound 2,6-dichloropurine (1.30 g, 6.86 mmol) was suspended in t-BuOH (25 mL) under a nitrogen atmosphere. Potassium tert-butoxide (778 mg, 6.92 mmol) was added portion-wise then the reaction mixture was stirred at room temperature. After 1 h, a solution of bromofuranoside 2 (1.0 g, 2.29 mmol) dissolved in anhydrous MeCN (20 mL) was added and the reaction mixture was heated at 65° C. overnight and then cooled down to room temperature. A saturated aq. NH4Cl solution was added and the resulting solution was extracted with EtOAc (3 times). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient PE/EtOAc 100:0 to 0:100) to afford product 10 (148 mg, 0.27 mmol, 12%) as a sticky solid.



1H NMR (300 MHz, CDCl3) δ 8.31 (s, 1H), 8.12-8.09 (m, 2H), 8.02-7.99 (m, 2H), 7.64-7.39 (m, 6H), 6.38 (d, J=17.2 Hz, 1H), 6.02 (dd, J=21.2, 8.9 Hz, 1H), 4.90-4.68 (m, 3H), 1.33 (d, J=22.4 Hz, 3H). 19F NMR (282 MHz, CDCl3) δ −158.0. MS (ESI) m/z calcd. for C25H20Cl2FN4O5 [M+H]+ 546.4; found 546.3.


Step 2. Preparation of (2R,3R,4R,5R)-5-(2,6-bis-methylamino-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (11)

A solution of compound 10 (148 mg, 0.27 mmol) in methylamine (33% in EtOH, 30 mL) was heated at 130° C. in a sealed tube for 4 days, cooled down to room temperature and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 50:50) followed by reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford product 11 (33 mg, 0.10 mmol, 37%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 8.00 (s, 1H), 6.12 (d, J=18.5 Hz, 1H), 4.51 (dd, J=24.4, 9.5 Hz, 1H), 4.06-3.85 (m, 3H), 3.04 (s, 3H), 2.93 (s, 3H), 1.20 (d, J=22.4 Hz, 3H). 19F NMR (282 MHz, CD3OD) δ −163.2. MS (ESI) m/z calcd. for C13H2OFN6O3 [M+H]+ 327.2; found 327.2.


Step 3. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2,6-bis-methylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (12)

To a solution of compound 11 (55 mg, 0.17 mmol) in dry THE (2 mL) at 0° C. was added tert-butyl magnesium chloride (1 M in THF, 304 DL, 0.30 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. and then 15 min at room temperature. The solution was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (115 mg, 0.25 mmol) dissolved in dry THF (1 mL) was dropwise added over 10 min. The mixture was warmed slowly to room temperature and stirred for 4 days. The reaction was quenched with a saturated aq. NH4Cl solution and extracted with EtOAc (3 times). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 50:50) to yield product 12 (mixture of diastereomers, 13 mg, 0.02 mmol, 13%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 7.78 (s, 1H), 7.35-7.12 (m, 5H), 6.13 (d, J=19.1 Hz, 0.53H), 6.10 (d, J=19.2 Hz, 0.47H), 4.99-4.78 (overlapped with H2O, m, 1H), 4.72-4.46 (m, 3H), 4.24-4.15 (m, 1H), 3.79-3.92 (m, 1H), 3.02 (br s, 3H), 2.92 (s+s, 3H), 1.29-1.11 (m, 12H). 19F NMR (282 MHz, CD3OD) δ −162.0 (s), −162.3 (s). 31P NMR (121 MHz, CD3OD) δ 3.97 (s), 3.89 (s). MS (ESI) m/z calcd. for C25H36FN7O7P [M+H]+ 596.6; found 596.2.




embedded image


Example 5. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-isobutylamido-6-methylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (16)

Step 1. Preparation of Compound 13.


To a solution of compound 4 (286 mg, 0.92 mmol) and imidazole (370 mg, 5.43 mmol) in dry DMF (6 mL) at 0° C. was added 1,3-dichloro-1,1,3,3-tetraisopropyldisiloxane (300 μL, 0.94 mmol). The reaction mixture was stirred for 2 h at RT, diluted with EtOAc (50 mL) and the suspension was washed with saturated aq. NH4Cl solution and brine (40 mL each). The organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient PE/EtOAc 7:3 to 3:7) to afford product 13 (283 mg, 0.51 mmol, 56%) as a white solid. MS (ESI) m/z calcd. for C24H44FN6O4Si2 [M+H]+ 555.8; found 555.2.


Step 2. Preparation of compound 14.


To a solution of compound 13 (200 mg, 0.36 mmol) in dry pyridine (3 mL) at 0° C. was added isobutyryl chloride (38 μL, 0.36 mmol). The reaction mixture was stirred for 2 h at RT. The reaction was quenched by the addition of water (500 μL). The mixture was concentrated and co-evaporated with toluene (3×10 mL). The residue was purified by column chromatography (gradient PE/EtOAc 1:0 to 1:1) to afford product 14 (99 mg, 0.16 mmol, 44%) as a white solid. MS (ESI) m/z calcd. for C28H50FN6O5Si2 [M+H]+ 625.9; found 625.3.


Step 3. Preparation of (2R,3R,4R,5R)-5-(2-isobutylamido-6-methylamino-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (15)

To a solution of compound 14 (90 mg, 0.14 mmol) in dry THF (2 mL) was added tetrabutylammonium fluoride (1 M in THF, 38 μL, 0.38 mmol). The mixture was stirred for 2 h at RT and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 10:0 to 9:1) followed by reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to give product 15 (42 mg, 0.11 mmol, 77%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 8.31 (s, 1H), 6.29 (d, J=17.9 Hz, 1H), 4.70-4.60 (m, 1H), 4.07-3.98 (m, 2H), 3.89 (dd, J=12.5, 3.4 Hz, 1H), 3.10 (br s, 3H), 2.87 (br s, 1H), 1.23-1.16 (m, 9H). 19F NMR (282 MHz, CD3OD) δ −163.8. MS (ESI) m/z calcd. for C16H24FN6O4 [M+H]+ 383.4; found 383.2.


Step 4. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-isobutylamido-6-methylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (16)

To a solution of compound 15 (27 mg, 0.07 mmol) in dry THF (1 mL) at 0° C. was added t-butyl magnesium chloride (1.0 M in THF, 130 μL, 0.13 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (50 mg, 0.11 mmol) dissolved in dry THF (1 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min followed by 18 h at room temperature then quenched with a saturated aq. NH4Cl solution (2 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 95:5) then reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford product 16 (mixture of 2 diastereoisomers, 25 mg, 0.04 mmol, 54%) as a white solid. 1H NMR (300 MHz, CD3OD) δ 8.05 (s, 1H), 7.33-7.13 (m, 5H), 6.27 (d, J=18.6 Hz) and 6.21 (d, J=19.1 Hz, 1H), 5.10-4.95 (m, 1H), 4.93-4.78 (overlapped with H2O, m, 1H), 4.60-4.42 (m, 2H), 4.26-4.18 (m, 1H), 3.90-3.80 (m, 1H), 3.09 (br s, 3H), 2.84-2.80 (m, 1H), 1.33-1.15 (m, 18H). 31P NMR (121 MHz, CD3OD) δ 3.69 (s). 31P NMR (121 MHz, CD3OD) δ 4.11 (s), 3.99 (s). MS (ESI) m/z calcd. for C2H40FN7O8P [M+H]+ 652.6; found 652.3.




embedded image


Example 6. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-ethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (18)
Step 1. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-ethylamino)-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (17)

To a solution of compound 3 (150 mg, 0.29 mmol) in MeOH (4 mL) was added N-methylethylamine (245 μL, 2.90 mmol). The reaction mixture was heated at 100° C. in a sealed tube for 15 h, cooled down to room temperature and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford product 31 (89 mg, 0.26 mmol, 89%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 8.06 (s, 1H), 6.13 (d, J=18.0 Hz, 1H), 4.40 (dd, J=24.9, 8.7 Hz, 1H), 4.11-4.01 (m, 4H), 3.98-3.83 (m, 1H), 3.34 (br. s, 3H), 1.24-1.11 (m, 6H). 19F NMR (282 MHz, CD3OD) δ −163.7. MS (ESI) m/z calcd. for C14H22FN6O3 [M+H]+ 341.2; found 341.2.


Step 2. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-ethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (18)

To a solution of compound 17 (30 mg, 0.09 mmol) in dry THE (2 mL) at 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 110 μL, 0.11 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (48 mg, 0.11 mmol) dissolved in dry THE (1 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford the product 18 (mixture of 2 diastereoisomers, 22 mg, 0.04 mmol, 40%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.69 (m, 1H), 7.26-7.04 (m, 5H), 6.05 (d, J=18.6 Hz) and 6.03 (d, J=18.9 Hz, 1H), 4.86-4.79 (overlapped with H2O, m, 1H), 4.50-4.32 (m, 3H), 4.12-4.06 (m, 1H), 3.96-3.79 (m, 3H), 3.25 (br. s, 3H), 1.24-1.02 (m, 15H). 31P NMR (121 MHz, CD3OD) δ 4.07 (s), 4.00 (s). MS (ESI) m/z calcd. for C26H38FN7O7P [M+H]+ 609.3; found 609.2.




embedded image


Example 7. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-propylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (20)
Step 1. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-propylamino)-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (19)

To a solution of compound 3 (150 mg, 0.29 mmol) in MeOH (4 mL) was added N-methylpropylamine (295 μL, 2.90 mmol). The reaction mixture was heated at 100° C. in a sealed tube for 15 h, cooled down to room temperature and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) then reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford product 19 (80 mg, 0.23 mmol, 78%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 8.04 (s, 1H), 6.13 (d, J=18.3, 1H), 4.40 (dd, J=24.2, 9.2 Hz, 1H), m, 4.06-3.84 (m, 5H), 1.68 (sept, J=7.5 Hz, 2H), 1.15 (d, J=22.2 Hz, 3H), 0.93 (t, J=7.5 Hz, 3H). 19F NMR (282 MHz, CD3OD) δ −163.8. MS (ESI) m/z calcd. for C15H24FN6O3 [M+H]+ 355.2; found 355.2.


Step 2. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-propylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (20)

To a solution of compound 19 (30 mg, 0.09 mmol) in dry THE (2 mL) at 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 110 μL, 0.11 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (46 mg, 0.11 mmol) dissolved in dry THE (1 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford product 20 (mixture of 2 diastereoisomers, 22 mg, 0.03 mmol, 33%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.78, 7.77 (s+s, 1H), 7.37-7.13 (m, 5H), 6.15 (d, J=18.6 Hz) and 6.13 (d, J=18.9 Hz, 1H), 4.97-4.89 (overlapped with H2O, m, 1H), 4.63-4.30 (m, 3H), 4.22-4.14 (m, 1H), 4.02-3.84 (m, 2H), 1.74-1.63 (3H, m), 1.32-1.27 (m, 3H), 1.23-1.13 (m, 9H), 0.94 (t, J=7.4 Hz) and 0.93 (t, J=7.4 Hz, 3H). 31P NMR (121 MHz, CD3OD) δ 4.05 (s), 4.00 (s). MS (ESI) m/z calcd. for C27H40FN7O7P [M+H] 623.3; found 623.2.




embedded image


Example 8. Preparation of Isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-cyclobutylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (22)
Step 1. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-cyclobutylamino)-9H-purin-9-yl)-4-fluoro-2-(hydroxymethyl)-4-methyltetrahydrofuran-3-ol (21)

To a solution of compound 3 (150 mg, 0.29 mmol) in MeOH (4 mL) was added N-methylcyclobutylamine hydrochloride (105 mg, 0.90 mmol) and triethylamine (190 μL, 1.00 mmol). The reaction mixture was heated at 100° C. in a sealed tube for 15 h and cooled down to room temperature. An aqueous solution containing 30% NH4OH (1 mL) was added and the reaction mixture was heated at 100° C. in a sealed tube for 2 h, cooled down and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford product 21 (90 mg, 0.25 mmol, 86%) as a pale yellow solid.



1H NMR (300 MHz, CD3OD) δ 8.09 (s, 1H), 6.14 (d, J=18.0 Hz, 1H), 5.80-5.70 (m, 1H), 4.44-4.33 (m, 1H), 4.06-4.02 (m, 2H), 3.88-3.84 (m, 1H), 3.34 (s, 3H), 2.38-2.19 (m, 4H), 1.79-1.71 (m, 2H), 1.17 (d, J=22.2 Hz, 3H). 19F NMR (282 MHz, CD3OD) δ −163.8. MS (ESI) m/z calcd. for C16H24FN6O3 [M+H]+ 367.2; found 367.2.


Step 2. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methyl-cyclobutylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (22)

To a solution of compound 21 (50 mg, 0.14 mmol) in dry THE (2 mL) at 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 210 μL, 0.21 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (74 mg, 0.16 mmol) dissolved in dry THE (2 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) and then by reverse phase column chromatography (gradient H2O/MeOH 100:0 to 0:100) to afford product 22 (mixture of 2 diastereoisomers, 24 mg, 0.04 mmol, 28%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.79 (s, 0.2H), 7.77 (s, 0.8H), 7.38-7.12 (m, 5H), 6.18 (d, J=17.6 Hz) and 6.16 (d, J=17.5 Hz, 1H), 4.95-4.81 (m, 2H), 4.62-4.43 (m, 3H), 4.25-4.18 (m, 1H), 3.96-3.83 (m, 1H), 3.38 (s) and 3.36 (s, 3H), 2.38-2.21 (m, 4H), 1.75-1.63 (m, 2H), 1.32-1.16 (m, 12H). 31P NMR (121 MHz, CD3OD) δ 4.04 (s), 3.97 (s). MS (ESI) m/z calcd. for C28H40FN7O7P [M+H]+ 636.3; found 636.2.


Modification of the 2-Amino Moiety in the Active Compounds One of ordinary skill in the art can add a substituent to the 2-amino purine moiety by methods well known to those skilled in the art. One non-limiting process is provided here, and others can be easily adapted. ((2R,3R,4R,5R)-3-(benzoyloxy)-5-bromo-4-fluoro-4-methyltetrahydrofuran-2-yl)methyl benzoate, is treated with commercially available 2,6-dichloropurine, a base and a mixture of organic solvents at an elevated temperature to generate (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(benzoyloxymethyl)-4-fluoro-4-methyltetrahydrofuran-3-yl benzoate. In one embodiment, the base is potassium tert-butoxide. In one embodiment, the mixture of organic solvents comprises tert-butanol and acetonitrile. The compound, (2R,3R,4R,5R)-5-(2,6-dichloro-9H-purin-9-yl)-2-(benzoyloxymethyl)-4-fluoro-4-methyltetrahydrofuran-3-yl benzoate is treated with an amine, a base and an organic solvent at ambient temperature to generate 2-chloro-N6-substituted purines. In one embodiment, the amine is methylamine. In one embodiment, the base is triethylamine. In one embodiment, the organic solvent is ethanol. One skilled in the art will also recognize that upon treatment with an amine and base, the benzoate groups on the nucleoside will simultaneously be removed to generate the deprotected furanose moiety. 2-Chloro-N6-substituted purines can then be treated with an amine, and an organic solvent in a sealed tube at an elevated temperature of about 100° C. to generate N2,N6-disubstituted purine nucleosides of the present invention. In one embodiment, the amine is methylamine. In one embodiment, the organic solvent is ethanol. N2,N6-Disubstituted purine nucleosides of the present invention can be treated with a base, isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate and an organic solvent at a reduced temperature to generate compounds of Formula I-V. In one embodiment, the base is tert-butyl magnesium chloride. In one embodiment, the organic solvent is tetrahydrofuran.


Preparation of Stereospecific Phosphorus Enantiomers


Certain of the active compounds described herein have a chiral phosphorus moiety. Any of the active compounds described herein can be provided as an isolated phosphorus enantiomeric form, for example, at least 80, 90, 95 or 98% of the R or S enantiomer, using methods known to those of skill in the art. For example, there are a number of publications that describe how to obtain such compounds, including but not limited to column chromatography, for example as described in Example 17 below and U.S. Pat. Nos. 8,859,756; 8,642,756 and 8,333,309 to Ross, et al.


Example 9. Separation of the Stereoisomers of Compound 5

The stereoisomers of Compound 5 were separated on a Phenominex Luna column using the following conditions:


Column: Phenominex Luna 5 micron C18 (2) 250×10 mm part # OOG-4252-BO


Sample concentration: Approximately 50 mg/ml in acetonitrile


Injection volume: 50


Mobile phase A: HPLC grade water


Mobile phase B: HPLC grade acetonitrile.


Flow: 5 ml/min


UV: 283 nm


Gradient:
















Time
% B



















0
2



40
50



41
50



41.1
2



45
2










Run time: 45 minutes


Column Temperature: 40° C.


A sample chromatogram of a semi-prep run is illustrated in FIG. 1.


The combined fractions were evaluated using an analytical column with the following conditions:


Column: Phenominex Luna 5 micron C18 (2) 250×2 mm part # OOG-4252-BO


Injection volume: 10


Mobile phase A: HPLC grade water


Mobile phase B: HPLC grade acetonitrile.


Flow: 0.2 ml/min


UV: 283 nm


Gradient:
















Time
% B



















0
2



30
50



40
50



40.1
2



45
2










Run time: 45 minutes


Column Temperature: 40° C.


The combined fractions for each stereoisomer were evaporated to dryness using a rotovap with a bath temperature of 30° C. The resulting solids were dissolved in 1 ml of acetonitrile, transferred into 1.7 ml microcentrifuge tubes and the solvent evaporated on the vacuum centrifuge at a temperature of 30° C.


The data on the final samples are as follows:


1. First eluding peak: Compound 5 #1 (5-1) (21.7 mgs—97.8% ee).


2. Second eluding Peak: Compound 5 #2 (5-2) (13.2 mgs—95.9% ee).


The final weights of the 1st and 2nd peak correspond well to their percentages in the original mixture. (62.2% and 37.8% respectively).


Stereospecific Syntheses of Compounds of Formula I-VII




embedded image


Example 10. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(hydroxymethyl)-4-fluoro-4-methyltetrahydrofuran-3-ol (23)
Step 1. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(hydroxymethyl)-4-fluoro-4-methyltetrahydrofuran-3-ol (23)

The compound (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(benzoyloxymethyl)-4-fluoro-4-methyltetrahydrofuran-3-yl benzoate, 3, (80 g, 140 mmol) was added to a solution of trimethylamine in methanol (7 M, 800 mL) and stirred at RT overnight. The mixture was concentrated and then purified by column chromatography (DCM:MeOH=100:1) to afford (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(hydroxymethyl)-4-fluoro-4-methyl-tetrahydrofuran-3-ol (23) (40 g, 90%).




embedded image


Example 11. Preparation of ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate



embedded image


Step 1. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (4)

To a solution of (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(hydroxymethyl)-4-fluoro-4-methyl-tetrahydrofuran-3-ol (2.0 g, 1.0 eq) in dioxane (15 mL) was added MeNH2 aqueous solution (5.0 eq). After stirring overnight at RT, TLC showed that the starting material was consumed. The mixture was concentrated and purified by column chromatography (DCM:MeOH=40:1-30:1) to afford (2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol as a white powder (1.6 g, 81.6%). [M+H]+=313.5




embedded image


Step 2. Preparation of ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate

The compound (2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (1.47 g, 1.0 eq) and PPAL-S (2.35 g, 1.1 eq) were dissolved in anhydrous THE (29 mL). After cooling the mixture to −10° C., t-BuMgCl (5.8 mL, 1.7 M, 2.1 eq) was slowly added under a blanket of N2. After stirring at RT for 45 min, the mixture was quenched with aq. saturated NH4Cl, and extracted with EtOAc (20 mL×3). The combined organic layers were washed with water, brine (30 mL), dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography (DCM:MeOH=50:1-20:1) to afford ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyl-tetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate as a white powder (1.1 g, 40.3%).



1H NMR (400 MHz, CD3OD) δ 7.81 (s, 1H), 7.33-7.16 (m, 5H), 6.10 (d, J=18.4 Hz, 1H), 4.90-4.84 (m, 5H), 4.55-4.46 (m, 3H), 4.20-4.16 (m, 1H), 3.91-3.87 (m, 1H), 3.30 (m, 1H), 3.03 (s, 3H), 1.30-1.20 (m, 12H). [M+H]+=582.8.




embedded image


Example 12. Preparation of isopropyl ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (25)



embedded image


Step 1. Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol

To a solution of (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(hydroxymethyl)-4-fluoro-4-methyl-tetrahydrofuran-3-ol (2.8 g, 8 mmol) in dioxane (20 mL) was added dimethylamine aqueous solution (5 mL). After stirring at RT for 3 h, TLC showed that the starting material was consumed. The mixture was concentrated and purified by column chromatography (DCM:MeOH=60:1) to afford (2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (2.2 g).



1H NMR (400 MHz, CD3OD) δ 8.08 (s, 1H), 6.13 (d, J=18.0 Hz, 1H), 4.43 (dd, J=9.2, 9.2 Hz, 1H), 4.06 (d, J=10.8 Hz, 2H), 3.90 (m, 1H), 3.37 (s, 3H), 3.06 (s, 3H), 1.18 (d, J=22 Hz, 3H).




embedded image


Step 2. Preparation of isopropyl ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (25)

The compound (2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (8 g, 1.0 eq) and PPAL-S (11.1 g, 1 eq) were dissolved in anhydrous THF (100 mL). The mixture was cooled to −5-0° C. and t-BuMgCl (30.5 mL, 1.7 M, 2.1 eq) was slowly added under a N2 atmosphere. After stirring at RT for 2 h, the mixture was quenched with aq. saturated NH4Cl solution and extracted with EtOAc (70 mL×3). The combined organic layers were washed with water, brine (30 mL), dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography (DCM:MeOH=50:1) to afford isopropyl ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate as a white powder (9.5 g, 65%).



1H NMR (400 MHz, CD3OD) δ 7.81 (s, 1H), 7.35-7.19 (m, 5H), 6.15 (d, J=18.8 Hz, 1H), 4.90 (m, 1H), 4.54-4.49 (m, 3H), 4.22-4.19 (m, 1H), 3.90 (m, 1H), 3.43 (s, 3H), 1.32 (d, J=7.2 Hz, 3H), 1.24-1.17 (m, 9H). 31P NMR (160 MHz, CD3OD) δ 3.89.




embedded image


Example 13. Preparation of isopropyl ((((R)-(2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate (26)

The compound (2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (3 g, 1.0 eq) and PPAL-R (4.17 g, 1 eq) were dissolved in anhydrous THE (60 mL). The mixture was cooled to −5-0° C. and t-BuMgCl (11.4 mL, 1.7 M, 2.1 eq) was slowly added under a N2 atmosphere. After stirring at RT for 16 h, the mixture was quenched with aq. saturated NH4Cl solution and extracted with EtOAc (50 mL×3). The combined organic layers were washed with water, brine (30 mL), dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography (DCM:MeOH=50:1) to afford isopropyl ((((R)-(2R,3R,4R,5R)-5-(2-amino-6-(dimethylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate as a white powder (2.2 g, 41%).



1H NMR (400 MHz, CD3OD) δ 7.8 (s, 1H), 7.35-7.29 (m, 5H), 6.18 (d, J=18.8 Hz, 1H), 4.92 (m, 1H), 4.60 (m, 1H), 4.51-4.23 (m, 3H), 3.90 (m, 1H), 3.44 (s, 6H), 1.29 (d, J=6 Hz, 3H), 1.22-1.16 (m, 10H). 31P NMR (160 MHz, CD3OD) δ 3.98.




embedded image


Example 14. Preparation of isopropyl ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate



embedded image


Step 1: Preparation of (2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (8)

K2CO3 (53 g, 500 mmol) was added to N-methylcyclopropanamino hydrochloride in aqueous solution (100 mL). After stirring at RT for 10 min, a solution of (2R,3R,4R,5R)-5-(2-amino-6-chloro-9H-purin-9-yl)-2-(hydroxymethyl)-4-fluoro-4-methyl-tetrahydrofuran-3-ol (35 g, 109 mmol) in dioxane (300 mL) was added. The mixture was stirred at RT for 16 h and HPLC indicated that the reaction was complete. The mixture was concentrated and purified by column chromatography (DCM:MeOH=60:1) to afford (2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (30 g, 82%).



1H NMR (400 MHz, CD3OD) δ 8.16 (s, 1H), 6.17 (d, J=18.0 Hz, 1H), 4.41 (dd, J=9.2, 9.2 Hz, 1H), 4.06 (m, 2H), 3.90 (m, 1H), 3.37 (s, 3H), 3.16 (m, 1H), 1.18 (d, J=22.4 Hz, 3H), 0.94 (m, 2H), 0.74 (m, 2H). [M+H]+=353.2.




embedded image


Step 2: Preparation of isopropyl ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate

The compound (2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (8 g, 1.0 eq) and PPAL-S (10.3 g, 1 eq) were dissolved in anhydrous THF (100 mL). After cooling the mixture to −5-0° C., t-BuMgCl (28 mL, 1.7 M, 2.1 eq) was slowly added under a N2 atmosphere. The mixture was stirred at RT for 1 h, quenched with aq. saturated NH4Cl solution, and extracted with EtOAc (70 mL×3). The combined organic layers were washed with water, brine (30 mL), dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography (DCM:MeOH=100:1 to 50:1) to afford isopropyl ((((S)-(2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate as a white powder (9.5 g, 65%).



1H NMR (400 MHz, CD3OD) δ 7.86 (s, 1H), 7.35-7.19 (m, 5H), 6.17 (d, J=19.2 Hz, 1H), 4.91 (m, 1H), 4.52 (m, 3H), 4.21 (m, 1H), 3.93 (m, 1H), 3.35 (s, 3H), 3.16 (m, 1H), 2.0 (s, 1H), 1.26-1.16 (m, 12H), 0.93 (m, 2H), 0.73 (m, 2H). 31P NMR (160 MHz, CD3OD) δ 3.90




embedded image


Example 15. Preparation of isopropyl ((((R)-(2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate

The compound (2R,3R,4R,5R)-5-(2-amino-6-(methylcyclopropanamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-methyltetrahydrofuran-3-ol (3 g, 1.0 eq) and PPAL-R (2.8 g, 1 eq) were dissolved in anhydrous THF (60 mL). After cooling the mixture to −5-0° C., t-BuMgCl (7.6 mL, 1.7 M, 2.1 eq) was slowly added under N2. Then the mixture was stirred at RT for 1 h and quenched with aq. saturated NH4Cl solution, and extracted with EtOAc (50 mL×3). The combined organic layers were washed with water, brine (30 mL), dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography (DCM:MeOH=100:1 to 50:1) to afford the product as a white powder (3 g, 55%).



1H NMR (400 MHz, CD3OD) δ 7.81 (s, 1H), 7.30-7.25 (m, 5H), 6.16 (d, J=24.8 Hz, 1H), 4.84 (m, 1H), 4.84-4.50 (m, 3H), 4.22-4.19 (m, 1H), 3.88 (m, 1H), 3.33 (s, 3H), 3.14 (m, 1H), 2.0 (s, 1H), 1.28-1.13 (m, 12H), 0.92 (m, 2H), 0.90 (m, 2H). 31P NMR (160 MHz, CD3OD) δ 3.99.




embedded image


Example 16. Preparation of Compound 32



embedded image



Step 1. Preparation of Compound 29.


To a solution of 6 (3.0 g, 1.0 eq) in pyridine (30 mL) was added TIPDSCl2 (4.35 g, 1.5 eq) at 0° C. After stirring at RT for 4 h, TLC showed that starting material was consumed. The mixture was diluted with EtOAc, washed with 1M aq. HCl solution, saturated NaHCO3 aqueous solution, brine, dried over anhydrous Na2SO4 and concentrated to afford 29 as a yellow oil (6.3 g, 100%).




embedded image



Step 2. Preparation of Compound 30.


To a mixture of Compound 29 (800 mg, 1.0 eq), DMAP (16 mg, 0.1 eq), pyridine (1.6 mL) and DCM (10 mL) was added isobutyryl chloride (209 mg, 1.5 eq) at 0° C. After stirring at RT for 2 h, TLC showed that the starting material was consumed. The mixture was quenched with water, washed with aq. 1M HCl solution, saturated NaHCO3 aqueous solution, brine, dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography to afford the product, 30, as a white oil (563 mg, 62.3%).



1H NMR (400 MHz, CDCl3) δ 7.98 (s, 1H), 787 (s, 1H), 6.20 (d, J=16.0 Hz, 1H), 4.32-4.07 (m, 4H), 3.50 (s, 6H), 2.3 (m, 1H), 1.29-1.05 (m, 45H).




embedded image



Step 3. Preparation of Compound 31.


To a mixture of 30 (560 mg, 1.0 eq) in THE (10 mL) was added Et3N.3HF (706 mg, 5 eq) and Et3N (890 mg, 10 eq) at RT. After stirring at RT for 1.5 h, TLC showed that the starting material was consumed. The mixture was concentrated and purified by column chromatography to afford 31 as a white powder (288 mg, 83%).



1H NMR (400 MHz, CDCl3) δ 7.72 (s, 1H), 5.96 (d, J=44.0 Hz, 1H), 5.22 (m, 1H), 4.13-3.99 (m, 4H), 3.42 (s, 6H), 2.83-2.63 (m, 2H), 1.29-1.17 (m, 9H).




embedded image



Step 4. Preparation of Compound 32.


Compound 31 (280 mg, 1.0 eq) and PPAL-S (320 mg, 1 eq) were dissolved in anhydrous THE (10 mL). After cooling the mixture to −5° C., t-BuMgCl (0.87 mL, 1.7 M, 2.1 eq) was slowly added under a N2 atmosphere. The mixture was stirred at RT for 2 h, quenched with aq. saturated NH4Cl solution, and extracted with EtOAc (10 mL×3). The combined organic layers were washed with water, brine (20 mL), dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography to afford the product as a white powder (260 mg, 50%).



1H NMR (400 MHz, CD3OD) δ 7.98 (s, 1H), 7.25 (m, 5H), 6.23 (d, J=18.8 Hz, 1H), 4.52 (m, 3H), 4.38 (m, 1H), 3.81 (m, 1H), 3.75 (m, 1H), 3.48 (s, 6H), 2.81 (m, 1H), 1.32 (m, 18H). [M+H]+=666.9.




embedded image


Example 17. Preparation of Compound 35



embedded image



Step 1. Preparation of Compound 33.


To a mixture of 29 (2.0 g, 1.0 eq), DMAP (0.04 g, 0.1 eq), pyridine (4 mL) and DCM (20 mL) was added AcCl (0.414 g, 1.5 eq) at 0° C. After stirring at RT for 2 h, TLC showed that the starting material was consumed. The mixture was quenched with water, washed with aq. 1M HCl solution, saturated NaHCO3 aqueous solution then brine, dried over anhydrous Na2SO4 and concentrated. The crude product was purified by column chromatography to afford the product, 33, as a white oil (1.73 g, 80.8%).



1H NMR (400 MHz, CDCl3) δ 7.99 (s, 1H), 7.74 (s, 1H), 6.20 (d, J=20.0 Hz, 1H), 4.33-4.11 (m, 4H), 3.50 (s, 6H), 2.63 (s, 3H), 2.3 (m, 1H), 1.26-1.05 (m, 29H). [M+H]+=611.9.




embedded image



Step 2. Preparation of Compound 34.


To a mixture of 33 (1.58 g, 1.0 eq) in THE (20 mL) was added Et3N.3HF (2.1 g, 5 eq) and Et3N (2.6 g, 10 eq) at RT. After stirring at RT for 1.5 h, TLC showed that the starting material was consumed. The mixture was concentrated and purified by column chromatography to afford 34 as a white powder (782 mg, 82%). [M+H]+=369.6.




embedded image



Step 3. Preparation of Compound 35.


Compound 34 (136 mg, 1.0 eq) and PPAL-S (184 mg, 1.1 eq) were dissolved in anhydrous THE (3 mL). After cooling the mixture to −5° C., t-BuMgCl (0.5 mL, 1.7 M, 2.1 eq) was slowly added under a N2 atmosphere. The mixture was stirred at RT for 30 min, quenched with aq. saturated NH4Cl solution and extracted with EtOAc (10 mL×3). The combined organic layers were washed with water, brine (20 mL), dried over anhydrous and concentrated. The crude product was purified by column chromatography (DCM:MeOH=50:1-20:1) to afford the phosphoramidate 35 as a white powder (150 mg, 63.8%).



1H NMR (400 MHz, CD3OD) δ 7.81 (s, 1H), 7.35-7.16 (m, 5H), 6.10 (d, J=18.4 Hz, 1H), 4.87 (m, 1H), 4.52-4.46 (m, 3H), 4.21 (m, 1H), 3.91-3.87 (m, 1H), 3.03 (s, 3H), 1.30-1.13 (m, 12H). 31P NMR (160 MHz, CD3OD) δ 3.84. 19F NMR (376 MHz, CD3OD) δ −162.79.


Synthesis of β-D-2′-deoxy-2′-α-fluoro-2′-β-ethynyl-N6-substituted-2,6-diaminopurine nucleotides



embedded image


embedded image


Example 18. General Route to β-D-2′-deoxy-2′-α-fluoro-2′-β-ethynyl-N6-substituted-2,6-diaminopurine nucleotides



embedded image



Step 1. Preparation of Compound 36.


To a solution of 6-chloroguanosine (100 g, 332 mmol) in pyridine (400 mL) was added TPDSCl2 (110 mL, 1.05 eq.) dropwise at −5-5° C. under a N2 atmosphere. After stirring at that temperature for 2 h, TLC showed the starting material was consumed. DCM (600 mL) was added, and then TMSCl (85 mL, 2 eq.) was added dropwise at 0-5° C. After stirring at that temperature for 2 h, TLC showed the intermediate was consumed.


Isobutyryl chloride was added dropwise at 0-5° C. After stirring at that temperature for 2 h, TLC showed the intermediate was consumed. Water was added, and the content was extracted with DCM. The organic phase was then washed with 0.5 N HCl to remove pyridine.


After thepH of the content was washed to 5˜6, pTSA.H2O (9.2 g, 484.5 mmol) was added at 0-5° C. After stirring at that temperature for 1 h, TLC showed the intermediate was consumed. Water was then added, and the organic phase was washed with water, saturated aqueous NaHCO3 and brine. After being dried over Na2SO4, the solvent was removed in vacuo. The residue was then purified with column chromatography (PE/EA=100-10/1) to afford the product as a light yellow solid (82 g, 40%).



1H NMR (400 MHz, DMSO-d6) δ 10.88 (s, 1H), 8.55 (s, 1H), 5.91 (d, J=1.6 Hz, 1H), 5.53 (d, J=4.6 Hz, 1H), 4.72-4.58 (m, 2H), 4.16 (dd, J=12.4, 4.8 Hz, 1H), 4.00 (ddd, J=7.7, 4.8, 2.6 Hz, 1H), 3.93 (dd, J=12.4, 2.7 Hz, 1H), 2.78 (h, J=6.9 Hz, 1H), 1.26-1.12 (m, 3H), 1.10 (d, J=6.7 Hz, 6H), 1.09-0.88 (m, 24H).




embedded image



Step 2. Preparation of Compound 37.


To a solution of 36 (10.0 g, 16.3 mmol) in DCM (100 mL) was added Dess-Martin periodinane at rt and the reaction was stirred for 12 h. TLC showed the starting material was consumed. The reaction mixture was then diluted with DCM (200 mL) and washed with saturated aqueous Na2S203 and brine. The organic phase was then dried over Na2SO4 and concentrated to afford crude 37 as a light yellow solid (12 g). The crude 53 can be used directly in the next step without purification.




embedded image



Step 3. Preparation of Compound 38.


To a solution of ethynyltrimethylsilane (18.6 mL, 142.7 mmol) in THE (240 mL) was added n-BuLi (46 mL, 2.5 M, 115.0 mmol) dropwise at −15˜−20° C. under a N2 atmosphere. After stirring for 30 min, the reaction was cooled to −70° C., and 37 (crude, 16.3 mmol) in THE (60 mL) was added at that temperature. The content was then warmed to 0° C. TLC showed the starting material was consumed. Saturated aqueous NH4Cl was added, and the reaction was extracted with EA (100 mL) three times. The organic phase was combined and then washed with brine, then further dried over Na2SO4. After being concentrated in vacuo, the residue was purified by column chromatography (PE/EA=100→10/1) to afford a light yellow solid (6.0 g, 52%).




embedded image



Step 4. Preparation of Compound 39.


To a solution of 38 (6.0 g, 8.4 mmol) in DCM (240 mL) was added pyridine (4.2 mL, 52.9 mmol) under a N2 atmosphere. The reaction was cooled to −70° C., and DAST (12 mL, 90.4 mmol) was added. The content was then warmed to −30° C. TLC showed that the starting material was consumed. The reaction was poured into saturated aqueous NaHCO3, and then extracted with DCM (200 mL). The organic phase was washed with brine and dried over Na2SO4. After being concentrated in vacuo, the residue was purified with column chromatography (PE/EA=100→10/1) to afford a light yellow solid (3.8 g, 63%).




embedded image



Step 5. Preparation of Compound 40.


To a solution of 39 (3.8 g, 5.3 mmol) in THE (120 mL) was added AcOH (1.3 g, 22 mmol) and TBAF (4.2 g, 15.9 mmol) at rt. The reaction was stirred at rt for 30 min. TLC showed the starting material was consumed. After being concentrated in vacuo, the residue was purified with column chromatography (EA) to afford the product as a white solid (2.0 g, 95%).




embedded image



General Procedure for Amino Displacement and Deprotection:


To a solution of 40 (350 mg, 0.88 mmol) in dioxane (20 mL) was added the methanol or water solution of the corresponding amine (free base or salt as hydrochloride plus DIEA) at rt. The content was stirred at rt for 1-12 h. TLC showed the starting material was consumed. After being concentrated in vacuo, the residue was used directly in the next step without purification. The above mentioned residue was dissolved in methanol (10 mL). Aqueous NaOH (2.5 N, 10 mL) was added. After stirring overnight at rt, TLC showed that starting material was consumed. The pH of the content was adjusted to 7-8 with 1 N HC. The solution was concentrated and purified with column chromatography (DCM/MeOH=100→20/1) to afford the product as an off-white solid (yield: 40-80% over two steps). Table 1 illustrates the structures of compounds 57-63 and the corresponding mass spectral and 1H NMR for the respective compounds.











TABLE 1





Compound




No.
Structure

1H NMR/MS








41


embedded image



1H NMR (400 MHz, Methanol-d4) δ 8.05 (s, 1H), 6.27 (d, J = 16.9 Hz, 1H), 4.75 (dd, J = 21.7, 9.1 Hz, 1H), 4.06 (dd, J = 11.0, 2.4 Hz, 2H), 3.87 (dd, J = 13.1, 3.2 Hz, 1H), 3.42 (s, 6H), 3.37 (s, 2H), 3.18 (d, J = 5.4 Hz, 1H). [M + H]+ = 336.9






42


embedded image



1H NMR (400 MHz, DMSO-d6) δ 7.94 (s, 1H), 7.30 (s, 1H), 6.20-6.09 (m, 2H), 5.98 (s, 2H), 5.33 (t, J = 5.3 Hz, 1H), 4.57 (dt, J = 22.1, 8.0 Hz, 1H), 4.12 (q, J = 5.3 Hz, 1H), 3.91 (d, J = 9.3 Hz, 1H), 3.70 (t, J = 8.6 Hz, 1H), 3.36 (s, 1H), 3.18 (d, J = 5.2 Hz, 2H), 2.89 (d, J = 7.0 Hz, 3H). [M + H]+ = 323.0






43


embedded image



1H NMR (400 MHz, Methanol-d4) δ 8.11 (s, 1H), 6.29 (d, J = 16.9 Hz, 1H), 4.76 (dd, J = 21.7, 9.0 Hz, 1H), 4.10-4.01 (m, 2H), 3.87 (dd, J = 13.1, 3.1 Hz, 1H), 3.37 (s, 1H), 3.24-3.11 (m, 2H), 1.00-0.87 (m, 2H), 0.74 (td, J = 4.6, 2.8 Hz, 2H). [M + H]+ = 363.0






44


embedded image



1H NMR (400 MHz, Methanol-d4) δ 8.07 (s, 1H), 6.26 (d, J = 16.9 Hz, 1H), 4.76 (dd, J = 21.8, 9.3 Hz, 1H), 4.11-4.01 (m, 2H), 3.89 (d, J = 3.0 Hz, 1H), 3.89-3.75 (m, 1H), 3.37 (s, 2H), 3.21 (d, J = 5.4 Hz, 1H), 2.97-2.86 (m, 1H), 1.00- 0.77 (m, 2H), 0.67-0.46 (m, 2H). [M + H]+= 348.8










Example 19. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-dimethylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-ethynyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate



embedded image


Step 1. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-dimethylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-ethynyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate

To a solution of compound 41 (30 mg, 0.09 mmol) in dry THE (2 mL) at 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 125 μL, 0.13 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (49 mg, 0.11 mmol) dissolved in dry THE (2 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford the product (mixture of 2 diastereoisomers, 12 mg, 0.02 mmol, 24%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.79 (s, 0.45H), 7.77 (s, 0.55H), 7.36-7.14 (m, 5H), 6.28 (d, J=17.4 Hz) and 6.26 (d, J=17.5 Hz, 1H), 5.00-4.44 (m, 5H), 4.23-4.16 (m, 1H), 3.69-3.81 (m, 1H), 3.42 (bs, 3H), 3.40 (bs, 3H), 1.32-1.26 (m, 3H), 1.20-1.15 (m, 6H). 31P NMR (121 MHz, CD3OD) δ 4.04 (s), 3.98 (s). MS (ESI) m/z calcd. for C26H34FN7O7P [M+H]+ 606.2; found 606.2.


Example 20. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-methylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-ethynyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate



embedded image


Step 1. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-methylamino-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-ethynyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate

To a solution of compound 42 (30 mg, 0.09 mmol) in dry THE (2 mL) at 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 125 μL, 0.13 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (49 mg, 0.11 mmol) dissolved in dry THE (2 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford the product (mixture of 2 diastereoisomers, 9 mg, 0.02 mmol, 18%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.81, 7.79 (0.9 s+0.1 s, 1H), 7.36-7.14 (m, 5H), 6.26 (d, J=17.4 Hz, 0.1H) and 6.24 (d, J=17.4 Hz, 0.9H), 4.93-4.89 (overlapped with H2O, m, 1H), 4.80-4.78 (m, 1H), 4.53-4.49 (m, 2H), 4.21-4.18 (m, 1H), 3.95-3.84 (m, 1H), 3.23-3.20 (m, 1H), 3.04 (bs, 1H), 1.31-1.14 (m, 9H). 31P NMR (121 MHz, CD3OD) δ 4.06 (s), 3.97 (s). MS (ESI) m/z calcd. for C25H32FN7O7P [M+H]+ 592.2; found 592.2.


Example 21. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methylcyclopropylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-ethynyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate



embedded image


Step 1. Preparation of isopropyl ((((R,S)-(2R,3R,4R,5R)-5-(2-amino-6-(N-methylcyclopropylamino)-9H-purin-9-yl)-4-fluoro-3-hydroxy-4-ethynyltetrahydrofuran-2-yl)methoxy)-phenoxy-phosphoryl)-L-alaninate

To a solution of compound 43 (40 mg, 0.11 mmol) in dry THE (2 mL) at 0° C. was added tert-butyl magnesium chloride (1.0 M in THF, 160 μL, 0.16 mmol) dropwise over 10 min. The reaction mixture was stirred 15 min at 0° C. then another 15 min at room temperature. The reaction mixture was cooled down to 0° C. and a solution of isopropyl ((R,S)-(pentafluorophenoxy)-phenoxy-phosphoryl)-L-alaninate (55 mg, 0.12 mmol) dissolved in dry THE (2 mL) was added dropwise over 10 min. The reaction mixture was stirred at 0° C. for 30 min and 18 h at room temperature. The reaction was quenched with a saturated aq. NH4Cl solution (4 mL) and extracted with EtOAc (3×5 mL). The combined organics were dried over Na2SO4 and concentrated. The residue was purified by column chromatography (gradient DCM/MeOH 100:0 to 90:10) to afford the product (mixture of 2 diastereoisomers, 18 mg, 0.03 mmol, 26%) as a white solid.



1H NMR (300 MHz, CD3OD) δ 7.84, 7.82 (s+s, 1H), 7.35-7.14 (m, 5H), 6.30 (d, J=17.4 Hz) and 6.26 (d, J=17.6 Hz, 1H), 4.99-4.89 (overlapped with H2O, m, 1H), 4.82-4.69 (m, 1H), 4.59-4.46 (m, 2H), 4.21 (m, 1H), 3.96-3.82 (m, 1H), 3.24-3.22 (m, 1H), 3.17-3.11 (m, 1H) 1.31-1.26 (m, 3H), 1.20-1.15 (m, 6H), 0.93-0.89 (m, 2H), 0.75-0.68 (m, 2H). 31P NMR (121 MHz, CD3OD) δ 4.06 (s), 3.98 (s). MS (ESI) m/z calcd. for C28H36FN7O7P [M+H]+ 632.2; found 632.2.


Example 22. Preparation of PPAL-S



embedded image



Step 1. Preparation of Racemic PPAL


To a stirred solution of phenyl dichlorophosphate (250 g) in EtOAc (800 mL) was added isopropyl L-alaninate (200 g) in triethylamine (120 g) at −10° C. The reaction was stirred at −10° C. for 1 h. The compound 2,3,4,5,6-pentafluorophenol (220 g) in triethylamine (120 g) and EtOAc (400 mL) was added at −5° C. and stirred at that temperature for 0.5 h. The reaction mixture was allowed to warm to 25° C. and stirred at that temperature for 2 h. The solution was filtrated and washed with EtOAc (2×200 mL), and the combined organic phases were evaporated under vacuum to afford the solid PPAL-RS (racemate).


Step 2. Preparation of PPAL-RS


To a stirred solution of PPAL-RS in EtOAc (200 mL) and n-heptane (1.4 L) was added 2,3,4,5,6-pentafluorophenol (10.1 g) in triethylamine (6 g), and stirring was continued for about 4-8 h. After the R-isomer of the solid was less than 0.5%, the solid was filtered. The solid was dissolved in EtOAc (4 L), washed with water (2×100 mL), brine (1 L), dried over anhydrous Na2SO4, and filtered. The solvent was removed under vacuum to afford the PPAL-S (350 g). 1H NMR (400 MHz, DMSO-d6) δ=7.42-7.40 (m, 2H), 7.24-7.22 (m, 3H), 6.87 (dd, J=14.1, 9.9 Hz, 1H), 4.90-4.84 (m, 1H), 3.94-3.88 (m, 1H), 1.27 (dd, J=7.1, 1.1 Hz, 3H), 1.15 (dd, J=6.2, 1.2 Hz, 6H) ppm. 13P NMR (160 MHz, DMSO-d6) δ=0.37 ppm.


Example 23. Preparation of PPAL-R



embedded image


To a three-necked round bottom flask fitted with a mechanic stirrer were added phenyl dichlorophosphate (1896 g, 0.90 mol) and anhydrous EtOAc (750 mL). The solution was cooled to −10° C. under a nitrogen atmosphere. Iso-propyl L-alaninate (118 g, 0.90 mmol) and triethylamine (100 g, 1.1 eq) were added to the above solution. A pre-cooled (below 10° C.) mixture of 2,3,4,5,6-pentafluorophenol (165 g, 1 eq) and triethylamine (90.5 g, 1 eq) in EtOAc (300 mL) was added to the mixture via an addition funnel at −5° C. and the resulting mixture was stirred between 20-25° C. for 1 hour. The white precipitate (TEA.HCl) was filtered off and rinsed with EtOAc. The filtrate was concentrated under reduced pressure to yield PPAL-RS about 280 g (S/R=1/1) as a white solid PPAL-RS (280 g) was triturated in 300 mL of heptane/EtOAc (20:1) at room temperature for 5 min. The white suspension was filtered and the solid was rinsed with a mixture of heptane/EtOAc (20:1). The filtrate was cooled to 8° C. and the solid was collected by filtration, Crude PPAL-R (10 g) was obtained with 95% chiral purity. The crude product was purified following above step. PPAL-R (5 g) was obtained in NLT 98% chiral purity,



1H NMR (400 MHz, DMSO-d6) δ=7.43-7.39 (m, 2H), 7.27-7.22 (m, 3H), 6.87 (dd, J=14.1, 9.9 Hz, 1H), 4.89-4.85 (m, 1H), 3.95-3.90 (m, 1H), 1.27 (dd, J=7.1, 1.1 Hz, 3H), 1.14 (dd, J=6.2, 1.2 Hz, 6H). 13P NMR (160 MHz, DMSO-d6) δ=0.35.


Example 24: Preparation of Compound 52



embedded image


embedded image



Step 1. Preparation of Compound 49.


To a solution of 48 (1.81 g, 3.23 mmol) in dioxane (18 mL) was added 40 aqueous CH3NH2 solution (16.2 mmol). The reaction was stirred at 40° C. for 2 h. The mixture was concentrated, diluted with EtOAc (50 mL), washed with water and brine. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated to afford a white solid 49 (1.66 g, 92).




embedded image



Step 2. Preparation of Compound 50.


To a solution of 49(1.34 g, 2.42 mmol) and 1-methylimidazole (794 mg, 9.68 mmol) in DCM (14 mL) was slowly added pentyl chloroformate (547 mg, 3.63 mmol) at 0° C. The reaction was stirred at r.t overnight. The mixture was concentrated, and purified by column chromatography (PE:EtOAc=5:1-2:1) to afford 50 (1.01 g, 62%) as a white solid. 1HNMR (400 MHz, DMSO) δ 7.96 (s, 1H), 6.73 (s, 1H), 6.06-6.10 (d, J=16.0 Hz, 1H), 4.09-4.30 (m, 2H), 3.97-4.09 (m, 4H), 3.28 (s, 3H), 1.39-1.46 (m, 2H), 1.0-1.2 (m, 35H), 0.73-0.76 (t, J=8.0 Hz, 3H).




embedded image



Step 3. Preparation of Compound 51.


To a solution of 50 (1.00 g, 1.5 mmol) in THE (11 mL) was added Et3N (2.0 mL, 15 mmol) and Et3N.3HF (1.21 g, 7.5 mmol) at 0° C. The reaction was stirred at r.t for 1.5 h. The mixture was concentrated, and purified by column chromatography (MeOH:CH2Cl2=50:1) to afford 75 (460 mg, 72.2%) as a white powder.




embedded image



Step 4. Preparation of Compound 52.


To a solution of 51 (460 mg, 1.08 mmol) and PPAL-S (538 mg, 1.19 mmol) in anhydrous THE (9 mL) was slowly added t-BuMgCl (2.27 mmol) at 5-10° C. under N2. The reaction was stirred at r.t for 40 min. The mixture was quenched with aq. saturated NH4Cl solution, extracted with EtOAc, washed with aq. 5% K2CO3 solution and brine, dried over anhydrous Na2SO4, filtered and concentrated. The crude product was purified by column chromatography (CH2C2:MeOH=15:1) to afford 52 (280 mg, 37.3%) as a white solid.



1H NMR (400 MHz, DMSO) δ 8.12 (s, 1H), 7.34-7.38 (m, 2H), 7.18-7.23 (m, 3H), 6.74 (s, 2H), 6.11-6.16 (d, J=16.0 Hz, 1H), 5.99-6.05 (m, 1H), 5.84 (m, 1H), 4.77-4.81 (m, 1H), 4.30-4.41 (m, 3H), 4.03-4.11 (m, 3H), 3.78-3.80 (m, 1H), 3.3 (s, 3H), 1.44-1.51 (m, 2H), 1.00-1.21 (m, 16H), 0.76-0.80 (t, J=8.0 Hz, 3H). [M+H]+=696.6.


Example 25: Preparation of Compound 56



embedded image


embedded image



Step 1. Preparation of Compound 48.


To a solution of 23 (600 mg, 1 eq) in pyridine (30 mL) was added TIPDSCl2 (1.5 eq) at 0° C. The resulting solution was allowed to stand at room temperature for 2 h. The mixture was quenched with ice water and extracted with EtOAc. The organic layer was washed with 1M aq. HCl solution, saturated aqueous sodium bicarbonate and saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, and concentrated to yield the crude residue. The residue was purified by chromatography (MeOH:CH2Cl2=1:50) to afford 48 (998 mg, 94.4%) as a white solid foam.




embedded image



Step 2. Preparation of Compound 53.


A mixture of 48 (800 mg, 1 eq), pyridine (3.2 mL), DMAP (34.9 mg, 0.2 eq) in DCM (20 mL) was stirred at room temperature. N-amyl chloroformate (3.2 mL) was added dropwise at 0° C., and the mixture was stirred at room temperature for 1 day. The organic layer was washed with 1M aqueous HCl solution, saturated aqueous sodium bicarbonate and saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, and evaporated in vacuo. The residue was purified by chromatography on silica gel (MeOH:CH2Cl2=1:50) to afford 53 (255 mg, 26%) as a white solid foam.




embedded image



Step 3. Preparation of Compound 54.


To the solution of 53 (270 mg, 1 eq) in 1,4-dioxane (10 mL), was dropwise added 40% aqueous CH3NH2 solution (225.7 mg, 5 eq). The mixture was stirred for 2 h at room temperature and then concentrated in vacuo. The residue was chromatographed on silica gel (methanol:dichloromethane=1:40) to afford 54 (220 mg, 81.7%) as a white solid foam.




embedded image



Step 4. Preparation of Compound 55.


Triethylamine (1011.9 mg, 10 eq) and Et3N.3HF (806.05 mg, 5 eq) were added to an ice-cooled solution of 54 (668 mg, 1 eq) in THE (10 mL), the mixture was stirred for 2 h at room temperature. The mixture was concentrated and chromatographed on silica gel (MeOH:CH2C2=1:30) to afford 55 (492 mg, 84%) as a white solid foam.




embedded image



Step 5. Preparation of Compound 56.


To the mixture of 55 (113 mg, 1 eq) and PPAL-S (120 mg, 1 eq) in THE (4 mL) was dropwise added 1.7 M t-BuMgCl in THE (0.327 mL, 2.1 eq) at −10° C. The mixture was stirred at room temperature for 1 h, and then quenched with saturated aq. NH4Cl solution. The aqueous phase was extracted with EtOAc and the organic phase was washed with brine, dried and concentrated to obtain crude residue. The residue was subjected to flash chromatography to afford 56 (126 mg, 68.5%) as a white solid.



1H NMR (400 MHz, DMSO) δ 8.00 (s, 1H), 7.10-7.45 (m, 5H), 6.15-6.20 (d, J=20.0 Hz, 1H), 5.00-5.25 (s, 1H), 4.80-4.86 (m, 1H), 4.45-4.70 (m, 2H), 4.12-4.19 (m, 3H), 3.80-3.85 (m, 1H), 3.04 (s, 3H), 1.60-1.75 (m, 2H), 1.10-1.40 (m, 16H), 0.76-0.80 (t, J=8.0 Hz, 3H).



31P NMR (160 MHz, DMSO) δ 3.57. [M+H]+=696.5.


Example 26: Preparation of Compound 60



embedded image


embedded image



Step 1. Preparation of Compound 57.


To a solution of 6 (20 g, 1 eq) in CH3CN (100 mL) was added imidazole (16.6 g), TIPDSCl2 (28.9 g, 1.5 eq) in sequence at 5±5° C. The resulting solution was allowed to stand at room temperature for 4 h. The mixture was quenched with ice water and extracted with EtOAc. The organic layer was washed with water, saturated aqueous sodium bicarbonate and saturated aqueous sodium chloride, dried over anhydrous sodium sulfate, and concentrated to afford the crude residue (32 g).




embedded image



Step 2. Preparation of Compound 58.


To the solution of 57 (9.8 g, 1 eq) in TH (4 mL) was dropwise added 1.7 M t-BuMgCl in THE (50 mL, 4.8 eq) at 0-5° C. The mixture was stirred at room temperature for 0.5 h, and n-amyl chloroformate (2.7 g, 1.05 eq) was slowly added. The mixture was stirred at 0-5° C. for 3-4 h. The mixture was quenched with saturated aq. NH4Cl solution. The aqueous phase was extracted with EtOAc (200 mL) and the organic phase was washed with brine, dried and concentrated to obtain 58 (10.7 g) as oil.




embedded image



Step 3. Preparation of Compound 59.


Triethylamine (10.119 g) and Et3N.3HF (8.6 g, 5 eq) were added to an ice-cooled solution of 58 (7.3 g, 1 eq) in THE (100 mL) and the mixture was stirred for 1 h at room temperature. The mixture was concentrated and chromatographed on silica gel (MeOH:CH2Cl2=1:30) to afford 59 (4.3 g, 91%) as a white solid.




embedded image



Step 4. Preparation of Compound 60.


To the mixture of 59 (2 g, 1 eq) and PPAL-S (2.3 g, 1.1 eq) in TH (40 mL) was dropwise added 1.7 M t-BuMgCl in THE (5.6 mL, 2.1 eq) at −5° C. The mixture was stirred at −20±5° C. for 1 h, and then quenched with saturated aq. NH4Cl solution. The aqueous phase was extracted with EtOAc and the organic phase was washed with brine, dried and concentrated to obtain crude residue. The residue was subjected to flash chromatography to afford 60 (1.5 g, 47%) as a white solid.



1H NMR (400 MHz, CD3OD) δ 7.9 (s, 1H), 7.1˜7.2 (m, 5H), 6.2 (d, J=20 Hz, 1H), 5.1 (br, 1H), 4.84 (m, 1H), 4.49 (m, 2H), 4.16 (m, 1H), 4.13 (m, 2H), 3.86 (m, 1H), 3.45 (br, 6H), 1.70 (m, 2H), 1.26 (m, 4H), 1.20 (m, 6H), 1.14 (m, 6H), 0.93 (m, 3H). [M+H]+=710.5.


Biological Data


Example 27. Assay Methodology and Additional Biological Data

Huh-7 luc/neo ET cells bearing a discistronic HCV genotype 1b luciferase reporter replicon were plated at 7.5×103 cells/ml in duplicate 96-well plates for the parallel determination of antiviral efficacy (EC50) and cytotoxicity (TC50). The plates were cultured for 24 hours prior to the addition of compounds. Six serial one half log dilutions of the test articles (high test concentration of 100.0 μM or high test concentration of 1.0 M) and human interferon-alpha2b (high test 10.0 U/ml) were prepared in cell culture medium and added to the cultured cells in triplicate wells for each dilution. Six wells in the test plates received medium alone as an untreated control. Following 72 hours of culture in the presence of compound, one of the plates was used for the determination of cytotoxicity by staining with XTT and the other for antiviral efficacy by determination of luciferase reporter activity. Cytotoxicity and efficacy data were collected and imported into a customized Excel workbook for determination of the TC50 and EC50 values. Data for compounds of Formula I-VII are illustrated in Table 7 below. In addition, FIG. 2 illustrates the HCV replication inhibition curves for Compound 5-2 and Sofosbuvir. As can be seen in FIG. 2, Compound 5-2 has an EC50=4 nM, while Sofosbuvir has an EC50=53 nM. The y-axis is the percent of virus control and the x-axis is the concentration of drug in μM. FIG. 3 illustrates the HCV replication inhibition curves for Compound 25 and Sofosbuvir. Compound 25 has an EC50=4 nM and Sofosbuvir has an EC50=53 nM. The y-axis is the percent of virus control and the x-axis is the concentration of drug in M. FIG. 4 illustrates an intra-assay comparison of the anti-HCV activity for Compounds 5-2, 25, 27 and Sofosbuvir. The y-axis is the percent of virus control and the x-axis is the concentration of drug in M.


Various patient-derived HCV genotypes containing wild-type and resistance-associated variants were used to determine their relative replication sensitivity to test compounds. Replicon resistance test vectors (RTVs) containing the NS5B genomic regions were prepared using viral RNA isolated from plasma of HCV patients. Each NS5B region was amplified by reverse-transcription polymerase chain reaction and cloned into an HCV replicon RTV which was then transferred by electroporation into Huh-7 cells. After incubation in the absence and presence of serially diluted test compounds for 72-96 hr, viral replication was measured by luciferase activity and 50% inhibitory concentrations (IC50 values) were determined.


Table reports the IC50 and IC95 values for compound 25, 27, 5-2 and Sofosbuvir against various clinical isolates containing wild-type and resistance-associated variants.


All compounds were significantly more effective against HCV replication than sofosbuvir and neither 25, 27, nor 5-2 compound showed any evidence of cross-resistance to L159F, L159F and S282T, and C316N mutants.









TABLE 2







Antiviral Activity of Test Compounds


in Patient-derived HCV Genotypes


















Fold
Fold


HCV
NS5B
Test
IC50
IC95
Change in
Change in


Geno-
Mu-
Com-
Value
Value
IC50 from
IC95 from


type
tation
pound
(nM)
(nM)
Sofosbuvir
Sofosbuvir
















1a
none
sofosbuvir
62.7
507.7






25
4.4
31.3
14.2
16.2




27
4.2
26.4
15.0
19.3




5-2
10.5
60.8
6.0
8.4


1b
none
sofosbuvir
86.0
642.2
1.0





25
5.9
32.0
1.0
20.0




27
5.0
28.9
0.9
22.2




5-2
10.6
72.4
0.8
8.9


2a
none
sofosbuvir
22.5
195.1






25
2.7
22.2
8.4
8.8




27
2.9
16.2
7.9
12.0




5-2
6.2
45.4
3.6
4.3


2b
none
sofosbuvir
44.8
295.3






25
3.0
14.9
15.2
19.9




27
3.1
14.7
14.4
20.1




5-2
6.3
32.5
7.1
9.1


3a-1
none
sofosbuvir
125.9
689.8






25
5.1
27.8
24.5
24.8




27
4.4
25.4
28.4
27.2




5-2
11.8
59.3
10.7
11.6


3a-2
none
sofosbuvir
123.5
808.1






25
4.7
24.2
26.3
33.4




27
4.5
23.3
27.5
34.6




5-2
10.4
56.5
11.9
14.3


4a
none
sofosbuvir
74.9
681.4






25
4.6
33.0
16.2
20.7




27
3.6
38.1
20.7
17.9




5-2
9.9
74.4
7.5
9.2


4d
none
sofosbuvir
93.7
1019.7






25
5.9
44.2
16.0
23.1




27
5.6
38.4
16.7
26.6




5-2
14.0
79.9
6.7
12.8


1a
L159F
sofosbuvir
114.7
1067.5






25
5.2
40.4
22.0
26.4




27
5.1
36.2
22.3
29.5




5-2
13.0
95.3
8.8
11.2


1a
L159F
sofosbuvir
1619.9
16950.9





and
25
17.2
158.5
94.0
107.0



S282T
27
14.9
141.6
108.4
119.7




5-2
38.7
313.5
41.9
54.1


1b
C316N
sofosbuvir
73.9
472.8






25
3.2
18.1
23.1
26.2




27
3.1
16.5
23.5
28.7




5-2
7.7
42.7
9.6
11.1









A transient transfection assay was performed to determine the sensitivity of the wild type S282T mutant of HCV to test compounds. Huh-7 cells were electroporated in the presence of RNA transcribed from wild type or S282T HCV replicon plasmids from the T7 promoter. The transfected cells were seeded in to 96-well plates at 7.5×103 cells per well in Dulbecco's Modified Eagle's medium. After 24 hr of incubation, medium was removed and replaced with fresh medium containing no or various concentrations of test compounds. Following an additional 96-hr incubation, the anti-HCV activity was measured by luciferase endpoint with Britelite™ Plus luminescence reporter gene kit (Perkin Elmer, Shelton, Conn.). Duplicate plates were treated and incubated in parallel for assessment of cellular toxicity by staining with the tetrazolium dye XTT.


Table 3 reports the IC50 and IC95 values for compounds 25, 27, 5-2 and Sofosbuvir against HCV wild type and S282T replicons.


All compounds were significantly more effective against HCV replication than sofosbuvir and neither 25, 27, nor 5-2 compounds showed any evidence of cross-resistance to S282T variant.









TABLE 3







Antiviral Activity of Test Compounds


in a HCV Transient Infection Assay
















Fold
Fold




IC50
IC95
change in
change in



NS5B
Value
value
IC50 from
IC95 from


Compound
Mutation
(nM)
(nM)
Sofosbuvir
Sofosbuvir















5-2
None
1.4
9.98
26
22.2



S282T
2.8
20.6
99.3
>48.5


25
None
<1
2.7
>36.4
80.7



S282T
<1
9.4
>278
>106.4


27
None
<1
4.1
>36.4
53.2



S282T
<1
11.8
>278
>84.7


Sofosbuvir
None
36.4
218



S282T
278
>1000









The stability of selected compounds in fresh human whole blood and in human liver S9 fraction was determined in incubations containing 10 μM test compound. After incubations of 0, 30, 60 min, and up to 120 min, aliquots were removed and immediately extracted with 3 volumes of ice-cold methanol/acetonitrile (1:1, v/v). Extracts were centrifuged and supernatants were analyzed by LC-MS/MS for concentrations of unchanged test compound and potential metabolites.



FIG. 5 illustrates the excellent stability of compound 5-2 and all 2-amino derivatives in human blood.


Interestingly, FIG. 6 illustrates the in vitro time course dealkylation of the 2′-deoxy-2′-α-fluoro-2′-β-methyl-N2-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate to 2′-deoxy-2′-α-fluoro-2′-β-methyl-N6-methyl-2,6-diaminopurine nucleoside phosphoramidate with a human liver S9 fraction. Furthermore, unexpected, faster, and a more extensive rate of cleavage of the carbamate moiety by human liver S9 fraction was observed as compared to compound 5-2 and its other 2-amino derivatives (FIG. 7).


Example 28. HCV (gt1b) NS5B Polymerase Assay

Inhibition of HCV (gt1b) NS5B polymerase was determined in triplicate by measuring de novo polymerization in reaction mixtures containing serial dilutions of TA, in vitro transcribed viral RNA complementary to the HCV (−) strand 3′UTR region, polymerase, radiolabeled ribonucleotide, 250 μM non-competing rNTPs, and 1 μM competing rNTP. TA concentrations that produced 50% inhibition (IC50) were determined from resulting inhibition curves.


Example 29. Human Bone Marrow Progenitor Cell Assay

Fresh human bone marrow progenitor cells (Invitrogen) suspended in either BFU-E or GM-CSF-specific culture medium were added, at 105 cells/well, to triplicate serial dilutions of TA in 6-well plates. After 14-day incubations, colony counts were used to determine CC50 values. BFU-E colonies were confirmed using the benzidene technique.


Compounds 25, 27 and 5-2 show no cytotoxicity against bone marrow stem cells in vitro.


Example 30. iPS Cardiomyocyte Assay

iPS Cardiomyocytes (Cellular Dynamics) were seeded in microliter plates at 1.5×104 cells per well. After 48-hr incubation, cells were washed and maintenance medium containing serially diluted TA was added in triplicate. After incubating for an additional 3 days, cell viability was measured by staining with XTT and CC50 values were calculated.


Compounds 25, 27 and 5-2 show no cytotoxicity against iPS cardiomyocytes in vitro.


Example 31. Human DNA Polymerase Assays

Inhibition of human DNA polymerases α, β and γ (CHIMERx) was determined in triplicate in reaction mixtures of serially diluted TA, 0.05 mM dCTP, dTTP, and dATP, 10 μCi [32P]-α-dGTP (800 Ci/mmol), 20 μg activated calf thymus DNA and additional reagents specific for each polymerase. After 30-min incubations, incorporation of [α-32P]-GTP was measured and resulting incubation curves were used to calculate IC50 values.


The triphosphate, β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine triphosphate, as well as the triphosphate analogs of compounds 25, 27 and 5-2 do not inhibit human DNA polymerases α, β or γ.


Example 32. Human Hepatocyte Co-Cultures

Cytotoxicity and hepatocyte health were assessed in triplicate by measuring ALT leakage, urea production, albumin secretion and cellular ATP contents in micro-patterned human hepatocyte co-cultures (HepatoPac®, Hepregen Corporation) prepared by seeding cryopreserved female human hepatocytes (single donor) and 3T3 J2 mouse fibroblasts in microtiter plates according to procedures established by Hepregen. Culture media was replaced with fresh media containing TA, test article, (0, 1, 10 or 30 μM) every 2 or 3 days through day 16. Spent culture media was assayed for ALT and urea content on days 2, 5, 7, 9, 12, 16 and 21 and for albumin content on days 2, 5, 7 and 9. Cellular ATP levels were measured on days 9 and 21. ATP signals in stromal-only control cultures (murine 3T3 fibroblasts) were subtracted from those of human HepatoPac co-cultures to obtain hepatocyte-specific effects. See, Table 4, 5 and 6 below.


Compound 5-2 at concentrations up to 30 μM, showed no signs of cytotoxicity as measured by ALT leakage, albumin secretion, urea production and cellular ATP content when incubated for up to 12 days with micro-patterned co-cultured human hepatocytes. The minor indications of cytotoxicity detected with extended exposure (up to 21 days of culture) were significantly less than those observed with sofosbuvir. See, Table 4, 5 and 6 below.


INX-189 was highly cytotoxic to human co-cultured hepatocytes, showing decreased albumin secretion as early as day 2 and cytotoxicity by all measures. Sofosbuvir showed more cytotoxicity than AT-511 under the same conditions.









TABLE 4







Effect of Test Article on Cellular ATP Concentrations










50% Inhibitory Concentration (IC50) - μM












Test Article
Day 9
Day 21















Cmpd 5-2
>30
12.8



Sofosbuvir
8.6
2.3



INX-189
8.1
0.1

















TABLE 5







Effect of Test Articles on Albumin Secretion










50% Inhibitory Concentration (IC50) - μM














Test Article
Day 2
Day 5
Day 7
Day 9

















Cmpd 5-2
>30
>30
>30
>30



Sofosbuvir
>30
19.5
10.9
9.3



INX-189
13.6
3.1
3.2
2.4

















TABLE 6







Effect of Test Articles on Albumin Secretion








Test
50% Inhibitory Concentration (IC50)-μM














Article
Day 2
Day 5
Day 7
Day 9
Day 12
Day 16
Day 21

















Cmpd 5-2
>30
>30
>30
>30
>30
24.2
14.5


Sofosbuvir
>30
>30
>30
12.1
6.8
 2.7
 2.3


INX-189
>30
4.2
1.8
1.8
1.3
<<1  
<<1  









Example 33. Metabolic Studies

The metabolism of compounds 25, 27 and 5-2, at a concentration of 10 μM, were investigated in fresh primary cultures of human, dog and mouse hepatocytes. Plated hepatocytes from humans (XenoTech, mixed gender, pooled from 10 donors), male Beagle dog (BioreclamationIVT), and male ICR/CD-1 mice (BioreclamationIVT, 8 donors) in 6-well plates with matrigel overlay were incubated in singlet with 10 μM TA. After 2, 4, 6, 8 or 24 hr, intracellular levels of nucleotide prodrugs and their potential metabolites (prodrugs, monophosphates, triphosphates and nucleosides) were quantitated by LC-MS/MS. Concentrations below the lower limit of quantitation (1.5 μmol/106 cells for prodrugs, monophosphates and nucleosides and 12 μmol/106 cells for triphosphates) were extrapolated from the standard curves.


The compound β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine triphosphate is the predominant metabolite of compounds 25, 27 and 5-2 observed in cultured human hepatocytes and is a potent inhibitor of the HCV (gt1b) NS5B polymerase, with an IC50 of 0.15 μM.



FIG. 8 shows the predominant Compound 25 metabolites in human hepatocytes.



FIG. 9 shows the predominant Compound 27 metabolites in human hepatocytes.



FIG. 10 shows the predominant Compound 5-2 metabolites in human hepatocytes.



FIG. 11 illustrates the activation pathways for Compounds 25, 27 and 5-2. As can be seen, Compounds 25, 27 and 5-2 are converted to their corresponding monophosphate analogs which are subsequently metabolized to a common MP analog; β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine monophosphate (Compound 61). The monophosphate is then stepwise phosphorylated to the active triphosphate: β-D-2′-deoxy-2′-α-fluoro-2′-β-methyl-guanine triphosphate (Compound 62).


Example 34. Controls

INX-189 (INX-08189BMS-986094) and sofosbuvir were used as controls in the Examples above.


The two most potent nucleotide prodrugs, Compounds 25 and 27, demonstrated excellent selectivity, with CC50 values greater than 100 μM in Huh-7 cells, human bone marrow stem cells and human cardiomyocytes. No inhibition of human DNA polymerase, α, β or γ, no activity against other RNA or DNA viruses, and no toxicity in all host cell lines was observed at concentrations up to 100 μM.


Table 7 is a table illustrating the compounds tested in a HCV Replicon Assay along with the EC50/EC95 (μM) and CC50 (μM) results.









TABLE 7







Replicon Assay Results for Compounds Tested.















Fold






increase




HCV
HCV
in activity




Relicon
Replicon
compared


Cmpd

EC50/EC95
CC50
to parent


No.
Structure
(μM)
(μM)
nucleoside

















embedded image


6.7
>100









embedded image


 2.1/9.04
>100
3





 4


embedded image


15.7
>100






 5


embedded image


0.026/0.124
>100
>600





5-1


embedded image


0.0551/0.282 
>100
>280





5-2


embedded image


0.004/0.028
>100
>3,900





 6


embedded image


10.7
>100






 7


embedded image


0.0121/0.071 
>100
>890





 8


embedded image


5.56
>100






 9


embedded image


0.0091/0.054 
>100
>600





15


embedded image


>100
>100






16


embedded image


0.576/3.69 
>100






17


embedded image


11.5/65.4
>100






18


embedded image


0.048/0.219
90.0






19


embedded image


7.47
>100






20


embedded image


0.073/0.315
>100






25


embedded image


0.004/0.019
>100
>2,600





26


embedded image


0.0351/0.057 
>100






27


embedded image


0.005/0.025
>100
>1,100





28


embedded image


0.014/0.076
>100






41


embedded image


0.508/25.1 
21.8






42


embedded image


4.18/20.4
>100






43


embedded image


6.43/24.7
21.6






45


embedded image


 0.16/0.876
0.68






46


embedded image


0.224/0.961
>100






47


embedded image


0.338/1.72 
1.68






61


embedded image










62


embedded image













embedded image


0.052/0.310
>100









embedded image


0.045/0.259
>100









The β-D-2′-D-2′-α-fluoro-2′-β-C-substituted-2-modified-N6-substituted purine nucleotides described herein exhibit significant activity against the HCV virus. Compounds according to the present invention are assayed for desired relative activity using well-known and conventional assays found in the literature.


For example, anti-HCV activity and cytotoxicity of the compounds may be measured in the HCV subgenomic RNA replicon assay system in Huh7 ET cells. (See, Korba, et al., Antiviral Research 2008, 77, 56). The results can be summarized in comparison to a positive control, 2′-C-Me-cytosine {2′-C-Me-C}(Pierra, et al., Journal of Medicinal Chemistry 2006, 49, 6614.


Another in-vitro assay for anti-hepatitis C virus activity is described in U.S. Pat. No. 7,718,790 by Stuyver, et al., and assigned to Pharmasset, Inc.


This specification has been described with reference to embodiments of the invention. Given the teaching herein, one of ordinary skill in the art will be able to modify the invention for a desired purpose and such variations are considered within the scope of the invention.

Claims
  • 1. A compound of the formula
  • 2. A pharmaceutical composition comprising an effective therapeutic amount of a compound of claim 1 or a pharmaceutically acceptable salt thereof in a pharmaceutically acceptable carrier.
  • 3. The pharmaceutical composition of claim 2, in an oral dosage form.
  • 4. The pharmaceutical composition of claim 3, wherein the oral dosage form is a solid dosage form.
  • 5. The pharmaceutical composition of claim 4, wherein the solid dosage form is a tablet.
  • 6. The pharmaceutical composition of claim 4, wherein the solid dosage form is a capsule.
  • 7. The pharmaceutical composition of claim 3, wherein the oral dosage form is a liquid dosage form.
  • 8. The pharmaceutical composition of claim 7, wherein the liquid dosage form is a suspension or solution.
  • 9. The pharmaceutical composition of claim 2, in an intravenous formulation.
  • 10. The pharmaceutical composition of claim 2, in a parenteral formulation.
  • 11. A compound of the formula:
  • 12. The compound of claim 11, wherein the compound is at least 98% free of the opposite phosphorus S-enantiomer.
  • 13. The compound of claim 11, wherein the compound is at least 99% free of the opposite phosphorus S-enantiomer.
  • 14. A pharmaceutical composition comprising an effective therapeutic amount of a compound of the formula
  • 15. The pharmaceutical composition of claim 14, wherein the compound is at least 98% free of the opposite phosphorus S-enantiomer.
  • 16. The pharmaceutical composition of claim 14, wherein the compound is at least 99% free of the opposite phosphorus S-enantiomer.
  • 17. The pharmaceutical composition of claim 14, in an oral dosage form.
  • 18. The pharmaceutical composition of claim 17, wherein the oral dosage form is a solid dosage form.
  • 19. The pharmaceutical composition of claim 18, wherein the solid dosage form is a tablet.
  • 20. The pharmaceutical composition of claim 18, wherein the solid dosage form is a capsule.
  • 21. The pharmaceutical composition of claim 17, wherein the oral dosage form is a liquid dosage form.
  • 22. The pharmaceutical composition of claim 21, wherein the liquid dosage form is a suspension or solution.
  • 23. The pharmaceutical composition of claim 14, in an intravenous formulation.
  • 24. The pharmaceutical composition of claim 14, in a parenteral formulation.
PRIORITY

This application is a continuation of U.S. Ser. No. 16/900,397, filed Jun. 12, 2020, which is a continuation of U.S. Ser. No. 16/278,621 filed Feb. 18, 2019, which is a continuation U.S. Ser. No. 16/001,549 filed Jun. 6, 2018, now U.S. Pat. No. 10,239,911 which is a continuation of U.S. Ser. No. 15/782,628, now U.S. Pat. No. 10,000,523, filed Oct. 12, 2017 which is a continuation of U.S. Ser. No. 15/063,461, now U.S. Pat. No. 9,828,410, filed Mar. 7, 2016 which claims priority to U.S. Ser. No. 62/129,319 filed Mar. 6, 2015; U.S. Ser. No. 62/253,958 filed Nov. 11, 2015; and, U.S. Ser. No. 62/276,597 filed Jan. 8, 2016. Each of these references is incorporated herewith in their entirety.

US Referenced Citations (201)
Number Name Date Kind
5977061 Holy et al. Nov 1999 A
6348587 Schinazi et al. Feb 2002 B1
6660721 Devos et al. Dec 2003 B2
6777395 Bhat et al. Aug 2004 B2
6784166 Devos et al. Aug 2004 B2
6812219 LaColla et al. Nov 2004 B2
6908924 Watanabe et al. Jun 2005 B2
6911424 Schinazi et al. Jun 2005 B2
6914054 Sommadossi et al. Jul 2005 B2
6949522 Otto et al. Sep 2005 B2
7094770 Watanabe et al. Aug 2006 B2
7105493 Sommadossi et al. Sep 2006 B2
7105499 Carroll et al. Sep 2006 B2
7125855 Bhat et al. Oct 2006 B2
7138376 Gosselin et al. Nov 2006 B2
7148206 Sommadossi et al. Dec 2006 B2
7157441 Sommadossi et al. Jan 2007 B2
7163929 Sommadossi et al. Jan 2007 B2
7169766 Sommadossi et al. Jan 2007 B2
7192936 LaColla et al. Mar 2007 B2
7202224 Eldrup et al. Apr 2007 B2
7211570 Schinazi et al. May 2007 B2
7268119 Cook et al. Sep 2007 B2
7285658 Cook et al. Oct 2007 B2
7307065 Schinazi et al. Dec 2007 B2
7323449 Olsen et al. Jan 2008 B2
7339054 Xu et al. Mar 2008 B2
7365057 LaColla et al. Apr 2008 B2
7384924 LaColla et al. Jun 2008 B2
7388002 Babu et al. Jun 2008 B2
7429571 Chand et al. Sep 2008 B2
7429572 Clark Sep 2008 B2
7456155 Sommadossi et al. Nov 2008 B2
7495006 Liotta et al. Feb 2009 B2
7514410 Babu et al. Apr 2009 B2
7534767 Butora et al. May 2009 B2
7547704 LaColla et al. Jun 2009 B2
7560434 Babu et al. Jul 2009 B2
7560550 Doring et al. Jul 2009 B2
7582618 Sommadossi et al. Sep 2009 B2
7601820 Wang et al. Oct 2009 B2
7608597 Sommadossi et al. Oct 2009 B2
7608599 Klumpp et al. Oct 2009 B2
7608600 Storer et al. Oct 2009 B2
7608601 Devos et al. Oct 2009 B2
7625875 Gosselin et al. Dec 2009 B2
7632821 Butora et al. Dec 2009 B2
7635689 LaColla et al. Dec 2009 B2
7638502 Schinazi et al. Dec 2009 B2
7662798 LaColla et al. Feb 2010 B2
7662938 Schinazi et al. Feb 2010 B2
7713941 Cook et al. May 2010 B2
7718790 Stuyver et al. May 2010 B2
7772208 Schinazi et al. Aug 2010 B2
7824851 Sommadossi et al. Nov 2010 B2
7842672 Boojamra et al. Nov 2010 B2
RE42015 Watanabe et al. Dec 2010 E
7879815 MacCoss et al. Feb 2011 B2
7902202 Sommadossi et al. Mar 2011 B2
7919247 Stuyver et al. Apr 2011 B2
7932240 Dousson et al. Apr 2011 B2
7951789 Sommadossi et al. May 2011 B2
7964580 Sofia et al. Jun 2011 B2
7973013 Cho et al. Jul 2011 B2
7994139 Babu et al. Aug 2011 B2
8008264 Butler et al. Aug 2011 B2
8012941 Cho et al. Sep 2011 B2
8012942 Butler et al. Sep 2011 B2
8071567 Devos et al. Dec 2011 B2
8071568 Narjes et al. Dec 2011 B2
8093380 Wang et al. Jan 2012 B2
8114994 Liotta et al. Feb 2012 B2
8114997 Otto et al. Feb 2012 B2
8119607 Francom et al. Feb 2012 B2
8133870 Babu et al. Mar 2012 B2
8148349 Meppen et al. Apr 2012 B2
8163703 Babu et al. Apr 2012 B2
8168583 Schinazi et al. May 2012 B2
8173621 Du et al. May 2012 B2
8193372 Dousson et al. Jun 2012 B2
8242085 Babu et al. Aug 2012 B2
8299038 Sommadossi et al. Oct 2012 B2
8318682 Butler et al. Nov 2012 B2
8324179 Chen et al. Dec 2012 B2
8334270 Sofia et al. Dec 2012 B2
8343937 Sommadossi et al. Jan 2013 B2
8362068 Dousson et al. Jan 2013 B2
8399429 Jonckers et al. Mar 2013 B2
8415308 Cho et al. Apr 2013 B2
8415309 Francom et al. Apr 2013 B2
8415322 Clark Apr 2013 B2
8431588 Jonckers et al. Apr 2013 B2
8440813 Babu et al. May 2013 B2
8455451 Cho et al. Jun 2013 B2
8470834 Kwong et al. Jun 2013 B2
8481510 Jonckers et al. Jul 2013 B2
8481712 Bhat et al. Jul 2013 B2
8481713 Wang et al. Jul 2013 B2
8492539 Chun et al. Jul 2013 B2
8501699 Francom et al. Aug 2013 B2
8507460 Surleraux et al. Aug 2013 B2
8541434 Kwong et al. Sep 2013 B2
8551973 Bao et al. Oct 2013 B2
8552021 Jonckers et al. Oct 2013 B2
8563530 Chang et al. Oct 2013 B2
8575119 Wang et al. Nov 2013 B2
8580765 Sofia et al. Nov 2013 B2
8609627 Cho et al. Dec 2013 B2
8618076 Ross et al. Dec 2013 B2
8629263 Ross et al. Jan 2014 B2
8633309 Ross et al. Jan 2014 B2
8637475 Storer et al. Jan 2014 B1
8642756 Ross et al. Feb 2014 B2
8673926 Chu Mar 2014 B2
8674085 Sommadossi et al. Mar 2014 B2
8680071 Surleraux et al. Mar 2014 B2
8691788 Sommadossi et al. Apr 2014 B2
8697694 Arasappan et al. Apr 2014 B2
8715638 Kwong et al. May 2014 B2
8716262 Sofia et al. May 2014 B2
8716263 Chun et al. May 2014 B2
8735345 Porter et al. May 2014 B2
8735372 Du et al. May 2014 B2
8735569 Ross et al. May 2014 B2
8742101 Storer et al. Jun 2014 B2
8759318 Chamberlain et al. Jun 2014 B2
8759510 Du et al. Jun 2014 B2
8765710 Sofia et al. Jul 2014 B2
8772474 Beigelman et al. Jul 2014 B2
8802840 Francom et al. Aug 2014 B2
8815829 Schinazi et al. Aug 2014 B2
8816074 Chu et al. Aug 2014 B2
8841275 Du et al. Sep 2014 B2
8846638 Or et al. Sep 2014 B2
8846896 Serebryany et al. Sep 2014 B2
8853171 Butler et al. Oct 2014 B2
8859595 Coats et al. Oct 2014 B2
8859756 Ross et al. Oct 2014 B2
8871737 Smith et al. Oct 2014 B2
8871785 Boojamra et al. Oct 2014 B2
8877731 Beigelman et al. Nov 2014 B2
8877733 Cho et al. Nov 2014 B2
8889159 Cleary et al. Nov 2014 B2
8889701 Ivachtchenko et al. Nov 2014 B1
8895531 Shi Nov 2014 B2
8895723 Serebryany et al. Nov 2014 B2
8906880 Du et al. Dec 2014 B2
8912321 Axt et al. Dec 2014 B2
8921384 Chu Dec 2014 B2
8933052 Jonckers et al. Jan 2015 B2
8946244 Chu et al. Feb 2015 B2
8951985 Surleraux et al. Feb 2015 B2
8957045 Sofia et al. Feb 2015 B2
8957046 Du et al. Feb 2015 B2
8980865 Wang et al. Mar 2015 B2
9012427 Blatt et al. Apr 2015 B2
9012428 Jonckers et al. Apr 2015 B2
9045520 Chun et al. Jun 2015 B2
9061041 Girijavallabhan et al. Jun 2015 B2
9085573 Du et al. Jul 2015 B2
9085599 Or et al. Jul 2015 B2
9090642 Cho et al. Jul 2015 B2
9109001 Parsy et al. Aug 2015 B2
9139604 Boojamra et al. Sep 2015 B2
9156872 Girijavallabhan et al. Oct 2015 B2
9173893 Cho et al. Nov 2015 B2
9187515 Mayes et al. Nov 2015 B2
9211300 Mayes et al. Dec 2015 B2
9243025 Surleraux et al. Jan 2016 B2
9603863 Blatt et al. Mar 2017 B2
9603864 Blatt et al. Mar 2017 B2
9828410 Sommadossi Nov 2017 B2
10000523 Sommadossi Jun 2018 B2
10005811 Sommadossi Jun 2018 B2
10239911 Sommadossi Mar 2019 B2
10519186 Moussa Dec 2019 B2
20020045599 Arimilli et al. Apr 2002 A1
20040063658 Roberts et al. Apr 2004 A1
20040229839 Babu et al. Nov 2004 A1
20040259934 Olsen et al. Dec 2004 A1
20100249068 Beigelman et al. Sep 2010 A1
20100279969 Schinazi et al. Nov 2010 A1
20110257121 Chang et al. Oct 2011 A1
20120070411 Beigelman et al. Mar 2012 A1
20120135951 Schinazi et al. May 2012 A1
20130064794 Surleraux et al. Mar 2013 A1
20130225636 Roberts et al. Aug 2013 A1
20140038916 Wang et al. Feb 2014 A1
20140066395 Cho et al. Mar 2014 A1
20140212382 Schinazi et al. Jul 2014 A1
20140235566 Amblard et al. Aug 2014 A1
20150011481 Vilchez et al. Jan 2015 A1
20150011497 Beigelman et al. Jan 2015 A1
20150105341 Beigelman et al. Apr 2015 A1
20150150897 Denning et al. Jun 2015 A1
20160002281 Mayes et al. Jan 2016 A1
20160257706 Sommadossi et al. Sep 2016 A1
20160271162 Moussa et al. Sep 2016 A1
20190153017 Sommadossi et al. May 2019 A1
20190201433 Sommadossi et al. Jul 2019 A1
20200087339 Moussa et al. Mar 2020 A1
Foreign Referenced Citations (91)
Number Date Country
103435672 Dec 2013 CN
103980332 Aug 2014 CN
547008 Jun 1993 EP
398231 Jul 1997 EP
WO 199816184 Apr 1998 WO
WO 200190121 Nov 2001 WO
WO 200192282 Dec 2001 WO
WO 200232920 Apr 2002 WO
WO 2003039523 May 2003 WO
WO 2003062256 Jul 2003 WO
WO 2003093290 Nov 2003 WO
WO 2004002999 Jan 2004 WO
WO 2004003000 Jan 2004 WO
WO 2004014312 Feb 2004 WO
WO 2004052906 Jun 2004 WO
WO 2004091499 Oct 2004 WO
WO 2004106356 Dec 2004 WO
WO 2005000864 Jan 2005 WO
WO 2005021568 Mar 2005 WO
WO 2005084192 Sep 2005 WO
WO 2005090370 Sep 2005 WO
WO 2006012078 Feb 2006 WO
WO 2006063149 Jun 2006 WO
WO 2006063717 Jul 2006 WO
WO 2006094347 Sep 2006 WO
WO 2006102533 Sep 2006 WO
WO 2006121820 Nov 2006 WO
WO 2006130217 Dec 2006 WO
WO 2007022073 Feb 2007 WO
WO 2007112028 Oct 2007 WO
WO 2007130783 Nov 2007 WO
WO 2008012555 Jan 2008 WO
WO 2008048128 Apr 2008 WO
WO 2008062206 May 2008 WO
WO 2008095040 Oct 2008 WO
WO 2009001097 Dec 2008 WO
WO 2009003042 Dec 2008 WO
WO 2009067409 May 2009 WO
WO 2009086192 Jul 2009 WO
WO 2009086201 Jul 2009 WO
WO 2009129120 Oct 2009 WO
WO 2010081082 Jul 2010 WO
WO 2010091386 Aug 2010 WO
WO 2010108135 Sep 2010 WO
WO 2010145778 Dec 2010 WO
WO 2011005595 Jan 2011 WO
WO 2011005860 Jan 2011 WO
WO 2012041965 Apr 2012 WO
WO 2012048013 Apr 2012 WO
WO 2012092484 Jul 2012 WO
WO 2012125900 Sep 2012 WO
WO 2012154321 Nov 2012 WO
WO 2012158811 Nov 2012 WO
WO 2013009737 Jan 2013 WO
WO 2013019874 Feb 2013 WO
WO 2013039855 Mar 2013 WO
WO 2013039920 Mar 2013 WO
WO 2013044030 Mar 2013 WO
WO 2013090420 Jun 2013 WO
WO 2013096680 Jun 2013 WO
WO 2013142125 Sep 2013 WO
WO 2013142157 Sep 2013 WO
WO 2013142159 Sep 2013 WO
WO 2013151975 Oct 2013 WO
WO 2013177219 Nov 2013 WO
WO 2013187978 Dec 2013 WO
WO 2014008236 Jan 2014 WO
WO 2014047117 Mar 2014 WO
WO 2014052638 Apr 2014 WO
WO 2014063019 Apr 2014 WO
WO 2014076490 May 2014 WO
WO 2014082935 Jun 2014 WO
WO 2014100498 Jun 2014 WO
WO 2014100505 Jun 2014 WO
WO 2014120981 Aug 2014 WO
WO 2014124430 Aug 2014 WO
WO 2014137930 Sep 2014 WO
WO 2014169278 Oct 2014 WO
WO 2014169280 Oct 2014 WO
WO 2014209979 Dec 2014 WO
WO 2015038596 Mar 2015 WO
WO 2015053662 Apr 2015 WO
WO 2015081133 Jun 2015 WO
WO 2015095305 Jun 2015 WO
WO 2015158913 Oct 2015 WO
WO 2016041877 Mar 2016 WO
WO 2016100441 Jun 2016 WO
WO 2016100569 Jun 2016 WO
WO 2016144918 Sep 2016 WO
WO 2016145142 Sep 2016 WO
WO 2019200005 Oct 2019 WO
Non-Patent Literature Citations (37)
Entry
Ahmad, T. et al. “Cardiac dysfunction associated with a nucleotide polymerase inhibitor for treatment of hepatitis C” Hepatology 2015, 62, 409.
Berge, M.S. et al “Pharmaceutical Salts” Journal of Pharmaceutical Sciences, 1977, 66, 1.
Chang, W. et al. “Discovery of PSI-353661, a Novel Purine Nucleotide Prodrug for the Treatment of HCV Infection” ACS Med Chem Lett. 2011, 2, 130.
Cretton-Scott, E. et al. “In Vitro Antiviral Activity and Pharmacology of IDX184, a Novel and Potent Inhibitor of HCV Replication” (Abstract 588) J. Hepatol. 2008, 48, Supplement 2, S220.
Freeman et al. 2-amino-9(3-azido-2,3-dideoxy-β-D-erythro-pentofuranosyl)-6-Substituted-9H-Purines: Synthesis and Anti-HIV Activity. Bioorganic and Medicinal Chemistry, 1995; 3(4): 447-448.
Herman, B. et al. “Substrate mimicry: HIV-1 reverse transcriptase recognizes 6-modified-30-azido-20,30-dideoxyguanosine-50-triphosphates as adenosine analogs” Nucleic Acids Research 2012, 40, 381.
Huang et al. “Impact of solid state properties on developability assessment of drug candidates” Advanced Drug Delivery Reviews. 2004, 56, 321.
McGuigan, C. et al. “Design, synthesis and evaluation of a novel double pro-drug: INX-08189. A new clinical candidate for hepatitis C virus” Bioorganic & Medicinal Chemistry Letters 2010, 20, 4850.
McGuigan, C. et al. “Dual pro-drugs of 2′-C-methyl guanosine monophosphate as potent and selective inhibitors of hepatitis C virus” Bioorganic & Medicinal Chemistry Letters 2011, 21, 6007.
Murakami, E. et al. “Adenosine Deaminase-like Protein 1 (ADAL1): Characterization and Substrate Specificity in the Hydrolysis of N6- or O6-Substituted Purine or 2-Aminopurine Nucleoside Monophosphates” J Med Chem 2011, 54, 5902.
Poordad et al. “Daclatasvir with Sofosbuvir and Ribavirin for Hepatitis C Virus Infection with Advanced Cirrhosis or Post-Liver Transplantation Recurrence” Hepatology, 2016, 63, 1493.
Pradere, U. et al. “Synthesis of 5′-Methylene-Phosphonate Furanonucleoside Prodrugs: Application to D-2′-Deoxy-2′-α-fluoro-2′-β-C-methyl Nucleosides” Organic Letters 2012, 14, 4426.
Reddy, P. et al. “2′-Deoxy-2′-α-fluoro-2′-β-C-methyl 3′,5′-cyclic phosphate nucleotide prodrug analogs as inhibitors of HCV NS5B polymerase: Discovery of PSI-352938” Bioorganic & Medicinal Chemistry Letters 2010, 20, 7376.
Serajuddin, A.T.M “Salt formation to improve drug solubility” Advanced Drug Delivery Reviews, 2007, 59, 603.
Sofia, M.J. “Nucleotide Prodrugs for HCV Therapy,” Antiviral Chemistry & Chemotherapy, 2011; 22, 23.
Tao, S., Zhou, L., Zhang, H., Zhou, S., Amiralaei, S., Shelton, J.R., Coats, S.J., Schinazi, R.F.: Comparison of Three 2′-C-Methyl Guanosine Prodrugs for Hepatitis C including a Novel 2-D-2′-C-Me-2,6-Diaminopurine Ribonucleoside Phosphoramidate (RS-1389): Interspecies Hepatocyte and Human Cardiomyocyte Metabolism Profiles. The Liver Meeting 2014. Boston, MA, USA. Nov. 6-11, 2014.
Zhang et al. “Synthesis and evaluation of 30-azido-20,30-dideoxypurine nucleosides as inhibitors of human immunodeficiency virus” Bioorganic and Medicinal Chemistry Letters 2010, 20, 60.
Zhou, L. et al. “β-D-2′-C-Methyl-2,6-diaminopurine Ribonucleoside Phosphoramidates are Potent and Selective Inhibitors of Hepatitis C Virus (HCV) and Are Bioconverted Intracellularly to Bioactive 2,6-Diaminopurine and Guanosine 5′-Triphosphate Forms” J Med Chem 2015, 58, 3445.
PCT/US2016/021276 International Search Report and Written Opinion, dated Jun. 17, 2016.
Stahl et al., “Handbook of Pharmaceutical Salts Properties, Selection, and Use”, International Union of Pure and Applied Chemistry (IUPAC), 2002 (Chapters 6 and 7).
US, 2020-0308215, A1, U.S. Appl. No. 16/900,397, Jean-Pierre Sommadossi, et al., Oct. 1, 2020.
US, 2020-0331955, A1, U.S. Appl. No. 16/918,898, Jean-Pierre Sommadossi, et al., Oct. 22, 2020.
US, 2020-0331954, A1, U.S. Appl. No. 16/918,918, Adel Moussa et al., Oct. 22, 2020.
US, U.S. Appl. No. 17/017,443, Jean-Pierre Sommodossi, et al., filed Sep. 10, 2020.
US, U.S. Appl. No. 17/028,724, Jean-Pierre Sommodossi, et al., filed Sep. 22, 2020.
US, U.S. Appl. No. 17/065,149, Jean-Pierre Sommodossi, et al., filed Oct. 7, 2020.
US, U.S. Pat. No. 9,828,410, U.S. Appl. No. 15/063,461, Sommadossi et al., May 9, 1989.
US, U.S. Pat. No. 10,000,523, U.S. Appl. No. 15/782,628, Sommadossi et al., Jun. 19, 2018.
US, U.S. Pat. No. 10,005,811, U.S. Appl. No. 15/782,638, Sommadossi et al., Jun. 26, 2018.
US, U.S. Pat. No. 10,202,412, B2, U.S. Appl. No. 15/645,701, Sommadossi et al., Feb. 12, 2019.
US, U.S. Pat. No. 10,239,911, B2, U.S. Appl. No. 16/001,549, Sommadossi et al., Mar. 26, 2019.
US, U.S. Pat. No. 10,519,186, B2, U.S. Appl. No. 15/885,630, Mousa et al., Dec. 31, 2019.
US, 2019-0177356, A1, U.S. Appl. No. 16/278,621, Sommadossi et al., Jun. 13, 2019.
US, 2019-0201433, A1, U.S. Appl. No. 16/293,423, Sommadossi et al., Jul. 4, 2019.
US, 2020-0087339, A1, U.S. Appl. No. 16/687,136, Mousa et al., Mar. 19, 2020.
US, 2020-0179415, A1, U.S. Appl. No. 16/703,599, Sommadossi et al., Jun. 11, 2020.
US, 2020-0222442, A1, U.S. Appl. No. 16/821,850, Sommadossi et al., Jul. 16, 2020.
Related Publications (1)
Number Date Country
20200331956 A1 Oct 2020 US
Provisional Applications (3)
Number Date Country
62276597 Jan 2016 US
62253958 Nov 2015 US
62129319 Mar 2015 US
Continuations (5)
Number Date Country
Parent 16900397 Jun 2020 US
Child 16918914 US
Parent 16278621 Feb 2019 US
Child 16900397 US
Parent 16001549 Jun 2018 US
Child 16278621 US
Parent 15782628 Oct 2017 US
Child 16001549 US
Parent 15063461 Mar 2016 US
Child 15782628 US