US008080536B2

(12) United States Patent (10) Patent No.: US 8,080,536 B2 Erion et al. (45) Date of Patent: *Dec. 20, 2011

(54) PHOSPHORUS-CONTAINING PRODRUGS 4,952,575 A 8, 1990 Sauerbier et al. 4,952,740 A 8/1990 Juge et al. 5,034,394 A 7/1991 Daluge (75) Inventors: Mark D. Erion, Del Mar, CA (US); K. 5,047,533 A 9, 1991 Reist et al. Raja Reddy, San Diego, CA (US); 5,089,500 A 2/1992 Daluge Serge H. Boyer, San Diego, CA (US) 5,130.303 A 7, 1992 Akiyama et al. 5,153,183 A 10, 1992 Kawabe et al. 5,157,027 A 10, 1992 Biller et al. (73) Assignee: Metabasis Therapeutics, Inc., La Jolla, 5,204,355 A 4/1993 ZSadon et al. CA (US) 5,210,085 A 5/1993 Liotta et al. 5,212,304 A 5/1993 Fung et al. (*) Notice: Subject to any disclaimer, the term of this 5,240,946 A 8/1993 Kinney et al. 5,246,937 A 9, 1993 Hamden et al. patent is extended or adjusted under 35 5,258,538 A 1 1/1993 Fung et al. U.S.C. 154(b) by 0 days. 5,366,965 A 11/1994 Strein 5,480,875 A 1/1996 Isomura et al. This patent is Subject to a terminal dis 5,532,225 A 7/1996 Reist et al. claimer. 5,583,122 A 12/1996 Benedict et al. 5,658,889 A 8, 1997 Gruber et al. (21) Appl. No.: 12/889,889 5,663,159 A 9/1997 Starrett, Jr. et al. 5,681,590 A 10, 1997 Bechard et al. Filed: Sep. 24, 2010 5,721,219 A 2/1998 Ingallet al. (22) 5,814,639 A 9, 1998 Liotta et al. 5,840,716 A 11/1998 Ubasawa et al. (65) Prior Publication Data 5,869.467 A 2/1999 Holy et al. US 2011 FOOO9356A1 Jan. 13, 2011 (Continued) Related U.S. Application Data FOREIGN PATENT DOCUMENTS (60) Continuation of application No. 1 1/779.689, filed on CH 492 738 6, 1970 Jul. 18, 2007, now Pat. No. 7,816,345, which is a (Continued) continuation of application No. 1 1/601,843, filed on Nov. 20, 2006, now abandoned, which is a division of OTHER PUBLICATIONS application No. 09/518,501, filed on Mar. 3, 2000, now Alarcon, “Studies on the InVivo Formation of Acrolein: 3-Hydroxy Pat. No. 7,205,404. propylmercapturic Acid as an Index of (NSC Provisional application No. 60/153,127, filed on Sep. 26271) Activiation.” Cancer Treatment Reports 60(4):327-335, U.S. (60) National Cancer Institute (1976). 8, 1999, provisional application No. 60/123,013, filed Alexander et al., “Preparation of 9-(2-Phosphonomethoxyethyl) on Mar. 5, 1999. Adenine Esters as Potential Prodrugs.” Collect. Czech. Chem. Chem. Commun. 59:1853-1869, Nakladatelstvi Ceskoslovenski Akademie (51) Int. C. Ved. (1994). A6 IK3I/66 (2006.01) Amin et al., “1-Hydroxy-3-(methylpentylamino)-propylidene-1, C07F 9/02 (2006.01) 1-bisphosphnic Acid as a Potent Inhibitor of Squalene Synthase.” (52) U.S. Cl...... 514/110; 558/81 Arzneim-Forsch/Drug Res. 46(8):759-762 Editio Cantor (1996). (58) Field of Classification Search ...... 558/81; (Continued) 514f110 See application file for complete search history. Primary Examiner — James O. Wilson Assistant Examiner — Ebenezer O Sackey (56) References Cited (74) Attorney, Agent, or Firm — Arnold & Porter LLP U.S. PATENT DOCUMENTS (57) ABSTRACT 3,018,302 A 1/1962 Bielefeld et al. Novel cyclic phosphoramidate prodrugs of drugs of formula 3,404,178 A 10/1968 Roy I 3,796,700 A 3, 1974 Yoshioka et al. 4,255,566 A 3, 1981 Carrico et al. 4,318,982 A 3/1982 Hornby et al. 4,340,668 A 7/1982 Hornby et al. V 4,376,165 A 3/1983 Hornby et al. H 4,447,529 A 5/1984 Greenquist et al. O Y 4,621,077 A 11, 1986 Rosini et al. V / 4,659,825 A 4, 1987 Holy et al. 4,705,651 A 11, 1987 Staibano V 4,724,232 A 2f1988 Rideout et al. Y H 4,724,233 A 2/1988 De Clercq et al. W. W 4,777,163 A 10, 1988 Bosies et al. 4,804,655 A 2f1989 Miller et al. 4,808,716 A 2/1989 Holy et al. 4,861,759 A 8/1989 Mitsuya et al. their use in delivery of drugs to the liver, their use in enhanc 4,879,277 A 1 1/1989 Mitsuya et al. ing oral bioavailability, and their method of preparation are 4,882,142 A 11, 1989 Simon et al. described. 4,898,724 A 2f1990 Simon et al. 4,939,130 A 7, 1990 Jaeggiet al. 26 Claims, No Drawings US 8,080,536 B2 Page 2

U.S. PATENT DOCUMENTS Banker et al., “Modern Pharmaceutics, 3ed. Marcel Dekker, New 5,914,331 A 6, 1999 Liotta et al. York, 1996, pp. 451 and 596. 5,962.522 A 10, 1999 Wacher et al. Beaucage et al., The Synthesis of Modified Oligonucleotides by the 6,004,927 A 12/1999 Benet et al. Phosphoramidite Approach and Their Applications, Tetrahedron 6,028,054 A 2/2000 Benet et al. 49(28):6123-61.94 (1993). 6,054,587 A 4/2000 Reddy et al. Bedfordet al., “Synthesis of Water-Soluble Prodrugs of the Cytotoxic 6,110,903. A 8, 2000 Kasibhatla et al. Agent Combretastatin A4.” Bioorg. Med. Chem. Lett. 6(2): 157-160 6,117.873 A 9, 2000 Acklin et al. (1996). 6,284,748 B1 9/2001 Dang et al. Beilstein Registry No. 3635189, Beilstein Institut zur Foerderung der 6,294,672 B1 9/2001 Reddy et al. Chemischen Wissenschatten (1991). 6,312,662 B1 1 1/2001 Erion et al. 6,399,782 B1 6/2002 Kasibhatla et al. Bentrude et al. Am. Chem. Soc., 108(21):6669-6675 (1986). 6,489.476 B1 12/2002 Dang et al. Bentrude et al., J. Am. Chem. Soc., 110(20:71 19-7127 (1988). 6,752,981 B1 6, 2004 Erion et al. Bentrude et al., “Stereo- and Reglochemistries of the Oxidations of 6,756,360 B1 6, 2004 Erion et al. 2-Methoxy-5-tert-butyl-1,3,2-dioxaphosphorinanes and the Cyclic 6,946,115 B2 9, 2005 Erion et al. Methyl 3'5'-Phosphite of Thymidine by HO/1 and O/AIBN to 7,205,404 B1 4/2007 Erion et al. P-Chiral Phosphates. ONMRAssignment of Phosphorus Configu 7,303,739 B2 12/2007 Erion et al. ration to the Diasteromeric Thymidine Cyclic Methyl 3'5'- Monophosphates.” J. Am. Chem. Soc. 111:3981-3987 (1989). FOREIGN PATENT DOCUMENTS Berry et al., “High-Yield Preparation of Isolated Rat Liver Parenchy DE 35 12781 A1 4f1985 mal Cells,” J. of Cell Biology 43:506-520 (1969). EP OOO2 062 A1 5, 1979 Bespalov et al., “Prolongation of Morphine Analgesia by Competi EP O 072987 A1 8, 1982 tive NMDA Receptor Antagonist D-CPPene (SDZ EAA 494) in EP O O72 531 A1 2, 1983 Rats.” Eur: J. Pharmacol. 351:299-305 (1998). EP O 158 057 A 10, 1985 Bijsterbosch et al., “Disposition of the Acyclic Nucleoside EP O 161955 A1 11, 1985 Phsophonate (S)-9-(3-Hydroxy-2-Phosphonylmethoxypropyl) EP O 261 283 A1 9, 1986 EP 0 180276 A1 12, 1988 Adenine.” Antimicrobial Agents and , 42:1146 EP O 338 372 A2 10, 1989 1150(1998). EP O 353 692 A2 7, 1990 Bijsterbosch et al., “Disposition of the Acyclic Nucleoside EP O 481 214 B1 4f1992 Phosphonate (S)9- (3-Hydroxy-2- EP 1 165570 B1 1, 2002 Phosphonylmethoxyproply)Adenine.” Antimicrobial Agents and GB 987378 9, 1970 Chemotherapy 42:1146-1150 (1998). JP 62-195392 A2 8, 1987 Bird et al., “Synthesis of Novel N-Phosphonoalkyl Dipeptide Inhibi JP 62.249.996 A2 10, 1987 tors of Human Collagenase.” J. Med. Chem. 37:158-169 (1994). WO WO 90,08155 A1 7, 1990 Boddy et al., “Individual Variation in the Activation and Inactivation WO WO 90,10636 A1 9, 1990 of Metabolic Pathways of Cyclophosphamide.” J. National Cancer WO WO91, 19721 A1 12, 1991 WO 96.O1267 1, 1996 Institute 84(22):744-748 (1992). WO 97.03679 2, 1997 Borch et al., “The Mechanism of Activation of WO WO 97.22614 A1 6, 1997 4-Hydroxycyclophosphamide.” J. Med. Chem. 30:427-431 (1987). WO 98,09668 3, 1998 Borch et al., “Synthesis and Antitumor Properties of Activated WO 98.39342 9, 1998 Cyclophosphamide Analogues.” J. Med. Chem. 34:3044-3052 WO 98.39343 9, 1998 (1991). WO 98.39344 9, 1998 Borch et al., “Synthesis, Activation and Cytotoxicity of WO WO99/45O16 9, 1999 Aldophosphamide Analogues.”.J. Med Chem. 34:3052-3058 (1991). WO WOO1/39724 A2 6, 2001 Boydet al., “Synthesis and Antitumor Activity of Cyclophosphamide Analogues. 3. Preparation, Molecular Structure Determination, and OTHER PUBLICATIONS Anticancer Screening of Racemic cis- and trans-4- Anderson et al., “Cyclophosphamide and Phenylcydophosphamide.” J. Med. Chem. 23:372-375 (1980). 4-Hydroxycyclophosphamide? Aldophosphamide Kinetics in Brain et al., “Modulation of P450-Dependent Patients Receiving High-Dose Cyclophosphamide Chemotherapy.” Pharmacokinetics: a Better Understanding of Drug Activation. In Clinical Cancer Research 2: 1481-1487 (1996). Vivo.” British.J. of Cancer 77(11):1768-1776 (1998). Annaert et al., “Transport, Uptake, and Metabolism of the Brenna et al., "Affinity-Chromatography Purification of Alkaline Bis(pivaloyloxymethyl)-Ester Prodrug of 9-(2- Phosphatase from Calf Intestine.” Biochem. J. 151:291-296 (1975). Phosphonylmethoxyethyl) Adenine in an In Vitro Cell Culture Sys Brill et al., “Arbuzov-like Dealkylation Reactions of Transition tem of the Intestinal Mucosa (Caco-2).” Pharm. Res. 14(4):492-496 Metal-Phosphite Complexes.” Chem. Rev. 84:577-585 (1984). (1997). Brocket al., “Acrolein, the Causative Factor of Urotoxic Side-effects Arnér et al., Mammalian Deoxyribonculeoside Kinases, Pharmac. of Cyclophosphamide, Ifosfamide, and Sufosfamide.” Ther: 67(2): 155-186 (1995). Arzneimittel Forschung Drug Research 29(4):659-661 (1979). Arnold et al., “Uber Beziehungen zwischen chemischer Konstitution Campagne et al., “Synthesis of Mixed Phosphonate Diester Ana and cancerotoxischer Wirkung in der Reihe der Phosphamidester des logues of Dipeptides using BOP or PyBOP Reagents.” Tetrahedron Lett. 34(42):6743-6744 (1993). Bis-(B-chlorathyl)-amins.” Arzelimittel Forschung. Drug Research Campbell, “The Synthesis of Phosphonate Esters, an Extension of the 11:143-158, ECV Editio Cantor Verlag, Aulendorf, Germany (1961). Mitsunobu Reaction.” J. Org. Chem. 57:6331-6335 (1992). Atiq et al., “Treatment of Unresectable Primary Liver Cancer with Casara et al. “Synthesis of Acid Stable 5'-O-Fluoromethyl Introhepatic Fluorodeoxyuridine and Through an Phosphonates of Nucleosides. Evaluation as Inhibitors of Reverse Implantable Pump.” Cancer 69(4):920-924 (1992). Transcriptase.” Bioorg. Med...Chem. Lett. 2(2): 145-148 (1992). Auberson et al., “N-Phosphonoalkyl-5-Aminomethylcquinoxaline-2, Casteel et al., “Steric and Electronic Effects in the Aryl Phosphate to 3-Diones: InVivo Active AMPA and NMDA-(Glycine) Antagonists.” Arylphosphonate Rearrangement.” Synthesis 691-693 (1991). Bioorg. Med. Chem. Lett. 9:249-254 (1999). Chang et al., “Enhanced Cyclophosphamide and Ifosfamide Activa Baker et al., “Microtiter Plate Assay for the Measurement of tion in Primary Human Hepatocyte Cultures: Response to Glutathione and Glutathione Disulfide in Large Numbers of Biologi Cytochrome P-450 Inducers and Autoinduction by cal Samples.” Annal. Biochem. 190:360-365 (1990). Oxazaphosphorines.” Cancer Research 57:1946-1954 (1997). Balthazor et al., “Nickel-Catalyzed Arbuzov Reaction: Mechanistic Chen et al., “Intratumoral Activation and Enhanced Chemotherapeu Observations.” J. Org. Chem. 45:5425-5426 (1980). tic Effect of Oxazaphosphorines following Cytochrome P-450 Gene US 8,080,536 B2 Page 3

Transfer: Development of a Combined Chemotherapy/Cancer Gene Enriquez et al., “Conjugation of Adenine Arabinoside Therapy Strategy.” Cancer Research 55:581-589 (1995). 5'-Monophosphate to Arabinogalactan: Synthesis, Characterization, Chen et al., “Sensitization of Human Breast Cancer Cells to and Antiviral Activity.” Bioconjugate Chem. 6: 195-202 (1995). Cyclophosphamide and Ifosfamide by Transfer of a Liver Erion et al., “Design, Synthesis, and Characterization of a Series of Cytochrome P450 Gene.” Cancer Research 56:1331-1340 (1996). Cytochrome Paso 3A-Activated Prodrugs (HepDirect Prodrugs) Use Clarket al., “Oxidative Metabolism of Cyclophosphamide: Identifi ful for Targeting Phosph(on)ate-Based Drugs to the Liver.” J. Am. cation of the Hepatic Monooxygenase Catalysts of Drug Activation.” Chem. Soc. 126:5154-5183 (2004). Cancer Research 49:2344-2350 (1989). Erion et al., “Liver-Targeted Drug Delivery Using HepDirect Cooper et al., “Use of Carbohydrate Derivatives for Studies of Phos Prodrugs.” J. of Pharmacology & Experimental Therapeutics phorus Stereo-Chemistry. Part II. Synthesis and Confugrational 312(2):554-560 (2005). Erion et al., “HepDirect Prodrugs for Targeting Nucleoside-Based Assignments of 1, -3, 2-Oxathiaphosphorinan-2-ones and 1, 3, Antiviral Drugs to the Liver.” Current Opinion in Investigational 2-Dioxaphosphorinan-2-thiones,” J. Chem. Soc. Perk. Trans. I. Drugs 7(2):109-117 (2006). 10: 1049-1052 (1974). Evans “Chemistry 206 Advanced Organic Chemistry.” Harvard Uni Coppi et al., “Lewis Acid Mediated Condensation of Alkenols and versity, online Sep. 11, 2003, retrieved on Jan. 19, 2005). Retrieved Aldehydes. A Selective Synthesis of Tetrahydropyrians and from the internet, . Database Beilstein Registry No. 1028505, Beilstein Institute for Farquhar et al., “5'-'4-(Pivaloyloxy)-1, 3, 2-dioxaphosphorin an Organic Chemistry, Frankfurt, Germany, Nov. 29, 1988 (2 pages). -2yl-2'-deoxy-5-fluorouridine: a membrane-permeating prodrug of Database Beilstein Registry No. 1083232, Beilstein Institute for 5-fluoro-2'-deoxyuridylic acid (FDUMP).” J. Med. Chem. 38:488 Organic Chemistry, Frankfurt, Germany, Nov. 29, 1988 (1 page). 495 (1995). Database Beilstein Registry No. 1085700, Beilstein Institute for Farquhar et al., “Biologically-Cleavable Phosphate Protective Organic Chemistry, Frankfurt, Germany, Nov. 29, 1988 (1 page). Groups: 4-Acyloxy-1,3,2-Dioxaphosphorinanes as Neutral Latent Database Beilstein Registry No. 6530655, Beilstein Institute for Precursors of Dianionic Phosphates.” Tetrahedron Lett. 36(5):655 Organic Chemistry, Frankfurt, Germany, Apr. 18, 1994 (2 pages). 658 (1995). Davis et al., “Effect of Withania somnifera on Cyclosphosphamide Farquhar et al., “Biologically Reversible Phosphate-Protective induced Urotoxicity.” Cancer Letters 148:9-17 (2000). Groups.” J. Pharm. Sci. 72(3):324-325 (1983). Dearfield et al., “Analysis of the Genotoxicity of Nine Acrylate? Farquhar et al., “Synthesis and Biological Evaluation of 9-5'-(2- Methacrylate Compounds in L5178Y Mouse Lymphoma Cells.” Oxo-1,3,2- oxazaphosphorinan-2-yl)-3-D-arabinosyladenine and Mutagenesis 4:381-393 (1989). 9-5'-(2-Oxo-1,3,2- dioxazaphosphorinan-2-yl)-3-D- Dechant et al., “Ifosfamide/Mesna. A Review of Its Antineoplastic arabinosylladenine: Potential Neutral Precursors of 9- B-D- Activity, Pharmacokinetic Properties and Therapeutic Efficacy in Arabinofuranosylladenine 5'-Monophosphate.” J. Med. Chem. Cancer.” Drugs 42(3):428-467 (1991). 28: 1358-1361 (1985). De Clercq et al., “A Novel Selective Broad-spectrum Anti-DNAVirus Farquhar et al., "Synthesis and Biological Evaluation of Neutral Agent.” Nature 323:464-467 (1986). Derivatives of 5-Fluoro-2'-deoxyuridine 5'-Phosphate.” J. Med. Deleve et al., "Cellular Target of Cyclophosphamide Toxicity in the Chem. 26: 1153-1158 (1983). Murine Liver: Role of Glutathione and Site of Metabolic Activation.” Farquhar et al., “Synthesis and Antibumor Evaluation of Hepatology 24(4): 830-837 (1996). Bispivaloyloxy)methyl 2'-Deoxy-5-fluorouridine DeLombaert et al., “N-Phosphonomethyl Dipeptides and Their 5'-Monophosphate (FdUMP): A Strategy to Introduce Nucleotides Phosphonate Prodrugs, a New Generation of Neutral Endoprptidase into Cells,” J. Med Chem. 37:3902-3909 (1994). (NEP, EC 3.4.24.11). Inhibitors.” J. Med. Chem. 37:498-511 (1994). Fiume et al., “Inhibition of Hepatitis B Virus Replication by Delombaert et al., “Pharmacological Profile of a Non-Peptidic Dual Vidarbine Monophosphate Conjugated with Lactosaminated Serum Inhibitor of Neutral Endopeptidase 24.11 and Endothelin-Converting Albumin.” The Lancet 13-15 (1988). Enzyme.” Biochem. Biophys. Res. Commun. 204(1):407-412 (1994). Fraiser et al., “Murine Strain Differences in Metabolism and Bladder Delphion English-language abstract of European Patent Publication Toxicity of Cyclophosphamide.” Toxicology 75:257-272 (1992). No. 0 072987 Al, 2 pages, accessed on Oct. 20, 2005 at https://www. Freed et al., “Evidence for Acyloxmethyl Esters of Pyrimidine delphion.com/details?pn=EP00072987A1&s FAMILY=1. 5'-Deoxyribonucleotides as Extracellular Sources of Active Denmarket al., Tetrahedron 48(11):2191-2208 (1992). 5'-Deoxyribonucleotides in Cultured Cells.” Biochem. Pharmac. Desos et al., “Structure-Activity Relationships in a Series of 2(1H)- 38:3193-3 198 (1989). Quinolones Bearing Different Acidic Function in the 3-Position: Freeman et al., “Prodrug Design for Phosphates and Phosphonates.” 6,7-Dichloro-2(1H)-oxoquinoline-3phosphonic Acid, a New Potent Progress in Medicinal Chemistry 34:111-147 (1997). and Selective AMPAKainate Antagonist with Neuroprotective Prop Friis et al., “Prodrugs of Phosphates and Phosphonates: Novel erties,” J. Med. Chem. 39:197-206 (1996). Lipophilie O-acloxyalkyl Ester Derivatives of Phosphate—or de Waziers et al., “Cytochrome P450 Isoenzymes, Epoxide Phosphonate Containing Drugs Masking the Negative Charges of Hydrolase and Glutathlone Transferases in Rat and Human Hepatic these Groups.” Euro J. Pharm. Sci., 4:49-59 (1996). and Extrahepatic Tissues 1.” The Journal of Pharmacology and Gao et al., “Asymmetric Synthesis of Both Enantiomers of Experimental Therapeutics, 253(1):387-394 (1989). Tomoxetine and Fluoxetine. Selective Reduction of 2,3-Epoxycin Dickson et al., “Orally Active Squalene Synthase Inhibitors: namyl Alcohol with Red-A1.” J. Org. Chem. 53:4081-4084 (1988). Bis((acyloxy)alkyl) Prodrugs of the O-Phosphonosulfonic Acid Moi Gilard et al., "Chemical Stability and Fate of the Cytostatic Drug ety.” J. Med. Chem. 39.661-664 (1996). Ifosfamide and Its N-Dechloroethylated Metabolities in Acidic Edmunson et al., “Cyclic Organophosphorus Compounds. Part 23. Aqueous Solutions.” J. Med. Chem. 42:2542-2560 (1999). Configurational Assignments in the 4-Phenyl-1, 3, 2.5- Gorenstein et al., “Stereoelectronic Effects in the Reactions of dioxaphosphorinane Series. X-Ray Molecular Structure of cis-2- Epimeric 2-Aryloxy-2-oxy-1, 3,2-dioxaphosphorinanes and Benzylamino-4-phenyl-1,3, 2-dioxaphosphorinane 2-Oxide.” J. Oxazaphosphorinanes.” J. Am. Chem. Soc. 102:5077-5081 (1980). Chem. Res. Synop. 5:122-123 (1989). Groenet al. “Intracellular Compartmentation and Control of Alanine Elliott et al., “Synthesis and Biological Evaluation of Metabolism in Rat Liver Parenchymal Cells.” Eur: J. Biochem. Phosphonamidate Peptide Inhibitors of Enkephalinase and 122:87–93 (1982). Angiotensin-Converting Enzyme.” J. Med. Chem. 28:1208-1216 Guida et al., “Structure-Based Design of Inhibitors of Purina (1985). Nucleoside Phosphorylase. 4. A Study of Phosphate Mimics.”.J. Med. English-language abstract for EP0002062, esp(a)cenet Worldwide Chem. 37: 1109-1114 (1994). database (1 page), (May 30, 1979). Gurtoo et al., “Role of Glutathione in the Metabolism-dependent English-language abstract for EP0072531, esp(a)cenet Worldwide Toxicity and Chemotherapy of Cyclophosphamide.” Cancer database (1 page), (Feb. 23, 1983). Research 41:3584-3591 (1981). US 8,080,536 B2 Page 4

Gustin et al., “A Rapid, Sensitive Assay for .” Lok et al., “Neurotoxicity associated with adenine arabinoside Analytical Biochemistry 71:527-532 (1976). monophosphate in the treatment of chronic hepatitis B virus infec Harada et al., “Resolution of 1,3-alkanediols Via Chiral Spiroketals tion.” J. Antimicrob. Chemotherap. 14:93-99 (1984). Derived from i-Menthone.” Tetrahedron 28(41):4843-4846 (1987). Lorey et al., “A New-Cyclic Phosphoramidate D4T Prodrug Hayakawa et al., “Benzimidazolium Triflate as an Efficient Promoter Approach CycloAmb-D4T-Phosphoramidates.” Nucleosides & for Nucleotide Synthesis via the Phosphoramidite Method.” J. Org. Nucleotides 18(4&5):947-948 (1999). Chem. 61:7996-7997 (1996). Low et al., "Conversion of 4-Hydroperoxycyclophosphamide and He et al., Inactivation of Cytochrome P450 3A4 by Bergamottin a 4-Hydroxycyclophosphamide to Phosphoramide Mustard and Component of Grapefruit Juice, Chem. Res. Toxicol. 11(4):252-259 Acrolein Mediated, by Bifunctional Catalysts.” Cancer Research (1998). Hillers et al., “Analogs of pyrimidinemono-and polynucleotides. IV. 42:830-837 (1982). Phosphates of 1-(1,4-dihydroxy-2-pentyl)thymine and 1-(1,3- Lu et al., “Palladium-Catalyzed Reaction of Aryl dihydroxy-2-propyl) uracil” Khim Geterotski Soedin 5:678-683 Polyfluoroalkanesulfonates with O.O- Dialkyl Phosphonates.” Syn (1978) Chem Abstr. vol. 89. No. 17; abst. No. 146864u. thesis 726-727 (1987). Hilton, “Role of Aldehyde Dehydrogenase in Cyclophosphamide Ludeman et al., “Synthesis and Antitumor Activity of resistant L1210 Leukemia,” Cancer Research 44:5156-5160 (1984). Cyclophosphamide Analogs. 1. Benzo Annulated Hirayama et al., “Structure and Conformation of a Novel Inhibitor of Cyclophosphamide and Related Systems,” J. Med. Chem. Angiotensin I Converting Enzyme—a Tripeptide Containing 18(12): 1251-1253 (1975). Phosphonic Acid.” Int. J. Pept Protein Res. 38:20-24 (1991). Ludeman et al., Synthesis and Antitumor Activity of Hulst et al., “A New PNMR Method for the Enantiomeric Excess Cyclophosphamide Analogues. 2. Preparation, Hydrolytic Studies, Determination of Alcohols, Amines and Amino Acid Esters.” Tetra and Anticancer Screening of 5-Bromocyclophosphamide, 3.5- hedron Letters 34(8): 1339-1342 (1993). Dehydrocyclophosphamide, and Related Systems; J. Med Chem. Hunston et al., “Synthesis and Biological Properties of Some Cyclic 22(2): 151-158 (1979). Phosphotriesters Derived from 2'-Deoxy-5-fluorouridine.” J. Med. Ludeman et al., “Synthesis and Antitumor Activity of Chem. 27:440-444 (1984). Cyclophosphamide Analogues. 4. Preparation, Kinetic Studies, and Jain et al., "Sulfonyl-Containing Aldophosphamide Analogues as Anticancer Screening of "Phenyl ketophosphamide' and Similar Novel Anticancer Prodrugs Targeted against Cyclophosphamide-Re Compounds Related to the Cyclophosphamide Metabolyte sistant Tumor Cell Lines.” J. Med. Chem. 47:3843-3852 (2004). Aldophosphamide.” J. Med. Chem. 29:716-727 (1986). Jounaidi et al., “Retroviral Transfer of Human Cytochrome P450 Genes for Oxazaphosphorine-based Cancer Gene Therapy,” Cancer Ludeman et al., “Synthesis of Reactive Metabolite-Analogues of Research 58:4391-4401 (1998). Cyclophosphamide for Comparisons of NMR Kinetic Parameters Jounaidi et al., “Frequent, Moderate-Dose Cyclophosphamide and Anticancer Screening Data.” Drugs Exptl. Clin. Res. XII(6/ Administration Improves the Efficacy of Cytochrome P-450/ 7):527-532 (1986). Cytochrome P-450 Reductase-based Cancer Gene Therapy.” Cancer May-Manke et al., “Investigation of the Major Human Hepatic Research 61:4437-4444 (2001). Cytochrome P450 Involved in 4-Hydroxylation and Kachel et al., “Cyclophosphamide-Induced Lung Toxicity: Mecha N-dechloroethylation of Trofosfamide.” Cancer Chemother: nism of Endothelial Cell Injury.” J. Pharmacology and Experimental Pharmacol. 44:327-334 (1999). Therapeutics 268(1):42-46 (1994). Maynard-Faureet al., “New Strategy for the Diastereoselective Syn Keenan et al., “Pathology Reevaluation of the Kociba et al. (1978) thesis of Bicyclic Pre-activated Analogues of Cyclophosphamide.” Bioassay of 2,3,7,8-TCDD: Implications for Risk Assessment.” J. Tetrahedron Letters 39:2315-2318 (1998). Tox. Envir: Health 34:279-296 (1991). McGuigan et al., “Intracellular Delivery of Bioactive AZT Kelley et al., “II(Guaninylalkyl)phosphinico methyl) phosphonic Nucleotides by Aryl Phosphate Derivatives of AZT”.J. Med Chem. Acids. Multisubstrate Analogue Inhibitors of Human Erythrocyte 36:1048-1052 (1993). Purine Nucleoside Phosphorylase.” J. Med. Chem. 38:1005-1014 McGuigan et al., “Kinase Bypass: A New Strategy for Anti-HIV Drug (1995). Design.” Bioorganic & Medicinal Chemistry Letters 3(6):1207-1210 Khamnei et al., “Neighboring Group Catalysis in the Design of (1993). Nucleotide Prodrugs,” J. Med. Chem. 39:4109-4115 (1996). Meier et al., “ADA Bypass by lipophilic Cyclo-Sal-ddAMP Pro Khorana et al., “Cyclic Phosphates. III. Some General Observations Nucleotides a second Example of the Efficiency of the cycloSat on the Formation and Properties of Five-, Six-and Seven-membered Concept.” Bioorg. Med. Chem. Lett. 7(12): 1577-1582 (1997). Cyclic Phophate Esters,” JACS 79:430-436 (1957). Meier et al., “Cyclic Saligenyl Phosphotreisters of 2',3'-Dideoxy-2", Korba et al., “Liver-targeted Antiviral Nucleosides: Enhanced Anti 3'-didehydrothymidine (d4T)—A New Pro-Nucleotide Approach.” viral Activity of Phosphatidyl-dideoxyguanosine Versus Bioorg. Med. Chem. Lett. 7:99-104 (1997). Dideoxyguanosine in Woodchuck Hepatitis Virus Infection InVivo.” Meijer et al., "Covalent and Noncovalent Protein Binding of Drugs: Hepatology 23(5):958-963 (1996). Implications for Hepatic Clearance, Storage, and Cell-Specific Drug Kryuchkov et al., Izy. Akad. Nauk SSSR. Ser: Khim. 6: 1201-1248 Delivery:Pharm'). Res. 6(2):105-118 (1989). (1987). Melvin, “An Efficient Synthesis of 2-Hydroxyphenylphosphonates.” Kuriyama et al., “Transient Cyclophosphamide Treatment Before Tetrahedron Lett. 22(35):3375-3376 (1981). Intraportal Readministration of an Adenoviral Vector can Induce Merckling et al., “Diasteroselectivity in Nucleophilic Displacement Re-expression of the Original Gene Construct in Rat Liver.” Gene Reactions at Phosphorus; Isolation and Characterization of Therapy 6:749-757 (1999). Pentacoordinated Intermediate.” Tetrahedron Letters 37(13):2217 Kwon et al., “Effects of N-Substitution on the Activation Mecha 2220 (1996). nisms of 4Hydroxycyclophosphamide Analogues.” J. Med Chem. Meyer et al., “2-O'-Acyl-6-thioinosine Cyclic 3',5'-Phosphates as 32: 1491-1496 (1989). Prodrugs of Thioinosinic Acid.” J. Med. Chem. 22:811-815 (1979). Lefebvre et al., “Mononucleotide Phosphotriester Derivatives with Misiura et al., “Synthesis and Antitumor Activity of Analogues of S-Acyl-2-thioethyl Bioreversible Phosphate-Protecting Groups: Ifosfamide Modified in the N-(2-Chloroethyl) Group”.J.Med. Chem. Intracellular Delivery of 3'-Azido-2', 3'-dideoxythymidine 31(1):226-230 (1988). 5'-Monophosphate.” J. Med. Chem. 38:3941-3950 (1995). Mitchell et al., "Acetaminophen-Induced Hepatic Necrosis. IV. Pro Lilo et al., “Synthesis and Configurational Assignment of Bicyclic tective Role of Glutathione.” J. Pharmacology and Experimental Preactivated Analogues of Cyclophosphamide.” Tetrahedron Let Therapeutics 187(1):211-217 (1973). ters 31(6):887-890 (1990). Mitchell et al., “Bioreversible Protection for the Phospho Group: Lohr et al., “Targeted chemotherapy by intratumor injection of Bioactivation of the Di(4-acyloxybenzyl) and Mono(4- encapsulated cells engineered to produce CYP2B1, and ifosfamide acyloxybenzyl) Phosphoesters of Methylphosphonate and activating cytochrome P450.” Gene Therapy 5:1070-1078 (1988). Phosphonacetate.”J. Chem. Soc. Perkin Trans. 1:2345-2353 (1992). US 8,080,536 B2 Page 5

Mitsunobu . “The Use of Diethyl Azodicarboxylate and Ren et al., “Inhibition of Human Aldehyde Dehydrogenase 1 by the Triphenylphosphine in Synthesis and Transformation of Natural 4Hydroxycyclophosphamide Degradation Product Acrolein.” Drug Products.” Synthesis 1-28 (1981). Metabolism and Disposition 27(1): 133-137 (1999). Moore et al., “Comparison of Mutagenicity Results for Nine Com Ren et al., “Pharmacokinetics of Cyclophosphamide and its Metabo pounds Evaluated at the hgprt Locus In the Standard and Suspension lites in Bone Marrow Transplantation Patients.” Clinical Pharmacol CHO Assays.” Mutagenesis 6:77-85 (1991). ogy and Therapeutics 64(3):289-301 (1998). Mosbo et al., “Dipole Moment, Nuclear Magnetic Resonance, and Reusch, “Virtual Text of Organic Chemistry.” Michigan State Uni Infrared Studies of Phosphorus Configurations and Equilibria in 2-R- versity, online Aug. 1, 2004, retrieved on Jan. 19, 2005). Retrieved 2-Oxo-1, 3, 2-dioxaphosphorinones,” J. Org. Chem. 42(9): 1549 from the internet,

Watkins, “Noninvasive Tests of CYP3A Enzymes.” Pharmacogenet Yip et al., “Use of High-Performance Liquid Chromatography in the ics 4:171-184 (1994). Preparation of Flavin Adenine DinuleotideAnalyte Conjugates.”.J. of Weber et al., “Activation of the Anti-cancer Drug Ifosphamide by Rat Chromatography 326:301-310 (1985). Liver Microsomal P450 Enzymes.” Biochem. Pharm. 45(8): 1685 1694 (1993). Yu et al., “In vivo Modulation of Alternative Pathways of P-450°Cata Weibel et al., “Potentiating Effect of {2-(2-(2-Amino-1,6-Dihydro lyzed Cyclophosphamide Metabolism: Impact on Pharmacokinetics 6-Oxo-9H-Purin-9ylMethytl-Phenyl Ethenyl Phosphonic Acid and Antitumor Activity.” J. Pharm. Exp. Ther: 288(3):928-937 (MDL 74,428), A Potent Inhibitor of Purina Nucleoside (1999). Phosphorylase, on the Antiretroviral Activities of 2',3'- Dideoxyinosine Combined to Ribavirin in Mice.” Biochem. Yule et al., “The Effect of Fluconazole on Cyclophosphamide Pharmacol. 48(2):245-252 (1994). Metabolism in Children.” Drug Metabolism and Disposition Weinhardt et al., “Synthesis and Antidepressant Profiles of Phenyl 27(3):417-421 (1999). Substituted 2- Amino- and 2-(Alkoxyearbonyl)amino-1,4,5,6- Zon et al. “4 Cyclophosphamide Analogues.” Prog. Med. Chem. tetrahydropyrimidines.” J. Med Chem. 28:694-898 (1985). 19:205-246 (1982). Wileman et al., “Receptor-Mediated Endocytosis.” Biochem. J. Zonet al., “NMR Spectroscopic Studies of Intermediary Metabolites 232:1-14 (1985). Wolff, “Burger's Medicinal Chemistry, 5ed, Part I.” John Wiley & of Cyclophosphamide. A Comprehensive Kinetic Analysis of the Sons, 1995 pp.975-977. Interconversation of cis- and trans-4-Hydroxycyclophosphamide Yamanaka et al., “Metabolic Studies on BMS-200475, a New Anti with Aldophosphamide and Concomitant Partitioning of viral Compound Active against Hepatitis B Virus.” Antimicrob, Aldophosphamide Between Irreversible Fragmentation and Revers Agents Chemoth. 43:190-193 (1999). ible Conjunction Pathways.” J. Med Chem. 27:466-485 (1984). US 8,080,536 B2 1. PHOSPHORUS-CONTAINING PRODRUGS

RELATED APPLICATION

This application is a continuation of U.S. application Ser. No. 1 1/779,689 filed Jul.18, 2007, now allowed, which is a continuation application of U.S. application Ser. No. 1 1/601, The cyclic 2,2'-difluoro-1',3'-propane ester is reported to 843 filed Nov. 20, 2006, now abandoned, which is a divisional 10 be hydrolytically unstable with rapid generation of the ring application of U.S. application Ser. No. 09/518,501 filed Mar. opened monoester. Starrett et al. J. Med. Chem. 37: 1857 3, 2000, now patented, which claims priority to U.S. Provi 1864 (1994). sional Application Ser. No. 60/153,127, filed Sep. 8, 1999, Cyclic 3',5'-phosphate esters of araA, araC and thioinosine and Provisional Application Ser. No. 60/123,013, filed Mar. have been synthesized. Meier et al., J. Med. Chem. 22:811 5, 1999, and each of which is incorporated by reference in 15 815 (1979). These compounds are ring-opened through the their entirety. action of phosphodiesterases which usually require one nega tive charge. Cyclic 1.3'-propanyl phosphonate and phosphate esters FIELD OF THE INVENTION are reported containing a fused aryl ring, i.e. the cyclosalig enyl ester, Meier et al., Bioorg. Med. Chem. Lett. 7: 99-104 (1997). These prodrugs are reported to generate the phos phate by a “controlled, non-enzymatic mechanisms at The present invention is directed towards novel prodrugs physiological pH according to the designed tandem-reaction that generate phosph(on)ate compounds which are biologi in two coupled steps”. The strategy was purportedly used to cally active or are further phosphorylated to produce biologi 25 deliver d4-T monophosphate to CEM cells and CEM cells cally active compounds, to their preparation, to their synthetic deficient in thymidine kinase infected with HIV-1 and HIV-2. intermediates, and to their uses. More specifically, the inven Unsubstituted cyclic 1',3'-propanyl esters of the mono tion relates to the area of substituted cyclic 1,3-propanyl phosphates of 5-fluoro-2'-deoxy-uridine (Farquhar et al., J. esters wherein the cyclic moiety contains at least one amino Med. Chem. 26: 1153 (1983)) and ara-A (Farquhar et al., J. attached to the phosphorus. 30 Med. Chem. 28: 1358 (1985)) were prepared but showed no in vivo activity. In addition, cyclic 1',3'-propanyl esters substi tuted with a pivaloyloxy methyloxy group at C-1" was pre BACKGROUND OF THE INVENTION pared for 5-fluoro-2'-deoxy-uridine monophosphate (5-FdUMP: (Freed et al., Biochem. Pharmac. 38: 3193 The following description of the background of the inven 35 (1989); and postulated as potentially useful prodrugs by oth tion is provided to aid in understanding the invention, but is ers (Biller et al., U.S. Pat. No. 5,157,027). In cells, the acyl not admitted to be, or to describe, prior art to the invention. All group of these prodrugs underwent cleavage by esterases to cited publications are incorporated by reference in their generate an unstable hydroxyl intermediate which rapidly entirety. broke down to the free phosphate and acrolein following a 40 B-elimination reaction as well as formaldehyde and pivalic Free phosphorus and phosphonic acids and their salts are acid. highly charged at physiological pH and therefore frequently Unsubstituted cyclic phosphoramidate esters, i.e. cyclic exhibit poor oral bioavailability, poor cell penetration and phosphonate and phosphate esters wherein one of the ring limited tissue distribution (e.g. CNS). In addition, these acids oxygens is replaced with an NR, are also known. For are also commonly associated with several other properties 45 example, cyclophosphamide (CPA) is representative of a that hinder their use as drugs, including short plasma half-life class of mustard oncolytics that utilize this prodrug moiety. due to rapid renal clearance, as well as toxicities (e.g. renal, Although CPA is activated primarily in the liver via a cyto gastrointestinal, etc.) (e.g. Antimicrob Agents Chemother chrome P450-catalyzed oxidation, its biological activity is 1998 May; 42(5): 1146-50). Phosphates have an additional outside the liver. CPA is an effective immunosuppressive limitation in that they are not stable in plasma as well as most 50 agent as well as an oncolytic agent for extrahepatic cancers tissues since they undergo rapid hydrolysis via the action of because one or more of the intermediate metabolites pro phosphatases (e.g. alkaline phosphatase, nucleotidases). duced following P450 activation diffuses out of the liver and Prodrugs of phosphorus-containing compounds have been into the circulation. With time the intermediate(s) enter extra sought primarily to improve the limited oral absorption and hepatic tissues and are thought to undergo a B-elimination 55 reaction to generate acrolein and the active phosphoramide poor cell penetration. In contrast to carboxylic acid proesters, mustard. Both products are reported to be cytotoxic to cells. many phosphonate and phosphate esters fail to hydrolyze in The mustard cytotoxicity results from alkylation of DNA (or Vivo, including simple alkyl esters. The most commonly used RNA). Acrolein is reported to exert its toxicity via several prodrug class is the acyloxyalkyl ester, which was first mechanisms, including depletion of glutathione, alkylation of applied to phosphate and phosphonate compounds in 1983 by 60 DNA and proteins via a Michael reaction. In addition, Farquhar et al., J. Pharm. Sci. 72(3):324 (1983). acrolein produces other toxicities such as the dose-limiting Cyclic phosphonate and phosphate esters have also been bladder toxicity commonly observed with cyclophosphamide described for phosphorus-containing compounds. In some therapy. Since the toxicity of these agents oftenhampers their cases, these compounds have been investigated as potential use as chemotherapy agents, numerous strategies are under phosphate or phosphonate prodrugs. Hunston et al., J. Med. 65 investigation that are designed to enhance P450 activity in or Chem. 27: 440-444 (1984). The numbering for these cyclic near tumors and thereby localize the activation and antipro esters is shown below: liferative effect of these agents to the tumor. One strategy uses US 8,080,536 B2 3 4 retroviruses or other well known techniques for introducing Numerous phosphorus-containing compounds are known genes into target tissues (e.g. Jounaidi et al., Cancer Research to exhibit pharmacological activity but remain far from opti 58, 4391 (1998)). Other strategies include the placement of mal due to one or more of the above-described limitations. encapsulated cells engineered to produce cytochrome P450s Some of the activities described include phosphonic acids (e.g. Lohr et al., Gene Therapy 5, 1070 (1998)) at or near the that are useful as antihypertensives and therapy for heart tumor. failure via inhibition of NEP24.11, phosphonic acids that are Unsubstituted cyclic phosphoramidate esters have also useful for treating a variety of CNS conditions (stroke, epi been prepared as potential prodrugs of the nucleosides araA lepsy, brain and spinal cord trauma, etc.) via binding to exci and 5-fluoro-2'-deoxyuridine (Farquhar et al., J. Med. Chem. tory amino acid receptors (e.g. NMDA receptor), bisphos 28, 1358 1361 (1985); J. Med. Chem. 26, 1153-1158 (1983)). 10 phonic acids that are useful for treating osteoporosis, The compounds were studied in a mouse model of leukemia where it was assumed that if the prodrug transformation was phosphonic acids that are useful as lipid lowering agents (e.g. similar to cyclophosphamide, then these agents would be squalene synthase inhibitors), phosphonates that are useful in useful for treating a variety of cancers including leukemias as treating inflammation (e.g. collagenase inhibitors), phospho well as carcinomas of the colon, breast and ovary. In addition, 15 nates and phosphates that are useful in treating diabetes, since some of the mechanisms that account for tumor cell cancer and parasitic and viral infections. drug resistance entail a decrease in the enzymes used to Phosphates and phosphonates that are known to be particu synthesize the monophosphate, the strategy was expected to larly useful in glucose lowering activity and therefore are possibly be beneficial in treating drug resistant tumors. The anticipated to be useful in treating diabetes are compounds compounds were only “marginally effective' in prolonging that bind to the AMP site of fructose 1,6-bisphosphatase life span in the mouse model. (FBPase) as described in U.S. Pat. No. 5,658,889, WO A variety of substituted 1',3' propanyl cyclic phosphorami 98/39344, WO 98/39343, and WO 98/39342. Other examples dates, wherein 1' represents the carbon alpha to the nitrogen of phosphorus-containing drugs include squalene synthetase were prepared as cyclophosphamide analogs (Zon, Progress inhibitor (e.g. BMS 188494). in Med. Chem. 19, 1205 (1982)). For example, a number of 2'- 25 A large class of drugs known to be active against hepatitis and 3'-substituted proesters were prepared in order to are generally nucleoside or nucleotide analogs that are phos decrease the propensity of the C. B-unsubstituted carbonyl phorylated inside cells to produce the biologically active by-product to undergo a Michael reaction. 2'-Substituents triphosphate. Examples include Lamivudine (3TC) and included methyl, dimethyl, bromo, trifluoromethyl, chloro, Vidarabine (ara A). In each case, the drug interferes with viral hydroxy, and methoxy whereas a variety of groups were used 30 replication via the triphosphate form through either inhibition at the 3'-position including phenyl, methyl, trifluoromethyl, of the viral DNA polymerases or DNA chain termination. ethyl, propyl, i-propyl, and cyclohexyl. Analogs with a 3'-aryl Some specificity for virus-infected cells is gained by both group e.g. trans-4-phenylcyclophosphamide were “moder preferential phosphorylation of the drug by virally-encoded ately effective in L1210 test system and showed no activity in kinases as well as by specific inhibition of viral DNA poly vivo” G. Zu Prog. Med. Chem. 19: 205-246 (1982). A variety 35 merases. Nevertheless, many of the nucleoside-based drugs of 1'-substituted analogs were also prepared. In general these are associated with significant non-hepatic toxicity. For compounds were designed to be "pre-activated cyclophos example, araA frequently produces neurological toxicity phamide analogs that bypass the oxidation step by already (40%) with many patients showing myalgia or a sensory existing as a 1'-substituted analog capable of producing the neuropathy with distressing pain and abnormalities in nerve final compound, e.g. hydroperoxide and 1-thioether. A series 40 conduction and a few showing tremor, dysarthria, confusion of 1-aryl analogs were also prepared in order to enhance the or even coma. Lok et al., J Antimicrob. Chemotherap. 14. oxidation potential. In contrast to the 1'-hydroperoxy analogs, 93-99 (1984). In other cases, the efficacy and/or therapeutic the 1'-aryl compounds exhibited either no activity or very index of nucleosides is compromised by poor phosphoryla poor activity in the standard anticancer in vivo Screen assay, tion efficiencies and therefore low levels of the biologically i.e. the mouse L1210 assay. The lack of activity was postu 45 active triphosphate (e.g. Yamanaka et al., Antimicrob. Agents lated to arise from the steric hindrance of the phenyl and and Chemother: 43, 190 (1999)). therefore limited oxidation of the prodrug. Support for this Phosphonic acids also show antiviral activity. In some postulate was the potent activity of the acyclic phenylketo cases the compounds are antivirals themselves (e.g. phospho analog which exhibited activity similar to cyclophosphamide. noformic acid), whereas in other cases they require phospho Luderman et al. J. Med. Chem. 29: 716 (1986). 50 rylation to the disphosphate, e.g. 9-(2-phosphonylmethoxy Cyclic esters of phosphorus-containing compounds are ethyl)adenine (PMEA, Adefovir). Frequently, these reported in the chemical literature, however they were not compounds are reported to exhibit enhanced activity due to tested as prodrugs in biological systems. These cyclic esters either poor Substrate activity of the corresponding nucleoside include: with viral kinases or because the viral nucleoside kinase 1 di and triesters of phosphoric acids as reported in Nifan 55 which is required to convert the nucleoside to the monophos tyev et al., Phosphorus, Sulfur Silicon and Related Ele phate is down regulated viral resistance. Monophosphates ments, 113: 1 (1996); Wijnberg et al., EP-180276A1; and phosphonic acids, however, are difficult to deliver to 2 phosphorus (III) acid esters. Kryuchkov et al., Izv. Akad. virally-infected cells after oral administration due to their Nauk SSSR, Ser. Khim. 6:1244 (1987). Some of the com high charge and in the case of the monophosphate instability pounds were claimed to be useful for the asymmetric Syn 60 in plasma. In addition, these compounds often have short thesis of L-Dopa precursors. Sylvain et al., DE3512781 half-lives (e.g. PMEA, Adefovir) due in most cases to high A1: renal clearance. In some cases, the high renal clearance can 3 phosphoramidates. Shih et al., Bull. Inst. Chem. Acad. Sin, lead to nephrotoxicities or be a major limitation in diseases 41: 9 (1994); Edmundson et al., J. Chem. Res. Synop. 5: Such as diabetes where renal function is often compromised. 122 (1989); and 65 Liver cancer is poorly treated with current therapies. In 4 phosphonates. Neidlein et al., Heterocycles 35:1185 general, liver tumors are resistant to radiotherapy, respond (1993). poorly to chemotherapy and are characterized by a high US 8,080,536 B2 5 6 degree of cell heterogeneity. Oncolytic nucleosides such as bonyloxy, attached to one of said additional carbonatoms that 5-fluoro-2'-deoxyuridine, have also shown a poor response is three atoms from a Y attached to the phosphorus: against primary liver cancers. together Zand Ware connected via an additional 3-5 atoms to form a cyclic group, optionally containing one heteroatom, SUMMARY OF THE INVENTION and V must be aryl, substituted aryl, heteroaryl, or substituted heteroaryl; The present invention is directed towards novel prodrugs together W and W are connected via an additional 2-5 that generate phosph(on)ate compounds, their preparation, atoms to form a cyclic group, optionally containing 0-2 het their synthetic intermediates, and their uses. In one aspect, the eroatoms, and V must be aryl, Substituted aryl, heteroaryl, or invention is directed towards the use of the prodrugs to 10 substituted heteroaryl; enhance oral drug delivery. Another aspect of the invention is Z is selected from the group consisting of —CHROH, directed to the use of the prodrugs to enhance the level of the CHROC(O)R, CHROC(S)R, CHROC(S)OR, biologically active drug in the liver. Another aspect of the CHROC(O)SR, CHROCOR, OR, SR, invention is the use of the prodrugs to treat diseases that —CHRN, -CHaryl, -CH(aryl)OH, -CH(CH=CR) benefit from enhanced drug distribution or specificity to the 15 liver and like tissues and cells, including hepatitis, cancer, OH, CH(C=CR)OH, R, NR, OCOR, liver fibrosis, malaria, other viral and parasitic infections, and OCOR, SCOR, SCOR, NHCOR, metabolic diseases where the liver is responsible for the over —NHCO.R. —CH-NHaryl, -(CH.) OR', and production of the biochemical end product, e.g. glucose (dia —(CH), SR': betes); cholesterol, fatty acids and triglycerides (hyperlipi p is an integer 2 or 3: demia) (atherosclerosis) (obesity). In another aspect, the with the provisos that: prodrugs are used to prolong pharmacodynamic half-life of a) V, Z, W, Ware not all-H; and the drug. In addition, the prodrug methodology of the current b) when Z is R, then at least one of V, W, and W is not invention is used to achieve sustained delivery of the parent —H, alkyl, aralkyl, or alicyclic; drug. In another aspect, the prodrugs are used to increase the 25 R’ is selected from the group consisting of Rand-H; therapeutic index of the drug. Another aspect of the invention R is selected from the group consisting of alkyl, aryl, is the use of the prodrugs in combination with techniques that alicyclic, and aralkyl, elevate P450 activity in specific tissues. In another aspect of R is selected from the group consisting of H. lower the invention, a method of making these prodrugs is alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl, and lower acyl: described. A further aspect is the novel intermediates to these 30 R'' is selected from the group consisting of H, and lower prodrugs. In another aspect, the prodrugs are also useful in the acyl: delivery of diagnostic imaging agents to the liver. each Y is independently selected from the group consisting One aspect of the present invention concerns compounds of-O-, - NR with the proviso that at least one Y is that are converted in vitro or in Vivo to the corresponding NR ; M-PO. , MPO. , MPO. , and MP(O)(NHR)O 35 Mis selected from the group that attached to PO, P.O., and are of formula P.O." or P(O)(NHR)O is a biologically active agent, but is not an FBPase inhibitor, and is attached to the phosphorus in formula I via a carbon, oxygen, Sulfur or nitrogen atom; with the provisos that: V 40 H 1) M is not NH(lower alkyl). —N(lower alkyl), NH O Y (lower alkylhalide), N(lower alkylhalide), or - N(lower V / M-P Z alkyl) (lower alkylhalide); and V 2) R' is not lower alkylhalide; Y H and pharmaceutically acceptable prodrugs and salts 45 W. W thereof. The present invention provides several novel methods of wherein: making the prodrugs of the present invention. One method V, W, and W are independently selected from the group relies on the reaction of the following novel P(III) reagent: consisting of —H, alkyl, aralkyl, alicyclic, aryl, Substituted 50 aryl, heteroaryl, Substituted heteroaryl, 1-alkenyl, and 1-alky V nyl; or H Y together V and Z are connected via an additional 3-5 atoms / to form a cyclic group containing 5-7 ring atoms, optionally 1 M-GH + L-P Z --> heteroatom, Substituted with hydroxy, acyloxy, alkoxycarbo 55 G' = O, Y H nyloxy, oraryloxycarbonyloxy attached to a carbon atom that NR is three atoms from bothY groups attached to the phosphorus: W Wi O L = leaving groups such as together V and Z are connected via an additional 3-5 atoms -NR2, halogen V to form a cyclic group, optionally containing 1 heteroatom, 60 H said cyclic group is fused to an aryl group at the beta and Y / gamma position to the Yadjacent to V. M-G-P Z together V and Ware connected via an additional 3 carbon V atoms to forman optionally substituted cyclic group contain Y H ing 6 carbon atoms and Substituted with one Substituent 65 W W selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycar US 8,080,536 B2 7 8 The resulting phosphite is then oxidized to the cyclic phos the remainder of the ring atoms being carbonatoms. Suitable phoramidate. heteroatoms include oxygen, Sulfur, and nitrogen. Suitable A second method relies on the reaction of a novel P(V) heteroaryl groups include furanyl, thienyl, pyridyl, pyrrolyl, reagent: N-lower alkyl pyrrolyl pyridyl-N-oxide, pyrimidyl, pyrazi nyl, imidazolyl, and the like, all optionally substituted. V The term “biary1' represents aryl groups containing more H than one aromatic ring including both fused ring systems and O. Y V / Z aryl groups Substituted with other aryl groups. Such groups M-G-H + L-P Ho may be optionally substituted. Suitable biaryl groups include i V H 10 G' = O, Y naphthyl and biphenyl. NH The term “alicyclic' means compounds which combine the W Wi properties of aliphatic and cyclic compounds. Such cyclic L' = leaving groups such as compounds include but are not limited to, aromatic, -NR2, -O-aryl, cycloalkyl and bridged cycloalkyl compounds. The cyclic halogen 15 V compound includes heterocycles. Cyclohexenylethyl and H cyclohexylethyl are suitable alicyclic groups. Such groups O Y V / Z may be optionally substituted. M-G-P The term “optionally substituted” or “substituted includes V H groups substituted by one to four Substituents, independently Y selected from lower alkyl, lower aryl, lower aralkyl, lower W W alicyclic, hydroxy, lower alkoxy, lower aryloxy, perha loalkoxy, aralkoxy, heteroaryl, heteroaryloxy, heteroaryla A third method relies on reacting another novel P(V) com lkyl, heteroaralkoxy, azido, amino, guanidino, amidino, halo, pound with a diamine or amino alcohol: 25 lower alkylthio, oxo, acylalkyl, carboxy esters, carboxyl, -carboxamido, nitro, acyloxy, aminoalkyl, alkylaminoaryl, alkylaryl, alkylaminoalkyl, alkoxyaryl, arylamino, aralky O OH O L" lamino, phosphono, Sulfonyl, -carboxamidoalkylaryl, -car V / V / M-G-P -> M-G-P -- boxamidoaryl, hydroxyalkyl, haloalkyl, alkylaminoalkylcar M V 30 boxy-, aminocarboxamidoalkyl-, cyano, lower alkoxyalkyl, OH L" lower perhaloalkyl, and arylalkyloxyalkyl. “Substituted aryl L' = halogen and “substituted heteroaryl” preferably refers to aryl and G" = O, NH, f heteroaryl groups substituted with 1-3 substituents. Prefer ably these Substituents are selected from the group consisting V 35 of lower alkyl, lower alkoxy, lower perhaloalkyl, halo, hydroxy, and amino. O Y V / The term “-aralkyl refers to an alkylene group substituted HY-CH(V)CH(Z)CWWYH -- M'-G"-P Z with an aryl group. Suitable aralkyl groups include benzyl, Y picolyl, and the like, and may be optionally substituted. “Het 40 eroarylalkyl refers to an alkylene group substituted with a W Wi heteroaryl group. The term "-alkylaryl refers to an aryl group substituted Since these compounds have asymmetric centers, the with an alkyl group. "Lower-alkylaryl refers to Such groups present invention is directed not only to racemic and diaste where alkyl is lower alkyl. reomeric mixtures of these compounds, but also to individual 45 The term “lower” referred to herein in connection with Stereoisomers. The present invention also includes pharma organic radicals or compounds respectively defines Such as ceutically acceptable and/or useful salts of the compounds of with up to and including 10, preferably up to and including 6. formula I, including acid addition salts. The present inven and advantageously one to four carbon atoms. Such groups tions also encompass prodrugs of compounds of formula I. may be straight chain, branched, or cyclic. 50 The terms “arylamino” (a), and “aralkylamino” (b). DEFINITIONS respectively, refer to the group —NRR' wherein respectively, (a) R is aryland R' is hydrogen, alkyl, aralkyl or aryl, and (b) In accordance with the present invention and as used R is aralkyl and R' is hydrogen or aralkyl, aryl, alkyl. herein, the following terms are defined with the following The term “acyl refers to —C(O)R where R is alkyl and meanings, unless explicitly stated otherwise. 55 aryl. The term “aryl refers to aromatic groups which have 5-14 The term “carboxy esters' refers to –C(O)OR where R is ring atoms and at least one ring having a conjugated pi elec alkyl, aryl, aralkyl, and alicyclic, all optionally Substituted. tron system and includes carbocyclic aryl, heterocyclic aryl The term “carboxyl refers to C(O)OH. and biaryl groups, all of which may be optionally Substituted. The term “oxo” refers to —O in an alkyl group. Suitable aryl groups include phenyl and furan-2,5-diyl. 60 The term “amino refers to NRR' where R and R' are Carbocyclic aryl groups are groups wherein the ring atoms independently selected from hydrogen, alkyl, aryl, aralkyl on the aromatic ring are carbon atoms. Carbocyclic aryl and alicyclic, all except H are optionally substituted; and R groups include monocyclic carbocyclic aryl groups and poly and R' can form a cyclic ring system. cyclic or fused compounds such as optionally Substituted The term “-carboxylamido” refers to CONR where naphthyl groups. 65 each R is independently hydrogen or alkyl. Heterocyclic aryl or heteroaryl groups are groups having The term “halogen' or “halo refers to —F. —Cl, —Br from 1 to 4 heteroatoms as ring atoms in the aromatic ring and and —I. US 8,080,536 B2 10 The term “alkylaminoalkylcarboxy' refers to the group The term “aryloxyalkyl-” refers to an alkyl group substi alkyl-NR-alk-C(O)—O— where “alk” is an alkylene group, tuted with an aryloxy group. and R is a H or lower alkyl. The term “aralkyloxyalkyl-” refers to the group aryl-alk The term “alkyl refers to saturated aliphatic groups O-alk- wherein “alk” is an alkylene group. “Lower aralky including straight-chain, branched chain and cyclic groups. 5 loxyalkyl-” refers to Such groups where the alkylene groups Alkyl groups may be optionally substituted. Suitable alkyl are lower alkylene. groups include methyl, isopropyl, and cyclopropyl. The term “alkoxy s or “alkyloxy- refers to the group The term “cyclic alkyl or “cycloalkyl refers to alkyl alkyl-O . groups that are cyclic of 3 to 10 atoms, more preferably 3 to The term “alkoxyalkyl- or “alkyloxyalkyl-” refer to the 6 atoms. Suitable cyclic groups include norbornyl and cyclo 10 group alkyl-O-alk- wherein “alk” is an alkylene group. In propyl. Such groups may be substituted. “lower alkoxyalkyl-, each alkyl and alkylene is lower alky The term "heterocyclic” and "heterocyclic alkyl refer to lene. The terms “alkylthio- and “alkylthio-” refer to the groups cyclic groups of 3 to 10 atoms, more preferably 3 to 6 atoms, alkyl-S . containing at least one heteroatom, preferably 1 to 3 heteroa 15 The term “alkylthioalkyl-” refers to the group alkyl-5-alk toms. Suitable heteroatoms include oxygen, Sulfur, and nitro wherein “alk” is an alkylene group. In “lower alkylthioalkyl gen. Heterocyclic groups may be attached through a nitrogen each alkyl and alkylene is lower alkylene. or through a carbon atom in the ring. The heterocyclic alkyl The term “alkoxycarbonyloxy- refers to alkyl-O C groups include unsaturated cyclic, fused cyclic and spirocy (O)—O—. clic groups. Suitable heterocyclic groups include pyrrolidi- 20 The term “aryloxycarbonyloxy- refers to aryl-O C nyl, morpholino, morpholinoethyl, and pyridyl. (O)—O—. The term “phosphono” refers to —POR, where R is The term “alkylthiocarbonyloxy-” refers to alkyl-S C selected from the group consisting of H, alkyl, aryl, aralkyl, (O)—O—. and alicyclic. The terms "amido” or “carboxamido” refer to NR C The term “sulphonyl' or “sulfonyl refers to -SOR, 25 (O)—and RC(O) NR' , where RandR' include H, alkyl, where R is H, alkyl, aryl, aralkyl, and alicyclic. aryl, aralkyl, and alicyclic. The term does not include urea, The term “alkenyl refers to unsaturated groups which - NR-C(O) NR-. contain at least one carbon-carbon double bond and includes The term “carboxamidoalkylaryland “carboxamidoaryl straight-chain, branched-chain and cyclic groups. Alkenyl refers to an aryl-alk-NR'—C(O), and ar-NR'—C(O)-alk-, groups may be optionally Substituted. Suitable alkenyl groups 30 respectively where “ar” is aryl, "alk” is alkylene, R' and R include allyl. “1-alkenyl refers to alkenyl groups where the include H, alkyl, aryl, aralkyl, and alicyclic. double bond is between the first and second carbon atom. If The term “hydroxyalky1' refers to an alkyl group substi the 1-alkenyl group is attached to another group, e.g. it is a W tuted with one —OH. Substituent attached to the cyclic phosphoramidate, it is The term “haloalkyl and “alkylhalide' refers to an alkyl attached at the first carbon. 35 group substituted with one halo. Preferably, the halo is in the The term “alkynyl refers to unsaturated groups which 2-position. contain at least one carbon-carbon triple bond and includes The term “cyano” refers to -C=N. straight-chain, branched-chain and cyclic groups. Alkynyl The term “nitro” refers to - NO. groups may be optionally substituted. Suitable alkynyl The term “acylalkyl refers to an alkyl-C(O)-alk-, where groups include ethynyl. “1-alkynyl refers to alkynyl groups 40 “alk” is alkylene. where the triple bond is between the first and second carbon The term “aminocarboxamidoalkyl-” refers to the group atom. If the 1-alkynyl group is attached to another group, e.g. NR C(O) N(R)-alk- wherein R is an alkyl group or Hand it is a W substituent attached to the cyclic phosphoramidate, “alk” is an alkylene group. "Lower aminocarboxami it is attached at the first carbon. doalkyl-” refers to such groups wherein “alk” is lower alky The term “alkylene' refers to a divalent straight chain, 45 lene. branched chain or cyclic Saturated aliphatic group. The term "heteroarylalkyl refers to an alkyl group substi The term “acyloxy' refers to the estergroup —O C(O)R. tuted with a heteroaryl group. where R is H. alkyl, alkenyl, alkynyl, aryl, aralkyl, or alicy The term “perhalo' refers to groups wherein every C. H clic. bond has been replaced with a C-halo bond on an aliphatic or The term “aminoalkyl-” refers to the group NR-alk- 50 aryl group. Suitable perhaloalkyl groups include —CF and wherein “alk” is an alkylene group and R is selected from H. - CFC1. alkyl, aryl, aralkyl, and alicyclic. The term “guanidino” refers to both NR C(NR) NR The term “alkylaminoalkyl-” refers to the group alkyl-NR as well as —N=C(NR) here each R group is independently alk- wherein each “alk” is an independently selected alky selected from the group of —H, alkyl, alkenyl, alkynyl, aryl, lene, and R is H or lower alkyl. “Lower alkylaminoalkyl- 55 refers to groups where each alkylene group is lower alkylene. and alicyclic, all except—Hare optionally Substituted. The term “arylaminoalkyl-” refers to the group aryl-NR The term "amidino” refers to —C(NR) NR where each alk- wherein “alk” is an alkylene group and R is H. alkyl, aryl, R group is independently selected from the group of —H. aralkyl, and alicyclic. In “lower arylaminoalkyl-, the alky alkyl, alkenyl, alkynyl, aryl, and alicyclic, all except—Hare lene group is lower alkylene. 60 optionally substituted. The term “alkylaminoaryl-” refers to the group alkyl-NR The term “pharmaceutically acceptable salt' includes salts aryl- wherein “aryl' is a divalent group and R is H, alkyl, of compounds of formula I and its prodrugs derived from the aralkyl, and alicyclic. In “lower alkylaminoaryl-, the alky combination of a compound of this invention and an organic lene group is lower alkyl. or inorganic acid or base. Suitable acids include HC1. The term “alkoxyaryl-” refers to an aryl group substituted 65 The term “prodrug as used herein refers to any compound with an alkyloxy group. In “lower alkyloxyaryl-, the alkyl that when administered to a biological system generates a group is lower alkyl. biologically active compound as a result of spontaneous US 8,080,536 B2 11 12 chemical reaction(s), enzyme catalyzed chemical reaction(s), The phrase “together V and Z are connected via an addi and/or metabolic chemical reaction(s), or a combination of tional 3-5 atoms to form a cyclic group, optionally containing each. Standard prodrugs are formed using groups attached to one heteroatom, said cyclic group is fused to an aryl group functionality, e.g. HO HS HOOC RN— associ attached at the beta and gamma position to the Yadjacent to ated with the drug, that cleave in vivo. Standard prodrugs V includes the following: include but are not limited to carboxylate esters where the

group is alkyl, aryl, aralkyl, acyloxyalkyl, alkoxycarbony loxyalkyl as well as esters of hydroxyl, thiol and amines where the group attached is an acyl group, analkoxycarbonyl, aminocarbonyl, phosphate or Sulfate. The groups illustrated 10 are exemplary, not exhaustive, and one skilled in the art could prepare other known varieties of prodrugs. Such prodrugs of the compounds of formula I, fall within the scope of the present invention. Prodrugs must undergo some form of a chemical transformation to produce the compound that is 15 biologically active or is a precursor of the biologically active compound. In some cases, the prodrug is biologically active, usually less than the drug itself, and serves to improve drug efficacy or safety through improved oral bioavailability, phar The phrase “together V and W are connected via an addi macodynamic half-life, etc. The biologically active com tional 3 carbonatoms to forman optionally Substituted cyclic pounds include, for example, anticancer agents, antiviral group containing 6 carbon atoms and Substituted with one agents, and antibiotic agents. Substituent selected from the group consisting of hydroxy, The term “bidentate' refers to an alkyl group that is acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and ary attached by its terminal ends to the same atom to form a cyclic loxycarbonyloxy, attached to one of said additional carbon group. For example, propylene imine contains a bidentate 25 atoms that is three atoms from a Yadjacent to Vincludes the propylene group. following: The structure

R6 V 2H V 30 O N V / O Y O -P V / 3 V -P O-C-CH N V A. Y H R6 W 35 W H CH has a plane of symmetry running through the phosphorus oxygen double bond when R=R.V=W. W=H, and Vand The structure above has an acyloxy substituent that is three Ware either both pointing up or both pointing down. carbon atoms from a Y. and an optional Substituent, —CH, The term “cyclic 1',3'-propane ester”, “cyclic 1,3-propane on the new 6-membered ring. There has to be at least one ester”, “cyclic 1',3'-propanyl ester, and “cyclic 1,3-propanyl 40 hydrogen at each of the following positions: the carbon ester” refers to the following: attached to Z; both carbons alpha to the carbon labeled '3”; and the carbon attached to “OC(O)CH above. O The phrase “together W and Ware connected via an addi Vy \ 45 tional 2-5 atoms to form a cyclic group, optionally containing -P 2 0-2 heteroatoms, and V must be aryl, substituted aryl, het VY ) 3' eroaryl, or substituted heteroaryl' includes the following: The phrase “together V and Z are connected via an addi tional 3-5 atoms to form a cyclic group containing 5-7 ring 50 atoms, optionally containing 1 heteroatom, Substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbony loxy attached to a carbon atom that is three atoms from both Y groups adjacent to V” includes the following: The structure shown above (left) has an additional 3 carbon 55 atoms that forms a five member cyclic group. Such cyclic groups must possess the listed Substitution to be oxidized.

60 The structure above has V-aryl, and a spiro-fused cyclo propyl group for W and W". The term "phosphoramidite” refers to compounds attached OH via C, O, S, or N to the phosphorus in —P(YR)(YR) including cyclic forms, where Y is independently —O— or NR , 65 but where at least one Y is NR -. The term "phosphoramidate' refers to compounds attached via C, O, S, or N to the phosphorus in —P(O)(YR) US 8,080,536 B2 13 14 (YR), including cyclic forms, where Y is independently The term "enhancing refers to increasing or improving a —O— or NR , but where at least one Y is NR—. specific property. It includes the situation where originally The term “cyclic phosphoramidate' refers to phosphora there was no activity. midates where -P(O)(YR)(YR) is The term “liver specificity' refers to the ratio:

parent drug or a drug metabolite in liver tissue H O Y parent drug or a drug metabolite in blood, V/ urine, or other non-hepatic tissue -P V 10 Y H as measured in animals treated with the drug or a prodrug. The W W ratio can be determined by measuring tissue levels of the parent drug or drug metabolite(s) including the biologically The carbon attached to V must have a C-H bond. The 15 active drug metabolite or both at a specific time or may carbon attached to Z must also have a C-H bond. represent an AUC based on values measured at three or more The term "carbocyclic Sugar refers to Sugar analogs that time points. contain a carbon in place of the oxygen normally found in the The term “increased or enhanced liver specificity' refers to Sugar ring. It includes 5-membered rings Such as ribofurano an increase in the liver specificity ratio in animals treated with Syl and arabinofuranosyl Sugars wherein the ring oxygen is the prodrug relative to animals treated with the parent drug. replaced by carbon. The term "enhanced oral bioavailability” refers to an The term “acyclic Sugar refers to Sugars that lack a ring, increase of at least 50% of the absorption of the dose of the e.g. ribofuranosyl ring. An example is 2-hydroxyethoxym parent drug or prodrug (not of this invention) from the gas ethyl in place of the ribofuranosyl ring. trointestinal tract. More preferably it is at least 100%. Mea The term “L-nucleoside' refers to enantiomer of the natu 25 surement of oral bioavailability usually refers to measure ral B-D-nucleoside analogs. ments of the prodrug, drug, or drug metabolite in blood, The term “arabinofuranosyl nucleoside' refers to nucleo tissues, or urine following oral administration compared to side analogs containing an arabinofuranosyl Sugar, i.e. where measurements following systemic administration. the 2'-hydroxyl of ribofuranosyl Sugars is on the opposite face 30 The term “parent drug” refers to MH for phosphoramidates of the Sugar ring. where M is connected to —P(O)(YR)(YR) via oxygen, sulfur The term “dioxolane Sugar refers to sugars that contain an or nitrogen and to M-PO, when M is connected to -P(O) oxygen atom in place of the 3' carbon of the ribofuranosyl (YR)(YR) via carbon. For example, AZT can be thought of as Sugar. a parent drug in the form of MH. In the body AZT is first The term “fluorinated sugars' refers to sugars that have 1-3 35 phosphorylated to AZT-PO, and then further phosphory carbon-fluorine atoms. lated to form AZT-triphosphate, which is the biologically The term “P450 enzyme” refers to cytochrome enzymes active form. The parent drug form MH only applies when M that oxidize organic compounds and includes naturally occur is attached via N. S. or O. In the case of PMEA, the parent ring P450 isozymes, mutants, truncated enzymes, post-tran drug form is M-PO. Scriptase modified enzymes, and other variants. 40 The term “drug metabolite” refers to any compound pro The term “liver” refers to liver and to like tissues and cells duced in vivo or in vitro from the parent drug, or its prodrugs. that contain the CYP3A4 isozyme or any other P450 isozyme The term “pharmacodynamic half-life” refers to the time found to oxidize the phosphoramidates of the invention. after administration of the drug or prodrug to observe a dimi Based on Example F, we have found that prodrugs of formula nution of one half of the measured pharmacological response. VI and VIII are selectively oxidized by the cytochrome P450 45 Pharmacodynamic half-life is enhanced when the half-life is increased by preferably at least 50%. isoenzyme CYP3A4. According to DeWaziers et al. (J. The term “pharmacokinetic half-life” refers to the time Pharm. Exp. Ther, 253,387-394 (1990)), CYP3A4 is located after administration of the drug or prodrug to observe a dimi in humans in the following tissues (determined by immuno nution of one half of the drug concentration in plasma or blotting and enzyme measurements): 50 tissues. The term “therapeutic index' refers to the ratio of the dose ofa drug or prodrug that produces atherapeutically beneficial Tissues % of liver activity response relative to the dose that produces an undesired response Such as death, an elevation of markers that are Liver 100 55 Duodenum 50 indicative of toxicity, and/or pharmacological side effects. jejunum 30 The term "enhancing the level of the biologically active illeum 10 drug” refers to increasing the level of the biologically active colon <5 (only P450 isoenzyme found) drug in the liver relative to the level achieved after adminis stomach <5 esophagus <5 tration of the parent drug where the same mode of adminis kidney not detectable 60 tration is used for both the prodrug and the parent drug. The term “cancer expressing a P450 enzyme” refers to tumor cells having a specific activity, either with or without an Thus, “liver more preferably refers to the liver, duodenum, agent that induces the P450 activity in the tumor cells, of at jejunum, ileum, colon, stomach, and esophagus. Most pref least 5% of the liver specific activity. erably, liver refers to the liver organ. 65 The term “low levels of enzymes' refers to levels below The term “hepatic' refers to the liver organ cells and not that necessary to produce the maximal response of the bio cells from the duodenum, for example. logically active agent. US 8,080,536 B2 15 16 The term “inadequate cellular production” refers to levels Bvara U; 1-b-D-arabinofuranosyl-E-5-(2-bromovinyl) that are below that necessary to produce the maximal uracil (Sorivudine) response of the biologically active agent. For example, the E-5-(2-bromovinyl)-2'-deoxyuridine response may be viral titre. TFT: Trifluorothymidine (Trifluorothymidine) The term “sustained delivery” refers to an increase in the 5-propynyl-l-arabinosyluracil (Zonavir) period in which there is a prolongation of therapeutically CDG, carbocyclic 2'-deoxyguanosine effective drug levels due to the presence of the prodrug. DAPD: (-)-B-D-2,6-diaminopurine dioxolane The term “bypassing drug resistance” refers to the loss or FDOC: (-)-B-D-5-fluoro-1-2-(hydroxymethyl)-1,3-di partial loss of therapeutic effectiveness of a drug (drug resis oxolanelcytosine tance) due to changes in the biochemical pathways and cel 10 d4C: 3'-deoxy-2',3'-didehydrocytidine DXG; dioxolane guanosine lular activities important for producing and maintaining the FEAU; 2'-deoxy-2'-fluoro-1-b-D-arabinofuranosyl-5- biologically active form of the drug at the desired site in the ethyluracil body and to the ability of an agent to bypass this resistance FLG: 2',3'-dideoxy-3'-fluoroguanosine through the use of alternative pathways and cellular activities. 15 FLT: 3'-deoxy-3'-fluorothymidine The term “FBPase inhibitors' refers to compounds that FTC; (-)-cis-5-fluoro-1-2-(hydroxymethyl)-1,3-oxathi inhibit the human enzyme fructose 1,6-bisphosphatase with olan-5-ylcytosine an IC50 of at least 100 uM and lower glucose in a normal 5-yl-carbocyclic 2'-deoxyguanosine (BMS200.475) 18-hour fasted rat following a 100 mg/kg dose i.v. The bio 1-(4'-hydroxy-1',2'-butadienyl)cytosine (Cytallene) logically active FBPase inhibitors are M-PO, wherein M is Oxetanocin A; 9-(2-deoxy-2-hydroxymethyl-beta-D- connected via a carbon, or via an oxygen when MH is a erythro-oxetanosyl)adenine imidazole containing nucleoside analog. Oxetanocin G: 9-(2-deoxy-2-hydroxymethyl-beta-D- The term “biologically active drug or agent” refers to the erythro-oxetanosyl)guanine chemical entity that produces the biological effect. In this ddAPR: 2,6-diaminopurine-2',3'-dideoxyriboside invention, biologically active agents refers to M-PO, M-P 25 3TC; (-)-2',3'-dideoxy-3' thiacytidine; (2R,5S) 1-2-(hy (O )NHR, MPO, or MP.O." where M can be the droxymethyl)-1,3-oxathiolane-5-ylcytosine (Lamivudine) same M as in the parent drug or a metabolite. Cyclobut A: (+/-)-9-(1 beta,2 alpha,3 beta)-2,3-bis(hy The term “therapeutically effective amount” refers to an droxymethyl)-1-cyclobutyladenine amount that has any beneficial effect in treating a disease or Cyclobut G; (+/-)-9-(1 beta,2 alpha,3 beta)-2,3-bis(hy condition. 30 droxymethyl)-1-cyclobutyllguanine (Lobucavir) The term “decreased activity of the phosphorylating 5-fluoro-2'-deoxyuridine () enzyme” includes decreased production of the enzyme, muta dFdC: 2,2'-difluorodeoxycytidine () tions that lead to decreased efficiency (lower K, or V.), or araC., arabinosylcytosine () enhanced production of endogenous inhibitors of the bromodeoxyuridine enzyme. 35 IDU; 5-iodo-2'-deoxyuridine (Idoxuridine) The term “phosph(on)ate' refers to compounds attached CdA; 2-chlorodeoxyadenosine (Cytadribine) via C, O, S, or N to PO' or P(O)(NHR)O. The counterion F-ara-A; fluoroarabinosyladenosine () may be H' or a metal cation. ACV:9-(2-hydroxyethoxylmethyl)guanine (Acyclovir) The term “nucleoside' refers to a purine or pyrimidine GCV: 9-(1,3-dihydroxy-2-propoxymethyl)guanine (gan base, including analogs thereof, connected to a Sugar, includ 40 cyclovir) ing heterocyclic and carbocyclic analogs thereof. 9-(4-hydroxy-3-hydroxymethylbut-1-yl)guanine (Penci The following well known drugs are referred to in the clovir) specification and the claims. Abbreviations and common (R)-9-(3,4-dihydroxybutyl)guanine (Buciclovir) names are also provided. phosphonoformic acid (FoScarnet) araA; 9-b-D-arabinofuranosyladenine (Vidarabine) 45 PPA; phosphonoacetic acid AZT, 3'-azido-2',3'-dideoxythymidine (Zidovudine) PMEA: 9-(2-phosphonylmethoxyethyl) adenine (Ade d4T, 2',3'-didehydro-3'-deoxythymidine (Stavudine) fovir) ddI; 2',3'-dideoxyinosine (Didanosine) PMEDAP: 9-(2-phosphonylmethoxyethyl)-2,6-diami ddA; 2',3'-dideoxyadenosine nopurine ddC; 2',3'-dideoxycytidine (Zalcitabine) 50 HPMPC: (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl) L-ddC: L-2',3'-dideoxycytidine cytosine (Cidofovir) L-FddC: L-2',3'-dideoxy-5-fluorocytidine HPMPA: (S)-9-(3-hydroxy-2-phosphonylmethoxypropyl) L-dT; b-L-thymidine (NV-02B) adenine L-dC; b-L-2-deoxycytidine (NV-02C) FPMPA; 9-(3-fluoro-2-phosphonylmethoxypropyl) L-d4C: L-3'-deoxy-2',3'-didehydrocytidine 55 adenine L-Fd4C: L-3'-deoxy-2',3'-didehydro-5-fluorocytidine PMPA; 9-2-(R)-phosphonylmethoxy)propyladenine 3TC; (-)-2',3'-dideoxy-3'-thiacytidine: 2R,5'S(-)-1-2- (Tenofovir) (hydroxymethyl)oxathiolan-5-ylcytosine (Lamivudine) araT; 9-b-D-arabinofuranosylthymidine 1-b-D-ribofuranosyl-1,2,4-triazole-3-carboxamide (Rib FMdC; (E)-2'-deoxy-2'(fluoromethylene)cytidine avirin) 60 AICAR, 5-aminoimidazole-4-carboxamido-1-ribofurano FIAU; 1-(2-deoxy-2-fluoro-b-D-arabinofuranosyl)-5-io Syl douridine FIAC: 1-(2-deoxy-2-fluoro-b-D-arabinofuranosyl)-5-io DETAILED DESCRIPTION OF THE INVENTION docytosine BHCG: (+)-(1a,2b.3a)-9-2,3-bis(hydroxymethyl)cy 65 The invention is directed to the use of new cyclic phospho clobutyllguanine ramidate methodology which allows compounds to be effi FMAU; 2'-Fluoro-5-methyl-b-L-arabino-furanosyluracil ciently converted to phosph(on)ate containing compounds by US 8,080,536 B2 17 18 P450 enzymes found in large amounts in the liver and other invention to increase the therapeutic index of various drugs tissues containing these specific enzymes. This methodology which have side effects related to the amount of the drug or its can be applied to various drugs and to diagnostic imaging metabolites which are distributed in extrahepatic tissues. agents. More specifically, the invention is directed to the use Increased therapeutic index can also result from increased of prodrugs that undergo non-esterase-mediated hydrolysis liver levels of the biologically active agent and therefore reactions to produce MP(O)(NHR)O which is either bio greater efficacy relative to the parent drug administered at logically active or is transformed into the biologically active similar doses. agent M-PO, M-PO, or M-PO". Because highly In another aspect of the invention, prodrugs can be used to charged phosph(on)ate containing compounds are not readily deliver phosph(on)ates to tissues that generate Suboptimal absorbed in the gastrointestinal tract, this prodrug methodol 10 levels of phosph(on)ates and their associated higher phospho ogy can be used to enhance absorption after oral administra rylated metabolites due to poor substrate activity of the parent tion. compound and/or poor expression of enzymes used to phos In another aspect of the invention, this prodrug methodol phorylate the parent compound. Poor expression of the phos ogy can also be used to enhance the pharmacodynamic half phorylating enzyme occurs naturally or from down regulation life relative to the parent drug because the cyclic phosph(on) 15 of the phosphorylating enzyme following chronic adminis ates of the invention can avoid metabolism by enzymes which tration of the drug or other agents. metabolize the parent drug. Similarly, prodrugs of the inven In another aspect of the invention, novel phosphoramidite tion can enhance the pharmacodynamic half-life of the and phosphoramidate intermediates are described. phosph(on)ate-containing compound because the prodrug In another aspect, methods of preparing the cyclic phos avoids clearance pathways used to clear negatively-charged phoramidate prodrugs are described. compounds from the bloodstream. These aspects are described in greater detail below. In another aspect of the invention, this prodrug methodol Phosphoramidate Drugs: ogy can also be used to achieve Sustained delivery of the Compounds of the formula M-P(O)(NHR)O are useful parent drug because oxidation of the novel prodrugs proceeds compounds for binding to nucleotide binding sites, such as in the liver at different rates and therefore selection of pro 25 AMP-binding sites, and certain sites known to recognize drugs that oxidize slowly but at a rate Suitable for maintaining negatively charged compounds, e.g. carboxylic and phospho therapeutically effective levels will produce a sustained thera nic acids. Enzymes that catalyze the addition of water to a peutic effect. carbonyl compound, specifically a peptide carbonyl, an ester, The novel cyclic 1,3-propanylester methodology of the a ketone or an aldehyde, are of particular interest since these present invention may also be used to increase the distribution 30 enzymes recognize compounds that have a tetrahedral group of a particular drug or imaging agent to the liver which con containing a negatively charged oxygen. An example is the tains abundant amounts of the P450 isozymes responsible for Zinc metalloproteinase class of enzymes which add water oxidizing the cyclic 1.3-propanylester of the present inven across a peptide carbonyl using a zinc-assisted catalytic tion so that the free phosph(on)ate is ultimately produced. mechanism. These enzymes are inhibited by phosphorami Accordingly, this prodrug technology should prove useful in 35 dates, e.g. NEP 24.11 is inhibited by the natural product the treatment of liver diseases or diseases where the liver is Phosphoramidon. The prodrug strategy can provide a useful responsible for the overproduction of the biochemical end means for delivery of these compounds orally, or delivery of product such a glucose, cholesterol, fatty acids and triglycer certain compounds to the liver in order to achieve greater ides. Such diseases include viral and parasitic infections, liver efficacy or a greater therapeutic window. Enzymes inhibitors cancer, liver fibrosis, diabetes, hyperlipidemia, and obesity. 40 that may be suitable for delivery as prodrugs include certain In one aspect, preferably the disease is not diabetes. In addi phosphoramidates that inhibit NEP24. 11, collagenase, stro tion, the liver specificity of the prodrugs should also prove molysin, gelatinase, ACE, endothelin converting enzyme, useful in the delivery of diagnostic agents to the liver. High and metalloproteinases involved in matrix remodeling Such levels of the biologically active drug are achieved in tissues as occurs in Rheumatoid arthritis and osteoarthritis and in the expressing P450 enzymes capable of cleaving the prodrug 45 heart following an acute myocardial infarction, and tumor due to significant retention by these cells of the activated metastasis. prodrug, Subsequent prodrug cleavage intermediates as well Phosphonic Acids and Phosphoric Acids: as the biologically active drug. In addition, high levels of the Cleavage of the prodrug of formula I produces the phos biologically active drug are achieved relative to the levels phoramidate M-P(O)(NHR)O which in some cases is further achieved after administration of the parent drug when the 50 converted to the corresponding M-PO, via either chemical parent drug is poorly phosphorylated in the tissue or has a hydrolysis or enzyme catalyzed hydrolysis (phosphatases, relatively short pharmacokinetic half-life due to metabolism amidases). The product M-PO, can either be the drug or or other clearance mechanisms. itself may be further metabolized to produce higher order The use of certain prodrug analogs of cyclophosphamide phosphates, e.g. M-PO, or M-PO, via cellular kinases. are also envisioned for treatment of cancers of the GI tract 55 In some cases the intermediate M-P(O) (NHR)O may be (e.g. primary and secondary liver cancers) as well as cancers converted to higher order phosphates via initial conversion to where cytochrome P450s are introduced artificially, e.g. by M-H retrovirus and other well known gene therapy strategies as Enhancing Oral Bioavailability well as via implanted cells engineered to express cytochrome The invention pertains to certain cyclic phosphoramidates P45OS. 60 and their use to deliver, most preferably via oral administra These specific P450 enzymes are also found in other spe tion, a therapeutically effective amount of the corresponding cific tissues and cells, and thus this methodology may also be phosph(on)ate compounds, preferably to an animal in need used to increase the delivery of these agents to those tissues. thereof. Prodrugs of the invention enhance oral bioavailabil In another aspect of the invention, the characteristic that ity of certain drugs by changing the physical property of the most of the cyclic phosphoramidates of the present invention 65 drug as a consequence of the prodrug moiety and its substitu are metabolized in the liver to produce the phosph(on)ate can ents V, Z, W, and W". The active drug may be M-P(O) (NHR) enable the use of the prodrug methodology of the present O or M-PO. Alternatively, both may instead undergo US 8,080,536 B2 19 20 further metabolism, e.g. phosphorylation by kinases to form quickly by the kidneys. Such compounds probably will not M-PO, and/or M-PO, as the active drug substance. reach therapeutic levels in extrahepatic tissues. However, Compounds containing a free phosphonic acid or a phos there are some phosph(on)ates and phosphates that are able to phoric acid group generally exhibit poor (<2%) oral bioavail remain in circulation because they are not rapidly cleared by ability since these groups are highly charged at physiological the kidneys (e.g. NEP inhibitors). Such compounds are able pH. Charged groups on compounds with molecular weights to achieve therapeutically effective levels in blood and extra greater than 250 Daltons impede passive diffusion across cell hepatic tissues. Thus, in another aspect, oral delivery to extra membranes as well as absorption across the gut epithelial cell hepatic tissues of phosph(on)ates which are not cleared by the layer. Neutral prodrugs of these compounds have therefore kidneys is preferred. Thus, such parent drugs that act at sites been studied since these compounds would be more lipo 10 accessible to the free phosph(on)ic acid such as targets within philic and therefore more likely to exhibit improved intestinal the vasculature system, or enzyme or receptor targets that are permeability. Although many prodrug classes have been located on cell membranes which are exposed to the blood or reported, few have been found that exhibit properties suitable fluid in the intrastitial space are preferred. Targets suitable for for drug development. this aspect of the invention would be targets in which the The most common prodrug class, and the class almost 15 phosphonic acid administered parenterally (e.g. via i.v. injec exclusively used for clinical candidates, is the acyloxyalkyl tion) produces a pharmacological or biochemical response esters. These prodrugs, however, often exhibit only a modest expected to be useful for treating a disease condition. improvement in oral bioavailability due to poor aqueous sta Since the inhibitors exhibit poor oral bioavailability bility, poor stability to acidic/basic pH and rapid degradation (<2%), prodrugs of the type described in this invention could by esterases in the gastrointestinal tract (Shaw & Cundy, enhance the oral bioavailability and produce the phosphonic Pharm. Res. 10, (Suppl), S294 (1993). Another class of pro acid following prodrug cleavage in the liver. Suitable circu drugs are the bis-aryl prodrugs (e.g. DeLombert et al. J. Med. lating drug levels are expected after prodrug cleavage in the Chem. 37, 498 (1994)) which have shown in a few isolated liver, since the liver is known to excrete phosphonic acids into cases to provide good to modest improvements in oral bio the circulation. availability. The major limitation with this class of com 25 Oral bioavailability can also be calculated by comparing pounds is that the prodrug ester often is degraded to the the area under the curve of prodrug, parent drug, and/or monoacid rapidly in vivo but conversion to the parent drug metabolite concentration over time in plasma, liver, or other occurs only slowly (sometimes over days) if at all. tissue or fluid of interest following oral and i.v. administra The prodrugs of the invention exhibit improved properties tion. (Example P) that lead to enhanced oral bioavailability relative to the parent 30 For example, for drugs excreted renally in large amounts, drug. Several characteristics of the present cyclic phosphora oral bioavailability can be measured by comparing the midate prodrugs may contribute to their ability to enhance amount of the parent drug or metabolite excreted in the urine, oral bioavailability. First, the prodrugs exhibit good stability for example, after oral and i.v. administration of the prodrug. in aqueous solutions across a wide range of pHs. (Example A) A lower limit of oral bioavailability can be estimated by This pH stability prevents immediate hydrolysis in the mouth 35 comparison with the amount of parent drug excreted in the and GI tract prior to absorption. The pH stability can also be urine after administration of the prodrug (p.o.) and the parent beneficial during formulation of the product. drug (i.v.). Prodrugs of the invention show improved oral Second, the prodrugs are resistant to esterases and phos bioavailability across a wide spectrum of prodrugs, with phatases which are abundant in the gastrointestinal tract. many preferably showing increases of 1.5 to 10-fold in oral Because much of the administered dose remains intact in the 40 bioavailability. More preferably, oral bioavailability is G.I. tract, the compound remains less highly charged than a enhanced by at least 2-fold compared to the parent drug. free phosph(on)ate which means more of the drug can be Agents are known that inhibit CYP3A4 in the gastrointes absorbed by passive diffusion and enter the blood stream. tinal tract. For example, grapefruit juice is known to decrease (Example B) the activity putatively via a component in the grapefruit juice Last, the prodrug can limit metabolism at other sites on the 45 (e.g. Bergamottin: Chem Res Toxicol 1998, 11, 252-259) molecule. For example, the prodrugs of the invention elimi which results in the inactivation and/or downregulation of the nate metabolism of the purine base of araA by adenosine enzyme. Since only GICYP3A4 is affected, the oral absorp deaminase which is also abundant in the GI tract. (Example tion of the prodrugs of this invention should be enhanced. A B) The amine of ara A which is normally deaminated by the combination of agents that inhibit, inactivate, or downregu enzyme is protected by the cyclic phosphate moiety which is 50 late P450s that metabolize the prodrugs will have the effect of located on the 5'-ribofuranosyl hydroxyl. Reduced metabo enhancing their absorption and thereby making more prodrug lism at other sites of the molecule enables more of the drug to available for metabolism in the liver. The net effect of the circulate in the blood stream. Although not all of these prop combination would therefore be to deliver more drug to the erties will be applicable to every prodrug of every drug, each liver after oral absorption. of these properties can enable more drug to survive the GI 55 Sustained Delivery tract and be available for absorption. Drugs that undergo rapid elimination in vivo often require The novel prodrug strategy of the invention will be useful multiple administrations of the drug to achieve therapeuti for the oral delivery of drugs that act in the liver as well as cally-effective blood levels over a significant period of time. certain drugs that act on targets located in the vascular system Other methods are also available including Sustained release or extrahepatic tissues. Because the highest concentration of 60 formulations and devices. Prodrugs that breakdown over time CYP3A4 (the enzyme responsible for activating the novel can also provide a method for achieving Sustained drug levels. prodrugs) is in the liver, the biologically active drug has a high In general, this property has not been possible with the known concentration in the liver, relative to other tissues. In one phosph(on)ate prodrugs since either they undergo rapid aspect, parent drugs which act in the liver are preferred. hydrolysis in vivo (e.g. acyloxyalkyl esters) or very slow However, some of the phosph(on)ates are exported by 65 conversion (e.g. di-aryl prodrugs). organic anion transporters in the liver and enter the blood The cyclic phosphoramidates of the invention are capable stream. Many phosph(on)ates in the blood stream are cleared of providing Sustained drug release by providing a steady US 8,080,536 B2 21 22 release of the drug over time. For example, most phosphates tion slow the elimination of these drugs by masking the nega undergo dephosphorylation in vivo within minutes after sys tive charge until after oxidation and hydrolysis in liver and temic administration via the action of phosphatases present in like tissues. the blood. Similarly, acyloxyalkyl esters of these phosphates Enhanced Selective Delivery of Agents to the Liver and Like undergo rapid esterase-mediated hydrolysis to the phosphate Tissues which then is rapidly dephosphorylated. Some prodrugs of Delivery of a drug to the liver with high selectivity is the current invention may enable prolonged drug delivery desirable in order to treat liver diseases or diseases associated since many of the present prodrugs are oxidized slowly over with the abnormal liver properties (e.g. diabetes, hyperlipi time to the phosph(on)ate in the livers (Example S). Suitably demia) with minimal side effects. positioned, prodrug moieties can prevent or slow systemic 10 Analysis of the tissue distribution of CYP3A4 indicates metabolism associated with the parent drug. that it is largely expressed in the liver (DeWaziers et al., J. Sustained delivery of the drugs is achievable by selecting Pharm. Exp. Ther. 253: 387 (1990)). Moreover, analysis of the prodrugs of formula I that are hydrolyzed in vivo at a rate tissue homogenates in the presence of prodrugs indicates that capable of maintaining therapeutically effective drug levels only the liver homogenate cleaves the prodrug and to a lesser over a period of time. The cleavage rate of the drug may 15 degree homogenates from tissues in the upper G 1. Kidney, depend on a variety of factors, including the rate of the P450 brain, heart, Stomach, spleen, muscle, lung, and testes showed oxidation, which is dependent on both the substituents on the no appreciable cleavage of the prodrug (Example D). prodrug moiety, the stereochemistry of these Substituents and Evidence of the liver specificity was also shown in vivo the parent drug. Moreover, Sustained drug production will after both oral and i.v. administration of the prodrugs. Admin depend on the rate of elimination of the intermediate gener istration of a prodrug of araAMP i.V. gives liver levels of the ated after oxidation and the rate and availability of the pro bioactive drug araATP greater than achieved by an equivalent drug to the liver, which is the major site of oxidation. Identi dose of either ara A or araAMP. In contrast, the prodrug fails fication of the prodrug with the desired properties is readily to produce detectable amounts of the araA by-product araH, achieved by Screening the prodrugs in an assay that monitors which, as reported in the literature, is readily detected after the rate of drug production in the presence of the major P450 25 eitherara A or araAMP administration. Similarly, the prodrug enzyme involved in the metabolism, in the presence of liver achieves high liver levels without production of the metabo microsomes or in the presence of hepatocytes. These assays lite arah after oral administration. Since the prodrugs are are illustrated in Examples C, D, F, G, I, J, respectively. cleaved by liver abundant enzymes, oral administration may It is contemplated that prodrugs of the present invention enable even higher liver specificity via a first pass effect. could be combined to include, for example, one prodrug 30 The prodrugs described in this invention can be tailored which produces the active agent rapidly to achieve a thera such that the elimination step is fast and therefore the product peutic level quickly, and another prodrug which would is produced near the site of oxidation, which for these pro release the active agent more slowly over time. drugs is in the liver or other P450-expressing tissue/cells. Improved Pharmacodynamic Half-Life In some cases liver specificity will be achieved most opti The pharmacodynamic half-life of a drug can be extended 35 mally using prodrugs of highly reactive drugs, which after by the novel prodrug methodology as a result of both its production, act locally at a fast rate relative to diffusion out of ability to produce drug over a Sustained period and in some the liver. cases the longer pharmacokinetic half-life of the prodrug. Agents that induce P450 activity, e.g. CYP3A4 activity, are Both properties can individually enable therapeutic drug lev known. For example, rifampicin, glucocorticoids, phenobar els to be maintained over an extended period resulting in an 40 bital, erythromycin are known to enhance CYP3A4 activity improvement in the pharmacodynamic half-life. The pharma in rat and human livers following. P450 activity can be moni codynamic half-life can be extended by impeding the tored in humans by non-invasive methods e.g. via 14C metabolism or elimination pathways followed by the parent erythromycin breath test. These studies are useful in the iden drug. For some drugs, the prodrugs of the present invention tification of agents that activate CYP3A4 in humans. Accord are able to avoid the metabolism or elimination pathways 45 ingly, for prodrugs where drug delivery is limited by prodrug associated with the parent drug and thereby exist as the pro metabolism rate (e.g. rate of clearance of prodrug is fast drug for extended periods in an animal. High levels of the relative to rate of prodrug cleavage), agents such as rifampicin prodrug for an extended period result in Sustained production can be used in combination or as adjuncts or pre-treatments to of the parent drug which can result in an improvement in the enhance CYP3A activity in the liver and thereby to increase drug pharmacodynamic half-life. 50 liver drug levels. (Examples J and O) An example of the ability of the prodrug class to impede Prodrug Cleavage Mechanism metabolic pathways associated with the parent drug is shown The prodrugs of the current invention are simple, low by the araAMP prodrugs. In comparison to araAMP prodrugs molecular weight modifications of the drug which enable show no ara-hypoxanthine (“araH') which is the known liver-selective drug delivery on the basis of the their sensitiv metabolic byproduct of araA produced in e.g. plasma and the 55 ity to liver-abundant enzymes. The prodrug cleavage mecha gastrointestinal tract after oral or i.V./administration. nism is postulated to entail an oxidation and B-elimination AraAMP on the other hand is rapidly and nearly completely based on studies analyzing reaction requirements and prod converted to araH, which is produced by first dephosphory ucts. In some cases, M-P(O)(NR)O is further metabolized lation to araA via phosphatases followed by deamination of to M-PO' and M-PO. Prodrugs are stable to aqueous the base via adenosine deaminase. The prodrug moiety pre 60 Solution across abroad pH range and therefore do not undergo vents both dephosphorylation and deamination from occur a chemical cleavage process to produce the parent drug. In r1ng. addition the prodrugs are stable to esterases and blood pro A common route of elimination of phosph(on)ate drugs is teins. In contrast, the prodrugs are rapidly cleaved in the via the kidneys and a transporter that recognizes anionic presence of liver microsomes from rats (Example C) and compounds. Complete elimination of phosphonate and phos 65 humans (Examples D and F). The drug is also produced in phate containing drugs from the circulation often occurs only freshly isolated rat hepatocytes where it is detected as the minutes after drug administration. The prodrugs of this inven corresponding phosph(on)ate and/or biologically active US 8,080,536 B2 23 24 agent (Examples G and J). Moreover, when the parent drug is -continued an FBPase inhibitor, the production of the drug is supported OH by the ability of the prodrug to result in potent gluconeogen esis inhibition (Example I). Air Possible specific enzymes involved in the cleavage process were evaluated through the use of known cytochrome P450 (3) inhibitors (Example E). The studies indicate that the isoen O Y OH Zyme cytochrome CYP3A4 is responsible based on keto Y/ O conozole inhibition of drug formation. V 10 O The biologically active agent is detected in the liver fol O Y O lowing administration of drugs of formulae VI-VIII, shown -PV / Ha below: V O H 15 V H O VI -P - -P(OH) V V H O H O Y V / M-P V Although the esters in the invention are not limited by the Y above mechanisms, in general, eachester contains a group or VII atom Susceptible to microsomal oxidation (e.g. alcohol, ben Zylic methine proton), which in turn generates an intermedi ate that breaks down to the parent compound in aqueous 25 solution via B-elimination of the M-P(O)(YH). This species is either the active component or is further transformed via VIII chemical or enzymatic processes to M-PO, M-PO, or M-P.O.". Furthermore, although these specific prodrugs are 30 cleaved by CYP3A4, other prodrugs in the class may be substrates for other P450s. Small changes in structure are known to influence substrate activity and P450 preference. The identification of the isoenzyme(s) activating the prodrug Prodrugs of the following formulas are particularly pre is accomplished according to the procedure described in ferred. 35 Example B. Prodrugs of formula VI cleave to generate aryl vinylketone Alternatively, cyclic phosphoramidates can serve as a pro whereas prodrugs of formula VIII cleave to generate phenol drug of phosph(on)ates or higher order phosph(on)ates since (Example L). The mechanism of cleavage could proceed by intermediate phosphoramidates can be converted to MPO, the following mechanisms, shown for compounds where one MPO, or MP.O., via the action of phosphatases/ami Y is NR. 40 dases which can produce MH or MPO, which in turn can be phosphorylated by kinases to the biologically active. (1)

45 O O N V / -- --NR -- -3- -P V O Y O V O V / | O -P O -- -P(YH)(OH) + O O V 50 V / O -P OH \O

compound. 55 Increased Therapeutic Index (2) The prodrugs of this invention can significantly increase Air Air the therapeutic index ("TI) of certain drugs. In many cases, OH the increased TI is a result of the high liver specificity. For O Y O Y example, araA and araAMP are known to produce significant V / O V / 60 -P - - --P Ha systemic side effects and these side effects are associated with M M blood levels of the araA byproduct araH. Presumably the side O O effects area result of toxicities of araHorara A in extrahepatic O YH O O tissues (e.g. nerves) which produce e.g. the neuropathies -RV / -( - - -P(YH)(OH)| + 65 associated with the drug in man (>40% of patients receiving O Air ara A). Prodrugs of araA show a substantial shift in the liver (araATP)/urine (araH) ratio in comparison with araAMP. US 8,080,536 B2 25 26 Renal toxicity is a common toxicity associated with phos administration of CYP inducers (e.g. rifampicin, glucocorti phonic acids. The toxicity results from transport, e.g. via the coids, phenobarbital, erythromycin) which can enhance con organic anion transporters located on the basolateral mem version in the liver and thereby enable higher levels of the brane of the renal proximal tubule, of the negatively charged biologically active drug without increasing the dose (EX drug into e.g. tubular cells which then accumulate the drug to amples J and O). These benefits of the prodrugs enable suc high concentrations unless there is an equally efficient trans cessful therapy of liver diseases, such as primary and second port of the drug out of the cell via luminal transport mecha ary liver cancers as well as other liver diseases. nisms (e.g., anion exchange or facilitated diffusion). Many Byproduct Toxicity examples have been reported in the literature of nephrotoxic Prodrugs of the invention are also useful because they phosphonic acids, e.g. PMEA and HPMPA. The novel pro 10 generate byproducts that are either non-toxic or rapidly drug of PMEA shows only small amounts of PMEA in the detoxified. Some of the byproducts generated by prodrugs of urine relative to either PMEA orbisPOMPMEA at doses that the invention are not considered to be highly toxic at doses achieved comparable liver drug levels. expected to be used in the clinic, especially given that the Another common toxicity associated with phosphonic acid byproduct would be generated in the liver (e.g. phenol). Other drugs is gastrointestinal toxicity via in some cases GI ero 15 byproducts generated by the prodrugs of this invention, e.g. sions. Prodrugs of the current invention can decrease GI aryl vinylketone, can be considered as alkylating agents and toxicities, especially toxicities produced by direct action of therefore could produce either or both cytotoxicity and geno the drug on the GI tract after oral administration. Similar to toxicity. These toxicities, however, are eliminated for pro the kidney, gut epithelial cells have organic anion transporters drugs activated in the liver since: 1) the byproduct is produced which can result in high intracellular drug levels an cytotox at or near the site of prodrug activation; and 2) natural defense icity. Since the negatively charged phosph(on)ate is not mechanisms exist at the site of byproduct generation and are revealed until after absorption and cleavage in the liver, pro able to completely detoxify the byproduct. Other mecha drugs of this invention reduce gut toxicity. nisms may also assist in the detoxification of the byproduct, Severe toxicities are also associated with nearly all anti including reactions that lead to oxidation, reduction or trans cancer agents. In an effort to decrease these toxicities during 25 formation (e.g. Sulfation) of the byproduct. treatment of primary or secondary liver cancers, drugs are The primary defense system expected to protect cells from Sometimes administered directly into the portal artery in the prodrug byproduct involves the natural thiol glutathione order to increase liver drug exposure. Since oncolytic drugs and possibly an enzyme that catalyzes the addition of glu typically are associated with significant side effects, local tathione to electrophiles, i.e. glutathione S-transferase (GST). administration enables greater hepatic uptake and thereby 30 Glutathione and GST are always co-expressed with CYPs as decreased extrahepatic toxicities. To further increase liver part of a natural defense mechanism that protects the cell from uptake, chemoembolization is sometimes used in conjunction highly reactive oxygen species generated during CYP-medi with hepatic artery drug infusion. The high liver specificity of ated oxidation of organic compounds. CYPs are expressed in the prodrugs in the current invention Suggest that systemic certain cells (primarily hepatocytes) to oxidize organic com side effects will be similarly minimized by the novel prodrug 35 pounds and thereby assist in their elimination from the body. approach. Tissue distribution studies show that CYP3A4 as well as most Moreover, primary and secondary liver cancers are particu other P450s are expressed highest in the liver followed by the larly resistant to both chemotherapy and radiotherapy. small intestine. All other tissues have <2% of the liver Although the mechanism for the resistance is not completely CYP3A4 activity. Similarly, analysis of glutathione levels understood, it may arise from increased liver gene products 40 indicates that the liver (10 mM) and GI have nearly 1000-fold that lead to rapid metabolism and/or export of chemothera higher glutathione levels than other tissues. These levels of peutic agents. In addition, the liver, which is generally asso glutathione are still high even in diseased livers. ciated with Xenobiotic metabolism and generation of cyto Glutathione reacts rapidly with vinyl ketones, and any toxic intermediates, is equipped by nature with multiple alkylating agent, to generate a conjugate which is then elimi protective mechanism so that damage from these intermedi 45 nated via the bile or kidney. Alkylation of other proteins or ates are minimized. For example, the intracellular concentra nucleic acids in the cell is not expected in cells with glu tion of glutathione is very high in the liver relative to other tathione since glutathione is a much better nucleophile than tissues presumably so that intermediates capable of alkylat heteroatoms on the bases of nucleic acids and exists at much ing proteins and DNA are detoxified through a rapid intrac higher concentration than thiol groups on the Surface of pro ellular reaction. Consequently, the liver may be resistant to 50 teins. chemotherapeutic agents because of these mechanisms and By products such as acrolein can produce bladder toxicity therefore require higher than normal concentrations of the which these prodrugs can prevent by their ability to generate oncolytic agent to achieve Success. Higher liver concentra the byproduct within the hepatocyte. Examples M and N are tions require higher doses of the drug which commonly result useful for testing for byproduct toxicity, which is not in extrahepatic toxicities. 55 expected in the liver as long as hepatic glutahione levels Prodrugs of the current invention can be effective against remain >20% of normal. Glutathione is readily measured in these cancers because they enable higher levels of the bio hepatocytes as well as in vivo (Examples M and N). Cytotox logically active drug through a variety of mechanisms. For icity is detected in hepatocytes via testing of cell viability example, the high liver specificity can enable higher doses with trypan blue and by analyzing for elevation of live since the dose-limiting extrahepatic side effects are reduced. 60 enzymes. In vivo, liver toxicity is evaluated by analysis of Second, Sustained delivery of the prodrug can enable high liver enzyme elevation in the plasma. drug levels to be maintained over a Sustained period. Third, in Non-Mutagenic Prodrugs Some cases, the parent drug is poorly phosphorylated in the Prodrugs of the invention are elevated by a postulated target cells. The prodrugs of this invention can bypass this mechanism involving an initial oxidation followed by a step and deliver the phosph(on)ate which in some cases is 65 B-elimination reaction. In some cases, e.g. certain prodrugs of further metabolized to the biologically active agent. Last, the formula VI and formula VII, the biproduct of the reaction is an therapeutic index can sometimes be further enhanced by co C.f3-unsaturated carbonyl compound, e.g. vinyl phenyl US 8,080,536 B2 27 28 ketone for prodrugs where V=Ph, Z, W and W=H. Com efficacy is achieved in vivo because administration of the pounds can act as Michael acceptors and react with nucleo nucleoside fails to achieve sufficiently high levels of the bio philes via a Michael addition. Mutagenesis is observed with logically active phosphate. Often the failure to achieve high Some C-B-unsaturated ketones and certain toxicities arise levels of the phosphate is because the nucleoside is a poor from Michael addition adducts (e.g. acrolein produces blad Substrate of the enzymes that catalyze the phosphorylation der toxicities). The degree to which these activities limit the reaction. Other reasons include the low natural levels of the use of compounds of Formula VI is dependent on the severity phosphorylating enzyme in the target cell which may repre of the toxicity and the indicated disease. sent low natural expression levels or be a result of chronic Prodrugs that produce non-toxic and non-mutagenic therapy with the drug or another drug which results in enzyme biproducts are especially preferred for the treatment of 10 chronic diseases (e.g. diabetes). Frequently, it is difficult to down regulation. In some cases, drug resistance results from predict the mutagenic properties of a compound. For a decrease inactivity of the enzymes responsible for synthesis example, a number of acrylates have been shown to produce of a nucleoside monophosphate (e.g. kinases such as positive mutagenic responses as indicated by increased chro thymidylate kinase or enzymes in the biosynthesis pathway mosome aberrations and micronucleus frequencies in cul 15 of 5-fluoro-2'-deoxy UMP). In other cases nucleoside trans tured L5179Y mouse lymphoma cells (Dearfield et al., porters are downregulated leading to lower intracellular Mutagenesis 4,381-393 (1989)). Other acrylates, however, nucleoside drug concentration and therefore less nucleoside are negative in this test (J. Tox. Envir: Health, 34, 279-296 is available for phosphorylation. Similarly, increased expres (1991)) as well as in the Ames test and the CHO assay which sion of multidrug resistant gene product is postulated to measures newly induced mutations at the hypoxanthine-gua increase the export of nucleotides from cancer cells. Admin nine phosphoribosyltransferase (hgprt) locus (Mutagenesis 6. istration of the prodrug generates the monophosphate by a 77-85 (1991)). Phenyl vinyl ketone lacks teratogenic activity different pathway avoiding the pathways that cause the resis in rat embryos in culture Suggesting that it may not be tance to the parent drug. Thus, the prodrugs of the present mutagenic nor highly toxic (Teratology 39, 31-37 (1989)). invention can achieve a therapeutic effect in cells resistant to Since mutagenicity and toxicity are not highly predictable 25 the parent drug. Some drugs as the mono- or triphosphate properties, non-mutagenic prodrugs of formula I and their analogs are highly potent inhibitors of the target enzyme (e.g. associated by-products can be readily identified by conduct HBV polymerase) but are poorly effective in cells or in vivo ing well known in vitro and in vivo assays. For example, due to poor phosphorylation. (Example H) These drugs are compounds can be tested in non-mammalian cell assays Such especially preferred drugs since the prodrug strategy delivers as the Ames test, a fluctuation test in Kl. pneumoniae, a 30 the monophosphate. Frequently, the first phosphorylation of forward mutation assay with S. typhimurium, a chromosome the nucleoside is the rate-limiting step whereas phosphoryla loss assay in Saccharomyces cerevisiae, or a D3 recombino tion of the monophosphate to the triphosphate by mammalian genicity assay in Saccharomyces cerevisiae. Compounds can kinases is rapid and relatively insensitive to structural varia also be tested in mammalian cell assays such as the mouse tions. lymphoma cells assay (TK+/-heterozygotes of L5178Y 35 Ring-Substituted Cyclophosphamide Analogs for Treatment mouse lymphoma cells), assays in Chinese hamster ovary of Certain Cancers: cells (e.g. CHO/HGPRT assay), and an assay in rat liver cell Cyclophosphamide (CPA) and Ifosfamide (IF) are com lines (e.g. RL1 or RL4). Each of these assays can be con monly used oncolytic drugs that undergo initial activation in ducted in the presence of activators (e.g. liver microsomes) the liver via a P450 mechanism (CYP2B6 and CYP3A4) to which may be of particular importance to these prodrugs. By 40 intermediate 7. This intermediate exists in equilibrium with conducting these assays in the presence of the liver the ring-opened species (8), which is the penultimate inter microsomes, for example, the prodrug produces products, mediate for a variety of metabolic pathways, including a Such as phenol or vinyl ketone. The mutagenicity of the B-elimination reaction that generates the alkylating mustard 9 by-product is measured either directly or as a prodrug where and acrolein. The latter reaction sequence occurs via a non the results are compared to the parent drug alone. Assays in 45 enzymatic reaction and is thought to proceed with a t/2 of liver cell lines are a preferred aspect of the invention since ~30-40 minutes at neutral pH. these cells have higher glutathione levels, which can protect the cell from damage caused by a Michael acceptor, as well as greater levels of intracellular enzymes used to detoxify com OH pounds. For example, the liver contains reductases that with 50 O O HN Some by-products might result in reduction of the carbonyl. -PV / - O He- -PV / -- A variety of end points are monitored including cell N V a growth, colony size, gene mutations, micronuclei formation, O O mitotic chromosome loss, unscheduled DNA synthesis, DNA 7 elongation, DNA breaks, morphological transformations, 55 and relative mitotic activity. In vivo assays are also known that assess the mutagenicity -PV NH -ie- -PV NH and carcinogenicity of compounds. For example, a non-mam malian in vivo assay is the Drosophila sex-linked recessive lethal assay. Examples of mammalian in vivo assays include 60 the rat bone marrow cytogenetic assay, a rat embryo assay, as well as animal teratology and carcinogenicity assays. Kinase Bypass Intermediates 8 and 9 are relatively lipophilic and long Nucleosides are often converted in cells to mono-, di- and lived. As a consequence, a significant proportion of the acti tri-phosphates with the latter usually acting as the biologi 65 vated CPA “escapes” the liver and enters the general circula cally active species. In some cases, the triphosphate is a tion. Both the oncolytic activity and the drug-associated side potent inhibitor of the target enzyme but poor to modest effects are due to extra-hepatic breakdown. Uptake of the US 8,080,536 B2 29 30 intermediate by tumor cells and generation of the mustard PMEA: PMEDAP; HPMPC; HPMPA; FPMPA; PMPA; fos results in inhibition of cell proliferation. Similarly, myelosup carnet; and phosphonoformic acid. pression and decreased white cells are side effects associated In one preferred aspect, MH does not include ara.A. with CPA therapy that result from inhibition of extra-hepatic More preferred antiviral drugs include: cell proliferation. 5 araA; AZT, d4T, ddI; ddA; ddC; L-ddC; L FddC: L-d4C: CPA and IF are under investigation in conjunction with L-Fd4C: 3TC: ribavirin; FIAU; FIAC; L-FMAU; TFT. CDG: methods useful for the introduction of cytochrome P450s DAPD, FDOC; d4C; DXG: FEAU; FLG: FLT; FTC; 5-y1 artificially, e.g. by retrovirus and other well known gene carbocyclic 2' deoxyguanosine; cytallene; oxetanocin A; therapy strategies as well as via implanted cells engineered to oxetanocin G: Cyclobut A, Cyclobut G: araT; ACV, GCV: express cytochrome P450s for the treatment of cancer. The 10 penciclovir, PMEA: PMEDAP; HPMPC; HPMPA; PMPA; hope is that with cells that express the CYP near the tumor, a foscarnet. more local and therefore a potentially more beneficial therapy In one preferred aspect, MH does not include ara.A. is possible. Prodrugs of this invention have the advantage for Most preferred antiviral drugs include: treating primary and secondary liver cancers as well as for tumors with enhanced P450 activity since they generate the 15 penciclovir, active oncolytic agent more locally, i.e. with less “escape' FMAU; from the tissue containing the P450 activity. The reasons for DAPD; the greater local effect are due to differences in the prodrug FTC; cleavage intermediates which include: (1) a ring-opening Cyclobut G: reaction that is rapid and irreversible; and (2) the ring-opened ACV: product, which can be negatively charged (M-P(O) (NHR) GCV: O) and therefore unable to exit the cell via passive diffusion. PMEA: Types of Parent Drugs HPMPA; Various kinds of parents drugs can benefit from the prodrug 5-yl-carbocyclic 2' deoxyguanosine; methodology of the present invention. Parent drugs of the 25 ribavirin form MH, which are phosphorylated to become the biologi Preferred anticancer drugs include: cally active drug are well Suited for use in the prodrug meth dFdC: 2,2'-difluorodeoxycytidine (gemcitabine); odology of the present invention. There are many well known araC., arabinosylcytosine (cytarabine); parent drugs of the form MH which become biologically F-ara-A: 2-fluoroarabinosyladenosine (fludarabine); and active via phosphorylation. For example, it is well known that 30 CdA; 2-chlorodeoxyadenosine (). antitumor and antiviral nucleosides are activated through 2'-deoxy-5-iodouridine phosphorylation. These compounds include LdC. LdT, araA: Coformycin AZT, d4T, ddI; ddAddC; L-ddC; L FddC: L-d4C: L-Fd4C: 2'-deoxycoformycin 3TC: ribavirin; 5-fluoro-2'-deoxyuridine: FIAU; FIAC: Tiazofurin BHCG; L FMAU; BvaraU; E-5-(2-bromovinyl-2' deoxyuri 35 Ribavirin dine: TFT: 5-propynyl-1 arabinosyluracil; CDG, DAPD; 5-fluoro-2'deoxyuridine FDOC: d4C: DXG: FEAU; FLG: FLT: FTC; 5-yl-carbocyclic 9-(arabinofuranosyl)-2,6-diaminopurine 2' deoxyguanosine; oxetanocin A; oxetanocin G. Cyclobut A, 9-(2'-deoxyribofuranosyl)-2,6-diaminopurine Cyclobut G; dFdC: araC; bromodeoxyuridine; IDU; CdA: 9-(2'-deoxy-2'-fluororibofuranosyl)-2,6-diaminopurine FaraA; Coformycin, 2'-deoxycoformycin; araT; tiazofurin; 40 9-(arabinofuranosyl)-guanine ddAPR; 9-(arabinofuranosyl)-2.6 diaminopurine; 9-(2- 9-(2'-deoxyribofuranosyl)-guanine deoxyribofuranosyl)-2.6 diaminopurine; 9-(2'-deoxy 2 fluo 9-(2'-deoxy-2'-fluororibofuranosyl)-guanine roribofuranosyl)-2,6-diaminopurine; 9 (arabinofuranosyl) In one preferred aspect, MH does not include 5-fluoro-2'- guanine; 9-(2' deoxyribofuranosyl)guanine; 9-(2'-deoxy deoxyuridine. 2'fluororibofuranosyl)guanine: FMdC; 5.6 dihydro-5-azacy 45 In one aspect, preferably the compounds of formula I are tidine; 5-azacytidine; 5-aza 2'deoxycytidine: AICAR: ACV. not used in the treatment of diabetes. GCV: penciclovir; (R)-9-(3,4-dihydroxybutyl)guanine: cytal More preferred anticancer drugs include: lene; PMEA, PMEDAP, HPMPC, HPMPA, FPMPA, and dFdC: PMPA. araC. In one preferred aspect, the compounds of formula I does 50 FaraA; not include compounds where MH is ara A or 5-fluoro-2'- CdA; deoxyuridine. 5-fluoro 2'deoxyuridine: Preferred antiviral drugs include: LdC, LdT, araA; AZT, d4T, ddI; ddA; ddC; L-ddC. L. 5,6-dihydro-5-azacytidine: FddC; L-d4C; L-Fd4C: 3TC: ribavirin; FIAU; FIAC; BHCG: 55 5-azacytidine; and L-FMAU; BvaraU; E-5-(2 bromovinyl-2'-deoxyuridine: 5-aza-2'-deoxycytidine. TFT: 5-propynyl 1-arabinosyluracil; CDG: DAPD; FDOC: In one preferred aspect, ME does not include 5-fluoro-2'- d4C: DXG: FEAU; FLG: FLT. FTC;5-yl-carbocyclic 2'deox deoxyuridine. yguanosine; cytallene; oxetanocin A; oxetanocin G. Cyclobut Drugs containing a phosphonic acid (C-PO) moiety A, Cyclobut G; bromodeoxyuridine; IDU; araT; tiazofurin; 60 are also Suitable parent drugs advantageously used in the ddAPR; 9-(arabinofuranosyl)-2.6 diaminopurine; 9-(2- present invention. These drugs are biologically active either deoxyribofuranosyl)-2.6 diaminopurine; 9-(2'-deoxy 2 fluo in the form of MPO, MPO, or MP.O. Phosphonic roribofuranosyl)-2,6-diaminopurine; 9 (arabinofuranosyl) acids that are also Suitable for this prodrug delivery strategy guanine; 9-(2'deoxyribofuranosyl)guanine; 9-(2'-deoxy 2. include protease inhibitors that are useful for example as fluororibofuranosyl)guanine: FMdC; 5.6 dihydro-5-azacyti 65 antihypertensives, anticancer or anti-inflammatory agents. dine; 5-azacytidine. 5-aza 2'deoxycytidine: ACV, GCV: pen The novel prodrug methodology can be applied to NEP ciclovir; (R) 9-(3,4-dihydroxybutyl)guanine: cytallene; 5 inhibitors, (DeLambert et al., J. Med. Chem. 37:498 (1994)), US 8,080,536 B2 31 32 ACE inhibitors, endothelin converting enzyme inhibitors, 9-(3.3-Dimethyl-5-phosphonopentyl)guanine by Guida et al. purine nucleoside phosphorylase inhibitors, inhibitors of in J Med Chem 1994 Apr. 15:37(8): 1109-14. metalloproteases involved in tumor metastasis, and inhibitors Alanine Racemase Inhibitors of collagenase (Bird et al., J. Med. Chem. 37,158-169 (1994). DL-(1-Amino-2-propenyl)phosphonic acid by Vo-Quang et Moreover, phosphonic acids useful as NMDA antagonists al. in J Med Chem 1986 April; 29(4):579-81 which are useful for treating a variety of conditions, including Squalene Synthase Inhibitors stroke, head trauma, pain, and epilepsy. Other phosphonic 1-Hydroxy-3-(methylpentylamino)-propylidene-1,1-bis acids that could benefit from the prodrug strategies are phos phosphonic acid by Amin et al. in Arzneimittelforschung. phonic acids reported by Squibb that inhibit squalene syn 1996 August: 46(8):759-62. 10 BMS188494 is POM prodrug of BMS187745 by Dickson et thase, by Hoechst that are immunomodulators, by Merck that al. in J Med. Chem. 1996 Feb. 2: 39(3):661-4. are antidepressants, by Ciba-Geigy and Marion Merrel Dow Treatment of Cancer: that are immunosuppressants via inhibition of purine nucleo The prodrug strategy in the current invention encompasses side phosphorylase, and by Bristol-Myers Squibb, Gilead that several features that are advantageously used in cancer thera are antivirals. Certainantibiotics might be suitable, especially 15 pies. Many of the known anticancer drugs are nucleosides that antibiotics Such as D-alanine racemase inhibitors and fosfo undergo phosphorylation to the monophosphate which fre mycin and associated analogs. quently is further converted to the triphosphate. The prodrug The following compounds and their analogs can be used in strategy increases the effectiveness of these drugs because the the prodrug methodology of the present invention: prodrug is cleaved by liver-abundant enzymes and therefore NEP Inhibitors higher levels of the active metabolite are achieved in the liver (S)-3-N-2-(phosphonomethyl)amino-3-(4-biphenylyl) relative to extrahepatic tissues. The net effect is greater effi propionylaminopropionic acid by DeLombaert et al. in J cacy and/or a greater therapeutic index. In some cases, the Med. Chem. 1994 Feb. 18; 37(4):498-511 prodrug strategy also increases the effectiveness of these Collagenase Inhibitors drugs by bypassing well-known resistance mechanisms N,N-((R)-1-phosphonopropyl(-(S)-leucyl-(S)-phenylala 25 including mechanisms involved in the uptake, intracellular nine N-methyl amide by Bird et al. in J Med Chem 1994 biosynthesis of the active metabolite, export and metabolism Jan. 7: 37(1):158-69 of the monophosphate. Examples of preferred drug candi Angiotensin Coverting Enzyme Inhibitors dates that are specifically amenable to the strategy include, (IR)-1-(N-(N-acetyl-L-isoleucyl)-L-tyrosyl)amino-2-(4- e.g. dFdC, araC, F-ara, and CdA, and 5-fluoro-2'-deoxyuri hydroxyphenyl)ethyl-phosphonic acid by Hirayama et al. 30 dine (“5-FU). In one aspect, MH is not 5-FU. in IntJ Pept Protein Res 1991 July;38(1):20-4. Some prodrugs may result in some accumulation of the Endothelin Inhibitor monophosphate in cells. Certain monophosphates are useful CGS 26303 by DeLombaert et al., Biochem Biophy's Res for the treatment of cancers, e.g. monophosphates that are Commun 1994 Oct. 14; 204(1):407-12 potent inhibitors of thymidylate synthase (TS) and IMP dehy (S,S)-3-Cyclohexyl-2-5-(2,4-difluorophenyl)-2- 35 drogenase. Some TS inhibitors are reported to be moderately (phosphonomethyl)aminopent-4-ynoylaminopropi effective in treating liver cancers. For example, 5-FU and onic acid by Wallace et al., J Med Chem 1998 Apr. 23; 5-FdUMP are effective. These drugs, however, are plagued by 41 (9): 1513–23 drug resistance and severe side effects. To avoid the latter, (S,S)-2-5-(2-fluorophenyl)-2-(phosphonomethyl)amino 5-FU analogs are often delivered via the portal artery to pent-4-ynoyl-lamino-4-methylpentanoic acid 40 achieve the highest possible liver levels. Accordingly, (S,S)-2-5-(3-fluorophenyl)-2-(phosphonomethyl)amino 5-FdTMP and associated analogs are suitable targets for the pent-4-ynoyl------amino-4-methylpentanoic acid prodrug strategy. Other nucleosides Such as ribavirin, tiaZo NMDA/AMPA Antagonists furin and coformycin and analogs thereof are also suitable N-phosphonoalkyl-5-aminomethylduinoxaline-2,3-diones targets for the prodrug strategy. as described in Bioorg Med. Chem. Lett. 1999 Jan. 18; 45 Liver cancers are also very resistant to radiotherapy. One 9(2):249-54 method for enhancing radiotherapy is to administer radiosen 3-(2-carboxypiperazin-4-yl)-1-propenyl-1-phosphonic acid sitizers. Radiosensitizers such as 2'-deoxy-5-iodouridine are by Bespalov et al. in EurJ Pharmacol 1998 Jun. 26: 351 nucleosides Suitable for the prodrug technology since the (3):299-305 prodrugs will be cleaved by only P450 containing cells. After 2-(8,9-dioxo-2,6-diazabicyclo5.2.0non-1 (7)-en-2-yl)- 50 cleavage the monophosphate can be further phosphorylated ethylphosphonic acid D.L-(E)-2-amino-4-3H-propyl-5- to the triphosphate and trapped inside the cell making the cell phosphono-3-pentenoic acid more sensitive to radiotherapy. 6,7-dichloro-2(1H)-oxoquinoline-3-phosphonic acid by Treatment of Viral Infections: Desos et al. in J. Med. Chem. 1996 Jan. 5: 39(1):197-206. Drugs useful for treating viruses that infect the liver and cis-4-(phosphonomethyl)piperidine-2-carboxylic acid 55 cause liver damage, e.g. hepatitis virus strains, exhibit similar (CGS19755) properties to the anticancer nucleoside drugs in terms of Purine Nucleoside Phosphorylase Inhibitors efficacy, side effects and resistance. Prodrugs of these nucleo 7-(2-amino-1,6-dihydro-6-chloro-9H-purin-9-yl)-1,1-fluo sides would therefore be useful in treating hepatitis. In some roheptylphosphonic acid and cases, the drugs are already targeted for hepatitis (e.g. araA, 4-(5-amino-6,7-dihydro-7-oxo-3H-1,2,3-triazolo 4,5-d- 60 3TC, L-FMAU, FTC, BMS 200.475). The prodrugs of these pyrimidin-3-yl)butylphosphonic acid. compounds could enhance the efficacy, increase the therapeu 5-(2-amino-1,6-dihydro-6-oxo-9H-purin-9-yl)pentyl) tic index, improve the pharmacodynamic half-life and/or phosphinicomethylphosphonic acid by Kelly et al. in J bypass drug resistance. Prodrugs of other agents used to treat Med Chem 1995 Mar. 17:38(6):1005-14 viral infections other than hepatitis may also be made useful (2-2-(2-amino-1,6-dihydro-6-oxo-9H-purin-9-yl)methyl 65 by administration of the prodrugs of this invention since these phenyl]ethenyl)-phosphonic acid by Weibel et al. in Bio drugs are good antivirals (e.g. acyclovir, but useful for other chem Pharmacol. 1994 Jul.19; 48(2):245-52. viral infections because they are phosphorylated to the mono US 8,080,536 B2 33 34 phosphono by a viral kinase. The monophosphate is con of CYPactivity are known that might be useful in minimizing Verted to the biologically active triphosphate by mammalian unwanted drug metabolism. For example, grapefruit juice is kinases. Accordingly, delivery of the monophosphate using known to inactivate gastrointestinal CYP3A4 and to result in this clan of prodrugs enables treatment of hepatitis by drugs enhanced absorption of numerous drugs metabolized by normally used to treat other viral infections. 5 CYP3A4. CYP inhibitors are also known for many of the Use for the Delivery of Diagnostic Agents: CYP subfamilies that can be useful for attenuating unwanted Nucleoside diagnostic agents, e.g. uridine analogs with the drug metabolism while maintaining CYP activity important 5H substituted with Tc, are useful as the prodrugs of this for prodrug cleavage. For example, the CYP3A inhibitor TAO invention as liver diagnostic agents. These compounds are was used to modulate cyclophosphamide metabolism in vivo converted first to the monophosphate and then onto the triph- 10 in a manner that decreased the formation of toxic metabolites osphate in cells that contain P450 activity, specifically that do not contribute to its antitumor activity. CYP3A4 activity. Since nearly all of the activity is in the liver, Methods for Monitoring Patient P450 Activity diagnostic agents of this type will primarily be metabolized in CYP activity is known to exhibit significant differences the liver and accumulate in cells that metabolize the prodrug. across individuals. The range for CYP3A4 is 5- to 20-fold Accordingly, liver tumors that have no CYP3A4 activity or 15 although most individuals are within a 3-fold range. Modest tumors such as hepatocellular carcinomas which have decreases are noted for individuals with liver disease (30 approximately 50% of the normal activity may be differenti 50%) or advanced age (25-50%). Differences for gender are ated from normal liver tissue. even more modest (<25%). Methods for phenotyping an indi Agents Used to Modulate CYP Activity vidual’s CYP activity are known and could be useful in pre A variety of methods may be used to enhance the in vivo dicting who should receive drugs that modulate CYPactivity. activity of compounds of formula I. For example, various Evasive procedures include liver biopsy. Non evasive proce drugs are known that enhance cytochrome P450(CYP) activ dures have been reported, including an “erythromycin breath ity. Enhancement frequently entails increased gene transcrip test” which is based on the exhalation of 14CO2 generated tion. Four families of CYPs are particularly susceptible to from the CYP3A-mediated N-demethylation of radiolabeled induction, namely CYP1-4. Induction is purportedly via 25 erythromycin (iv). (Watkins, Pharmacogenetics 4, 171-184 receptors that are activated by various xenobiotics. For (1994)). example, CYP1 gene activation frequently involves activa Gene Therapy tion of the Ah receptor by polycyclic aromatic hydrocarbons. Introduction into tumor cells genes that encode for CYP2-4 are activated via orphan nuclear receptors. Data Sug enzymes not normally expressed represents a new therapeutic gests that the nuclear receptor CAR (constitutively Active 30 strategy for increasing the therapeutic effectiveness of anti Receptor) is responsible for phenobarbital CYP activation, cancer . The general strategy entails expres especially CYP2 genes. The pregnane nuclear receptors sion of an enzyme that catalyzes the breakdown of a prodrug (PXR or PAR or SXR) are thought to activate CYP3A genes ofan anticancer drug thereby localizing the drug in or near the whereas the PPAR (peroxisome proliferator activate recep tumor mass and limiting exposure elsewhere. The strategy tor) is linked to CYP4 gene activation. All three xenobiotic 35 has been demonstrated using the HSV-TK gene wherein the receptors are highly expressed in the liver which accounts for thymidylate kinase specifically expressed in the transfected the liver specificity of the P450 gene induction. cells activates ganciclovir to the monophosphate which is Xenobiotics known to induce CYP3 genes include phe then converted by other kinases to the tumor cell killing nobarbital, a variety of steroids, e.g. dexamethasone, antibi triphosphate. A similar strategy uses the bacterial cytosine otics, e.g. rifampicin, and compounds such as pregnenolone 40 deaminase gene for conversion of 5- to 5-fluoro 16a carbonitrile, phenyloin, carbamazepine, phenylbutaZone, cytosine. Other genes have been considered including car etc. A variety of methods are known that enable identification boxypeptidase G2, nitro reductase, purine nucleoside phos of xenobiotics that induce P450s, including a reporter gene phorylation, etc. In addition, CYP gene transfer has been assay in HepG2 cells (Ogg et al., Xenobiotica 29, 269-279 explored as a way to enhance the chemotherapeutic effect of (1999). Other inducers of the CYP3A subfamily are known 45 cyclophosphamide and ifosfamide, two drugs known to be that act at the post-transcriptional level either by mRNA or activated by CYPs. For example, human breast cancer cells protein stabilization, e.g. clotrimazole, TA and erythromycin. were sensitized by transfection with the CYP2B1 gene (Chen Compounds known to induce CYPs or identified in in vitro et al., Cancer Research, 56, 1331 1340 (1996)). The advan assays are then used to enhance CYP activity in vivo. For tage of this strategy relative to the HSV-TK gene strategy is example, CYP activity is monitored in rats pre-treated with 50 that the product of the CYP catalyzed oxidation readily dif CYP modulators by e.g. evaluating liver microsomes over a fuses outside of the tumor cell and into nearby cells. In con period of time to determine the optimal pre-treatment period, trast to monophosphate products of the HSV-TK strategy, the dose and dosing frequency. Rats with enhanced CYPactivity, product can enter cells that are not in cell-cell contact and especially the CYP activity responsible for activation of the therefore produce a more widespread tumor killing effect prodrugs (e.g. CYP3A4), are then treated with compounds of 55 (Chen and Waxman, Cancer Research, 55, 581-589 (1995)). formula I. Enhanced CYP activity can then lead to enhanced Compounds of formula I can be made more effective by prodrug conversion and liver specificity. For example, using gene therapy to introduce the gene that encodes the enhanced metabolism of cyclophosphamide was found with CYP specifically involved in prodrug cleavage. The specific pre-treatment with phenobarbital (Yu et al., J. Pharm. Exp. CYP that breaks down the prodrug is readily determined Ther. 288,928-937 (1999). 60 using some or all of the following steps: 1) demonstrate In some cases, enhanced CYP activity may lead to prodrug cleavage using human microsomes; 2) classify the unwanted drug metabolism. For example, enhanced activity Subfamily by comparing activity with microsomes induced of CYPs not involved in prodrug activation can result in with various subfamily specific inducers (e.g. CYP3 enzymes increased drug metabolism and therefore decreased efficacy. are induced with a variety of steroids, e.g. dexamethasone, In addition, increased CYP activity in other tissues, e.g. 65 antibiotics, e.g. rifampicin, and compounds such as preg CYP3A4 in the gastrointestinal tract, could result in nenolone-16a carbonitrile, phenyloin, carbamazepine, phe decreased prodrug absorption and liver drug levels Inhibitors nylbutazone, etc.; 3) identify the CYP or CYPs responsible US 8,080,536 B2 35 36 for prodrug activation by using known CYP subfamily spe CHRN, -CHaryl, -CH(aryl)OH, -CH(CH=CR) cific inhibitors (e.g. troleandomycin, erythromycin, keto OH, -CH(C=CR)OH, -R, NR, OCOR, conazole and gestodene) and/or by using neutralizing anti OCOR, SCOR, SCOR, NHCOR, bodies; 4) confirm CYP subfamily by demonstrating turnover —NHCO.R. —CH-NHaryl, -(CH), OR, and via the recombinant enzyme. —(CH.). SR': Genes are introduced to the tumor using a Suitable vector p is an integer 2 or 3: (e.g. retroviral vectors, adenoviral vectors) or via direct DNA with the provisos that: injection. P450 activity can also be introduced into or near the a) V, Z, W, Ware not all-H; and tumor mass using cells engineered to express P450s. Pre b) when Z is R, then at least one of V, W, and W is not ferred are encapsulated cells (e.g. Lohr et al., Gene Therapy 5: 10 —H, alkyl, aralkyl, or alicyclic; 1070 (1998)) The compounds of formula I are then intro R’ is selected from the group consisting of R and —H: duced following significant enhancement of the CYPactivity R is selected from the group consisting of alkyl, aryl, in the tumor. alicyclic, and aralkyl, Especially preferred are those prodrugs disclosed in the R is selected from the group consisting of H, and lower invention that are converted to the parent phosph(on)ate in 15 alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl and lower acyl: cells and tissues, especially hepatocytes and liver, as indi R" is selected from the group consisting of H, and lower cated by measurement of the intracellular drug metabolites in acyl: hepatocytes using the procedure described in Examples G each Y is independently selected from the group consisting and J. of-O-, - NR with the proviso that at least one Y is Preferred Compounds NR ; The compounds of the invention are substituted 6-mem Mis selected from the group that attached to PO, P.O., bered cyclic 1.3 propane prodrugs of certain phosph(on)ates PO, or P(O)(NHR)O is a biologically active agent, but is as represented by Formula I: not an FBPase inhibitor, and is attached to the phosphorus in 25 formula I via a carbon, oxygen, Sulfur or nitrogen atom; with the provisos that: 1) M is not NH(lower alkyl). —N(lower alkyl), NH H O. Y (lower alkylhalide), N(lower alkylhalide), or - N(lower V / alkyl) (lower alkylhalide); and M-P Z V 30 2) R is not lower alkylhalide; Y H and pharmaceutically acceptable prodrugs and salts thereof. W. W In general, preferred substituents, V. Z. W. and W" of for mula I are chosen such that they exhibit one or more of the wherein: 35 following properties: V, W, and W are independently selected from the group (1) enhance the oxidation reaction since this reaction is consisting of —H, alkyl, aralkyl, alicyclic, aryl, Substituted likely to be the rate determining step and therefore must aryl, heteroaryl, Substituted heteroaryl, 1-alkenyl, and 1-alky compete with drug elimination processes. nyl; or (2) enhance stability in aqueous solution and in the pres together V and Z are connected via an additional 3-5 atoms 40 ence of other non-P450 enzymes; to form a cyclic group containing 5-7 ring atoms, optionally 1 (3) enhance cell penetration, e.g. Substituents are not heteroatom, Substituted with hydroxy, acyloxy, alkoxycarbo charged or of high molecular weight since both properties can nyloxy, oraryloxycarbonyloxy attached to a carbon atom that limit oral bioavailability as well as cell penetration; is three atoms from bothY groups attached to the phosphorus: (4) promote the B-elimination reaction following the initial O 45 oxidation by producing ring-opened products that have one or together V and Z are connected via an additional 3-5 atoms more of the following properties: to form a cyclic group, optionally containing 1 heteroatom, a) fail to recyclize; said cyclic group is fused to an aryl group at the beta and b) undergo limited covalent hydration; gamma position to the Yadjacent to V. c) promote B-elimination by assisting in the proton abstrac together V and Ware connected via an additional 3 carbon 50 tion; atoms to forman optionally substituted cyclic group contain d) impede addition reactions that form stable adducts, e.g. ing 6 carbon atoms and Substituted with one Substituent thiols to the initial hydroxylated product or nucleophilic selected from the group consisting of hydroxy, acyloxy, addition to the carbonyl generated after ring opening; alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycar and bonyloxy, attached to one of said additional carbonatoms that 55 e) limit metabolism of reaction intermediates (e.g. ring is three atoms from a Y attached to the phosphorus: opened ketone); together Zand Ware connected via an additional 3-5 atoms (5) lead to a non-toxic and non-mutagenic by-product with to form a cyclic group, optionally containing one heteroatom, one or more of the following characteristics. Both properties and V must be aryl, substituted aryl, heteroaryl, or substituted can be minimized by using substituents that limit Michael heteroaryl; 60 additions, e.g.: together W and W are connected via an additional 2-5 a) electron donating Z groups that decrease double bond atoms to form a cyclic group, optionally containing 0-2 het polarization; eroatoms, and V must be aryl, Substituted aryl, heteroaryl, or b) W groups that sterically block nucleophilic addition to substituted heteroaryl; the B-carbon; Z is selected from the group consisting of —CHROH, 65 c) Z groups that eliminate the double bond after the elimi CHROC(O)R, CHROC(S)R, CHROC(S)OR, nation reaction either through retautomerization CHROC(O)SR, CHROCOR, OR, SR, (enol->keto) or hydrolysis (e.g. enamine); US 8,080,536 B2 37 38 d) V groups that contain groups that add to the C.B-unsat In another aspect, it is preferred when togetherV and Ware urated ketone to form a ring; connected via an additional 3 carbonatoms to forman option e) Z groups that form a stable ring via Michael addition to ally substituted cyclic group containing 6 carbon atoms and double bond; and monosubstituted with one substituent selected from the group f) groups that enhance detoxification of the by-product by consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylth one or more of the following characteristics: iocarbonyloxy, and aryloxycarbonyloxy attached to one of (i) confine to liver; and said additional carbon atoms that is three atoms from a Y (ii) make Susceptible to detoxification reactions (e.g. attached to the phosphorus. In Such compounds, it is more ketone reduction); and preferred when togetherV and W form a cyclic group selected (6) capable of generating a pharmacologically active prod 10 from the group consisting of —CH2—CH(OH)—CH2—, uct. —CHCH(OCOR)-CH , and —CHCH(OCO)R)— Suitable alkyl groups include groups having from 1 to CH2—. about 20 carbon atoms. Suitable aryl groups include groups Another preferred V group is 1-alkene. Oxidation by P450 having from 1 to about 20 carbon atoms. Suitable aralkyl 15 enzymes is known to occur at benzylic and allylic carbons. groups include groups having from 2 to about 21 carbon In one aspect, preferred V groups include —H, when Z is atoms. Suitable acyloxy groups include groups having from 1 CHR-OH, CHOCOR, or CHOCOR. to about 20 carbon atoms. Suitable alkylene groups include In another aspect, when V is aryl, substituted aryl, het groups having from 1 to about 20 carbon atoms. Suitable eroaryl, or substituted heteroaryl, preferred Z groups include alicyclic groups include groups having 3 to about 20 carbon OR, SR, R, NR, OCOR, OCOR, atoms. Suitable heteroaryl groups include groups having SCOR, SCOR, NHCOR, NHCOR, from 1 to about 20 carbonatoms and from 1 to 4 heteroatoms, -(CH2), OR, and -(CH2). SR. More preferred Z groups preferably independently selected from nitrogen, oxygen, include OR, R, OCOR, OCOR, NHCOR, and Sulfur. Suitable heteroaliacyclic groups include groups —NHCO.R. —(CH.) OR', and (CH), SR''. Most having from 2 to about twenty carbon atoms and from 1 to 5 25 preferred Z groups include —OR. H. —OCOR, heteroatoms, preferably independently selected from nitro OCOR, and NHCOR. gen, oxygen, phosphorous, and Sulfur. Preferred W and W groups include H. R., aryl, substituted In compounds of formula I, preferably one Y is —O—and aryl, heteroaryl, and substituted aryl. Preferably, W and W. one Y is NR -. When only one Y is NR , preferably are the same group. More preferred is when W and Ware H. the Y closest to W and W" is —O—. In another aspect, pref 30 In one aspect, prodrugs of formula VI which, in addition, erably the Y closest to V is —O—. are preferred: In another aspect, both Y groups are NR . More preferred are compounds wherein one Y is —O , and VI V V, W, and W are independently selected from the group 35 H consisting of —H, alkyl, aralkyl, alicyclic, aryl, Substituted O Y V / aryl, heteroaryl, Substituted heteroaryl, 1-alkenyl, and 1-alky M-P nyl, or V together V and Ware connected via an additional 3 carbon Y atoms to forman optionally substituted cyclic group contain 40 ing 6 carbon atoms and Substituted with one Substituent wherein selected from the group consisting of hydroxy, acyloxy, V is selected from the group consisting of aryl, Substituted alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycar aryl, heteroaryl, and substituted heteroaryl. More preferred bonyloxy, attached to one of said additional carbonatoms that are Such compounds where M is attached to the phosphorus is three atoms from a Y attached to the phosphorus. 45 via an O or Natom. Especially preferred are 3,5-dichlorophe More preferred are such compounds where V is selected nyl, 3-bromo-4-fluorophenyl, 3-chlorophenyl, 3-bromophe from the group consisting of aryl, Substituted aryl, heteroaryl, nyl. Preferably Y is —O—. Particularly preferred aryl and and substituted heteroaryl. Substituted aryl groups include phenyl and Substituted phe More preferred V groups of formula VI are aryl, substituted nyl. Particularly preferred heteroaryl groups include mono aryl, heteroaryl, and substituted heteroaryl. Preferably, one Y 50 cyclic Substituted and unsubstituted heteroaryl groups. Espe is —O—. Particularly preferred aryl and substituted aryl cially preferred are 4-pyridyl and 3-bromopyridyl. groups include phenyl, and phenyl Substituted with 1-3 halo In one aspect, the compounds of formula VI preferably gens. Especially preferred are 3,5-dichlorophenyl, 3-bromo have a group Z which is H. alkyl, alicyclic, hydroxy, alkoxy, 4-fluorophenyl, 3-chlorophenyl, and 3-bromophenyl. OC(O)R, OC(O)OR, or NHC(O)R. Preferred are such It is also especially preferred when V is selected from the 55 groups in which Zdecreases the propensity of the by-product, group consisting of monocyclic heteroaryl and monocyclic vinylaryl ketone to undergo Michael additions. Preferred Z Substituted heteroaryl containing at least one nitrogen atom. groups are groups that donate electrons to the vinyl group Most preferred is when such heteroaryl and substituted het which is a known strategy for decreasing the propensity of eroaryl is 4-pyridyl, and 3-bromopyridyl, respectively. C.f3-unsaturated carbonyl compounds to undergo a Michael In another particularly preferred aspect, one Y group is 60 addition. For example, a methyl group in a similar position on —O—, Z. W. and Ware H, and V is phenyl substituted with acrylamide results in no mutagenic activity whereas the 1-3 halogens. Especially preferred are 3,5-dichlorophenyl, unsubstituted vinyl analog is highly mutagenic. Other groups 3-bromo-4-fluorophenyl, 3-chlorophenyl, and 3-bromophe could serve a similar function, e.g. Z-OR'', NHAc, etc. nyl. Other groups may also prevent the Michael addition espe It is also especially preferred when V is selected from the 65 cially groups that result in removal of the double bond alto group consisting of heteroaryland Substituted heteroaryl. gether such as Z= OH, OC(O)R, OCOR, and NH, Most preferred is when such heteroaryl is 4-pyridyl. which will rapidly undergo retautomerization after the elimi US 8,080,536 B2 39 40 nation reaction. Certain W and W groups are also advanta heteroaryl, substituted heteroaryl, and Z is selected from the geous in this role since the group(s) impede the addition group consisting of H, OR, and NHCOR. More pre reaction to the B-carbon or destabilize the product. Another ferred are such compounds where Z is —H. Preferably, such preferred Z group is one that contains a nucleophilic group compound have Mattached via oxygen. Most preferred are capable of adding to the C.B-unsaturated double bond after Such compounds where oxygen is in a primary hydroxyl the elimination reaction i.e. (CH),SH or (CH)OH where p is 2 or 3. Yet another preferred group is a group attached to V group. Also more preferred, are those compounds where V is which is capable of adding to the C.f3-unsaturated double phenyl or substituted phenyl. bond after the elimination reaction: In one aspect, tumor cells expressing a P450 enzyme can be treated by administering a cyclophosphamide analog selected 10 from the group consisting of VI

V H O Y 15 V / R'R''N-P Y H In another aspect, prodrugs of formula VII are preferred: W W

wherein: O Y VII V/ V, W, and W are independently selected from the group M-P Z consisting of —H, alkyl, aralkyl, alicyclic, aryl, Substituted V aryl, heteroaryl, Substituted heteroaryl, 1-alkenyl, and 1-alky Y H 25 nyl; or togetherV and Z are connected via an additional 3-5 atoms wherein to form a cyclic group containing 5-7 ring atoms, optionally 1 Z is selected from the group consisting of: heteroatom, Substituted with hydroxy, acyloxy, alkoxycarbo CHROH, CHROC(O)R’, CHROC(S)R, nyloxy, oraryloxycarbonyloxy attached to a carbon atom that CHROCOR, CHROC(O)SR, CHROC(S)OR, 30 and —CHaryl. Preferably, M is attached to the phosphorus is three atoms from both Ygroups attached to the phosphorus: via a nitrogen. More preferred groups include —CHROH, O CHR’OC(O)R, and CHROCOR. Preferably R is H. togetherV and Z are connected via an additional 3-5 atoms In another aspect, prodrugs of formula VIII are preferred: to form a cyclic group, optionally containing 1 heteroatom, 35 said cyclic group is fused to an aryl group at the beta and gamma position to the Y adjacent to V: VIII together V and Ware connected via an additional 3 carbon O. Y atoms to forman optionally substituted cyclic group contain M-P D Z ing 6 carbon atoms and Substituted with one Substituent 40 selected from the group consisting of hydroxy, acyloxy, VY )) alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycar bonyloxy, attached to one of said additional carbonatoms that wherein is three atoms from a Y attached to the phosphorus: Z is selected from the group consisting of —OH, - OC together Zand Ware connected via an additional 3-5 atoms (O)R, OCOR, and OC(O)SR; 45 to form a cyclic group, optionally containing one heteroatom, D" and Dare independently selected from the group con and V must be aryl, substituted aryl, heteroaryl, or substituted sisting of H, alkyl, -OH, and —OC(O)R; with the pro heteroaryl; viso that at least one of D and D are - H. Preferably Y is together W and W are connected via an additional 2-5 —O—. An especially preferred Z'group is OH. atoms to form a cyclic group, optionally containing 0-2 het In one preferred embodiment, W' and Z are —H, W and V 50 eroatoms, and V must be aryl, Substituted aryl, heteroaryl, or are both the same aryl, substituted aryl, heteroaryl, or substi substituted heteroaryl; tuted heteroaryl, and both Y groups are the same —NR , Z is selected from the group consisting of —CHROH, Such that the phosphonate prodrug moiety: CHROC(O)R, CHROC(S)R, CHROC(S)OR, CHROC(O)SR, CHROCOR, OR2, SR, 55 —CHRN, -CHaryl, -CH(aryl)OH, -CH(CH=CR) V OH, CH(C=CR)OH, R°, NR, OCOR, O Y OCOR, SCOR, SCOR, NHCOR, V/ —NHCO.R. —CH-NHaryl, -(CH), OR, and -P V —(CH), SR': Y p is an integer 2 or 3: with the provisos that: W a) V, Z, W, Ware not all-H; and b) when Z is —R, then at least one of V, W, and W is not has a plane of symmetry through the phosphorous-oxygen —H, alkyl, aralkyl, or alicyclic; double bond. 65 R’ is selected from the group consisting of Rand-H; In another preferred embodiment, W and W are H, V is R is selected from the group consisting of alkyl, aryl, selected from the group consisting of aryl, Substituted aryl, alicyclic, and aralkyl, US 8,080,536 B2 41 42 R' is selected from the group consisting of H. lower MPO, or MP.O." may be useful in treating diseases 2-haloalkyl, and lower alkyl; where the target is outside the liver in tissues or cells that can R’ is selected from the group consisting of H, and lower oxidize the prodrug. acyl: Other preferred M groups include drugs useful in treating R" is lower 2-haloalkyl; 5 diabetes, cancer, viral infections, liver fibrosis, parasitic R" is selected from the group consisting of H. lower alkyl, infections, hypertension, and hyperlipidemia. and R"; The preferred compounds of formula VIII utilize a Z'group each Y is independently selected from the group consisting that is capable of undergoing an oxidative reaction that yields of —O , NR' with the proviso that at least one Y is an unstable intermediate which via elimination reactions 10 breaks down to the corresponding M-P(O)(NHR), or M-P NR (O)(O)(NHR). An especially preferred Z' group is OH. and pharmaceutically acceptable prodrugs and salts Groups D and D are preferably hydrogen, alkyl, OR, thereof. –OCOR, but at least one of Dor D must be H. Preferably, the tumor cell is a hepatocellular carcinoma. The following compounds and their analogs can be used in These cyclophosphamide analogs can also be used to treat 15 the prodrug methodology of the present invention. tumor cells by enhancing the activity of a P450 enzyme that In one preferred aspect, M is attached to the phosphorus in oxidizes cyclophosphamide analogs and by administering the formula I via an oxygen atom. Preferably, M is a nucleoside. cyclophosphamide analogs. Preferably, M is attached via an oxygen that is in a primary In another aspect, the P450 activity is enhanced by the hydroxyl group on a ribofuranosyl or an arabinofuranosyl administration of a compound that increases the amount of group. Preferably Such compounds include LdT, LdC. araA; endogenous P450 enzyme. Preferably, the compound that AZT, d4T, ddI; ddAddC; L-ddC; L FddC: L-d4C: L-Fd4C: increases the amount of endogenous P450 enzyme is selected 3TC: ribavirin; 5-fluoro 2" deoxyuridine: FIAU; FIAC: from the group consisting of phenobarbitol, dexamethasone, BHCG; L FMAU; BvaraU; E-5-(2-bromovinyl-2' deoxyuri rifampicin, phentoin, and preganolon-16C-carbonitrile. Pre dine: TFT: 5-propynyl-1 arabinosyluracil; CDG, DAPD; ferred cyclophosphoramide analogs include those where R" 25 FDOC: d4C; DXG: FEAU; FLG: FLT; FTC; 5-yl-carbocyclic is 2-chloroethyl, and R" is selected from the group consisting 2' deoxyguanosine; oxetanocin A; oxetanocin G. Cyclobut A, of—H, and 2-chloroethyl. Cyclobut G; dFdC: araC; bromodeoxyuridine; IDU; CdA: Also, preferred are cyclophosphamide analogs where R' FaraA; Coformycin, 2'-deoxycoformycin; araT; tiazofurin; is selected from the group consisting of —H. methyl, and ddAPR; 9-(arabinofuranosyl)-2.6 diaminopurine; 9-(2- 2-chloroethyl. 30 deoxyribofuranosyl)-2.6 diaminopurine; 9-(2'-deoxy 2 fluo In one aspect, the activity of a P450 enzyme is enhanced by roribofuranosyl)-2,6-diaminopurine; 9 (arabinofuranosyl) administration of a compound that increases the amount of guanine; 9-(2' deoxyribofuranosyl)guanine; 9-(2'-deoxy endogenous P450 enzyme. 2'fluororibofuranosyl)guanine: FMdC; 5.6 dihydro-5-azacy Also preferred cyclophosphamide analogs are those where tidine; 5-azacytidine; 5-aza 2'deoxycytidine; or AICAR. In Z, W. W., and Rare-H, and R" and R" are 2-chloroethyl. 35 another aspect, it is preferred when M is attached via an Preferred cyclophosphamide analogs have V is selected oxygen in a hydroxyl on an acyclic Sugar it is preferred when from the group consisting of aryl, Substituted aryl, heteroaryl, such MH is ACV, GCV: penciclovir; (R)-9-(3.4 dihydroxy and substituted heteroaryl. More preferred are those com butyl)guanine; or cytallene. pounds where Z, W. W., and Rare -H, and R" and R" are In one preferred aspect, the compounds informula I do not 2-chloroethyl. Also more preferred are those wherein V is 40 include compounds where MH is ara A or 5-fluoro-2'-deox selected from the group consisting of phenyl, 3-chlorophenyl, yuridine. and 3-bromophenyl. Also preferred are those wherein V is In general, it is preferred that when M is attached via an 4-pyridyl. oxygen, said oxygen is in a primary hydroxy group. In Such an Also more preferred, are those compounds where V is an instance, it is preferred that MH is araA; AZT, d4T, ddI; ddA: optionally Substituted monocyclic heteroaryl containing at 45 ddC: L-ddC; L FddC: L-d4C: L-Fd4C: 3TC: ribavirin; least one nitrogen atom. Preferably such compounds have M 5-fluoro 2" deoxyuridine: FIAU; FIAC; BHCG: L FMAU; attached via oxygen. Most preferred are Such compounds BvaralJ; E-5-(2-bromovinyl-2' deoxyuridine: TFT: 5-propy where said oxygen is in a primary hydroxyl group. Especially nyl-1 arabinosyluracil; CDG; DAPD; FDOC: d4C; DXG: preferred are such compounds where V is 4-pyridyl. FEAU; FLG: FLT. FTC: 5-yl-carbocyclic 2' deoxyguanosine: Preferably, oral bioavailability is at least 5%. More prefer 50 oxetanocin A; oxetanocin G: Cyclobut A, Cyclobut G; dFdC: ably, oral bioavailability is at least 10%. araC; bromodeoxyuridine; IDU; CdA; FaraA; Coformycin, P450 oxidation can be sensitive to stereochemistry which 2'-deoxycoformycin; araT; tiazofurin; ddAPR; 9-(arabino might either be at phosphorus or at the carbon bearing the furanosyl)-2.6 diaminopurine; 9-(2'-deoxyribofuranosyl)- aromatic group. The prodrugs of the present invention have 2.6 diaminopurine; 9-(2'-deoxy 2 fluororibofuranosyl)-2,6- two isomeric forms around the phosphorus. Preferred is the 55 diaminopurine; 9 (arabinofuranosyl)guanine; 9-(2 stereochemistry that enables both oxidation and the elimina deoxyribofuranosyl)guanine; 9-(2'-deoxy 2 fluororibofura tion reaction. Preferred is the cis-stereochemistry. In contrast, nosyl)guanine: FMdC; 5.6 dihydro-5-azacytidine. 5-azacyti the reaction is relatively insensitive to the group M since dine. 5-aza 2'deoxycytidine: AICAR: ACV, GCV: penciclo cleavage occurred with a variety of phosphonate, phosphate vir; (R)-9-(3.4 dihydroxybutyl)guanine; or cytallene. and phosphoramidates. The atom in Mattached to phospho 60 In another aspect, MH is araA. rus may be O, S or N. The active drug is M-PO, MPO, In another aspect, MH is 5-fluoro-2'-deoxyuridine. or MP.O., useful for treatment of diseases in which the liver In another aspect, compounds of formula I wherein M is is a target organ, including diabetes, hepatitis, liver cancer, attached to the phosphorus informula I via a carbonatom are liver fibrosis, malaria and metabolic diseases where the liver preferred. In such compounds, preferably M-PO, is is responsible for the overproduction of a biochemical end 65 phosphonoformic acid, or phosphonoacetic acid. products such as glucose (diabetes), cholesterol, fatty acids In another preferred aspect, MPO, MPO. , or and triglycerides (atherosclerosis). Moreover, M-PO, MP.O." is useful for the treatment of diseases of the liver or US 8,080,536 B2 43 44 metabolic diseases where the liver is responsible for the over phenyl substituted with 1-3 halogens and 4-pyridyland MH is production of a biochemical end product. Preferably, such selected from the group consisting of araA; AZT, d4T, 3TC: disease of the liver is selected from the group consisting of ribavirin; 5 fluoro-2'deoxyuridine: FMAU; DAPD; FTC: diabetes, hepatitis, cancer, fibrosis, malaria, gallstones, and 5-yl-carbocyclic 2'deoxyguanosine; Cyclobut G; dFdC: chronic cholecystalithiasis. It is more preferred when treating 5 araC; IDU; FaraA: ACV: GCV: or penciclovir. such diseases that MH, MPO. , MP2O, or MPO is an Also preferred is when MH is selected from the group antiviral or anticancer agent. consisting of ACV, GCV: penciclovir; (R)-9-(3.4 dihydroxy Preferably, the metabolic disease that MPO, MP.O., butyl)guanine; cytallene. or MP.O." are useful for diabetes, atherosclerosis, and obe When W and W are H, V is aryl, substituted aryl, het sity. 10 eroaryl, or substituted heteroaryl, and Z is H, OR, or NH In another aspect, it is preferred when the biochemical end COR, it is also preferred when M is attached to the phospho product is selected from the group consisting of glucose, rus via a carbonatom. Preferred are such compounds wherein cholesterol, fatty acids, and triglycerides. More preferred is MPO, is selected from the group consisting of phosphono when MP(O)(NHR)O or MPO' is an AMP activated pro 15 formic acid, and phosphonoacetic acid. Also preferred are tein kinase activator. MH is selected from the group consisting of PMEA, In one preferred embodiment, W' and Z are —H, W and V PMEDAP, HPMPC, HPMPA, FPMPA, and PMPA. are both the same aryl, substituted aryl, heteroaryl, or substi Preferably, oral bioavailability is at least 5%. More prefer tuted heteroaryl Such that the phosphonate prodrug moiety: ably, oral bioavailability is at least 10%. P450 oxidation can be sensitive to stereochemistry which R6 W might either be at phosphorus or at the carbon bearing the O Y aromatic group. The prodrugs of the present invention have V / two isomeric forms around the phosphorus. Preferred is the -P V stereochemistry that enables both oxidation and the elimina Y 25 tion reaction. Preferred is the cis stereochemistry. In contrast, the reaction is relatively insensitive to the group M since R6 W cleavage occurred with a variety of phosphonate, phosphate and phosphoramidates. Accordingly, the group M represents has a plane of symmetry. a group that as part of a compound of formula I enables In another preferred embodiment, W and W are H, V is 30 generation of a biologically active compound in vivo via selected from the group consisting of aryl, Substituted aryl, conversion to the corresponding M-PO. The atom in M heteroaryl, substituted heteroaryl, and Z is selected from the attached to phosphorus may be O, S, C or N. The active drug group consisting of H, OR, and NHCOR. More pre may be M-PO, or a metabolite of M-PO, such as the ferred are such compounds where Z is —H. Preferably, such triphosphate useful for treatment of diseases in which the compound have Mattached via oxygen. Most preferred are 35 liver is a target organ, including diabetes, hepatitis, liver can Such compounds where oxygen is in a primary hydroxyl cer, liver fibrosis, malaria and metabolic diseases where the group. liver is responsible for the overproduction of a biochemical Also more preferred, are those compounds where V is end products such as glucose (diabetes), cholesterol, fatty phenyl or substituted phenyl. Most preferred are such com acids and triglycerides (atherosclerosis). Moreover, M-PO, pounds where said oxygen is in a primary hydroxyl group. 40 and MP(O)(NHR)O may be useful in treating diseases Preferably, such compounds have Mattached via oxygen. where the target is outside the liver but accessible to a phosph Also more preferred, are those compounds where V is an (on)ate. Preferred M groups are groups in which M is a optionally Substituted monocyclic heteroaryl containing at nucleoside and the phosphate is attached to a hydroxyl, pref least one nitrogen atom. Preferably such compounds have M erably a primary hydroxyl on a Sugar or Sugar-analog. More attached via oxygen. Most preferred are Such compounds 45 preferred M groups include LdC. LdT, araA; AZT, d4T, ddI; where said oxygen is in a primary hydroxyl group. ddA; ddC: L-ddC; L FddC: L-d4C: L-Fd4C: 3TC: ribavirin; Especially preferred are such compounds where V is 5-fluoro 2'deoxyuridine: FIAU; FIAC; BHCG: L FMAU; selected from the group consisting of phenyl Substituted with BvaralJ; E-5-(2-bromovinyl-2' deoxyuridine: TFT: 5-propy 1-3 halogens, and 4-pyridyl. In these compounds it is also nyl-1 arabinosyluracil; CDG; DAPD; FDOC: d4C; DXG: preferred when MH is selected from the group consisting of 50 FEAU; FLG: FLT. FTC: 5-yl-carbocyclic 2'deoxyguanosine: LdT, LdC, araA; AZT, d4T, ddI; ddAddC: L-ddC; L FddC: oxetanocin A; oxetanocin G: Cyclobut A, Cyclobut G; dFdC: L-d4C; L-Fd4C: 3TC: ribavirin; 5-fluoro 2" deoxyuridine: araC; bromodeoxyuridine; IDU; CdA; FaraA; Coformycin, FIAU; FIAC; BHCG: L FMAU; Bvara U; E-5-(2-bromovi 2'-deoxycoformycin; araT; tiazofurin; ddAPR; 9-(arabino nyl-2'deoxyuridine: TFT: 5-propynyl-1 arabinosyluracil; furanosyl)-2.6 diaminopurine; 9-(2'-deoxyribofuranosyl)- CDG; DAPD, FDOC: d4C; DXG: FEAU; FLG: FLT; FTC; 55 2.6 diaminopurine; 9-(2'-deoxy 2 fluororibofuranosyl)-2,6- 5-yl-carbocyclic 2'deoxyguanosine; oxetanocin A; oxetano diaminopurine; 9 (arabinofuranosyl)guanine; 9-(2 cin G: Cyclobut A, Cyclobut G; dFdC: araC; bromodeoxyuri deoxyribofuranosyl)guanine; 9-(2'-deoxy 2"fluororibofura dine; IDU; CdA; FaraA; Coformycin, 2'-deoxycoformycin; nosyl)guanine: FMdC; 5.6 dihydro-5-azacytidine. 5-azacyti araT; tiazofurin; ddAPR; 9-(arabinofaranosyl)-2.6 diami dine. 5-aza 2'deoxycytidine: AICAR: ACV, GCV: penciclo nopurine; 9-(2'-deoxyribofuranosyl)-2.6 diaminopurine; 60 vir; (R)-9-(3.4 dihydroxybutyl)guanine; or cytallene. 9-(2'-deoxy 2 fluororibofuranosyl)-2,6-diaminopurine: 9 Especially preferred are lobucavir, FTC,3TC, BMS200,475; (arabinofuranosyl)guanine; 9-(2' deoxyribofuranosyl)gua DAPD, DXG, L-FMAU, LdC, LdT, ribavirin, F-ara-A, araC, nine; 9-(2'-deoxy 2 fluororibofaranosyl)guanine: FMdC; 5.6 CdA, dFdC, 5-fluoro-2'-deoxyuridine, ACV, GCV, and pen dihydro-5-azacytidine; 5-azacytidine. 5-aza 2 deoxycyti ciclovir. dine: AICAR: ACV, GCV: penciclovir; (R)-9-(3,4-dihydroxy 65 The preferred M groups include phosphonic acids useful butyl)guanine; or cytallene. Particularly preferred are such for treating viral infections. Such preferred antivirals include compounds where V is selected from the group consisting of PMEA: PMEDAP; HPMPC; HPMPA; FPMPA; PMPA; fos US 8,080,536 B2 45 46 carnet; phosphonoformic acid. More preferred are PMEA, PMPA, HPMPC, and HPMPA. Especially preferred are WIa PMEA and PMPA. Other preferred M groups include phosphonic acids useful in treating diabetes, liver fibrosis, e.g. collagenase inhibitors 5 such as reported in Bird et al., J. Med. Chem. 37, 158-169 (1994), parasitic infections, diseases responsive to metallo protease inhibition (e.g. hypertension, liver, cancer), and hyperlipidemia. Variable M': Group M1: In another aspect, preferred MH groups including 10 1)3TC where - P(O)(Y CH(V)CH2CH2-Y) is attached L-nucleosides. Preferred L-nucleosides include FTC, 3TC, to the primary hydroxyl. L-FMAU, LdC, and LdT. 2) (-)FTC where -P(O)(Y CH(V)CH2CH2-Y) is In one aspect, preferred MH groups are acyclic nucleo attached to the primary hydroxyl. sides. Preferred acyclic nucleosides include ACV, GCV: pen 3) L-FMAU where - P(O)(Y CH(V)CH2CH2-Y) is ciclovir; (R)-9-(3.4 dihydroxybutyl)guanine; and cytallene. 15 attached to the primary hydroxyl. More preferred are ACV: GCV, and penciclovir. 4) Penciclovir where -P(O)(Y CH(V)CH2CH2-Y) is In another aspect, preferred MH groups include dideoxy attached to the primary hydroxyl that is phosphorylated nucleosides. Preferred dideoxy nucleosides include AZT: in cells. d4T, ddI; ddA; ddC: L-ddC: L-FddC; L d4C; L-Fd4C; d4C: 5) BMS 200.475 where P(O)(Y CH(V)CH2CH2-Y) and ddAPR. More preferred are AZT, d4T, ddI; and ddC. is attached to the primary hydroxyl. In another aspect, preferred MH groups include arabino 6) L(-)Fd4C where - P(O)(Y CH(V)CH2CH2-Y) is furanosyl nucleosides. Preferred are araA: araT; 5-propynyl attached to the primary hydroxyl. 1-arabinosyluracil; araC; FaraA; 9-(arabinofuranosyl)-2,6 7) Lobucavir where - P(O)(Y CH(V)CH2CH2-Y) is diaminopurine; and 9-(arabinofuranosyl)guanine. More pre 25 attached to the primary hydroxyl that is phosphorylated ferred are araA, araC; and FaraA. in cells. In another aspect, preferred MH groups include carbocy 8) DXG where - P(O)(Y CH(V)CH2CH2-Y) is clic nucleosides. Preferred are 5-yl-carbocyclic 2' deoxygua attached to the primary hydroxyl. nosine; CDG cyclobut A: cyclobut G; and BHCG. More 9) LdC where -P(O)(Y CH(V)CH2CH2-Y) is attached preferred are 5-yl-carbocyclic 2' deoxyguanosine; and 30 to the primary hydroxyl. cyclobut G. Variable M': Group M2: 1) ddI where - P(O)(Y CH(V)CH2CH2-Y) is attached In another aspect, preferred MH groups include fluorinated to the primary hydroxyl. Sugars on the nucleosides. Preferred fluorinated Sugars 2) LdTwhere -P(O)(Y CH(V)CH2CH2-Y) is attached include FLT. FLG: FIAC; FIAU; FMAU; FEAU; dFdC:9-(2'- 35 to the primary hydroxyl. deoxy-2'fluororibofuranosyl) 2,6-diaminopurine; and 9-(2- 3) ddC where-P(O)(Y CH(V)CH2CH2-Y) is attached deoxy 2"fluororibofuranosyl)guanine More preferred are to the primary hydroxyl. L-FMAU; and dFdC. 4) AZT where - P(O)(Y CH(V)CH2CH2-Y) is In another aspect, preferred MH groups include dioxolane attached to the primary hydroxyl. nucleosides. Preferred dioxolane nucleosides include DAPD; 40 5) d4T where -P(O)(Y CH(V)CH2CH2-Y) is attached DXG; and FDOC. More preferred is DAPD and DXG. to the primary hydroxyl. In another aspect, preferred MH and MPO for treating 6) DAPD where - P(O)(Y CH(V)CH2CH2-Y) is viral infections include lobucavir, FTC, 3TC, BMS 200,475; attached to the primary hydroxyl. DAPD, DXG, L-FMAU, LdC, LdT, ribavirin, ACV, GCV, 7) L-FddC where - P(O)(Y CH(V)CH2CH2-Y) is penciclovir, PMEA, and PMPA. 45 attached to the primary hydroxyl. In another aspect, preferred MH groups for treating cancer 8) Ribavirin where - P(O)(Y CH(V)CH2CH2-Y) is include F-ara-A, araC, CdA, dFdc, and 5-fluoro-2'-deoxyuri attached to the primary hydroxyl. dine. 9) CdA where -P(O)(Y CH(V)CH2CH2-Y) is The following prodrugs are preferred compounds of the attached to the primary hydroxyl invention. The compounds are shown without depiction of 50 Variable M': Group M'3: 1) Ganciclovir where -P(O)(Y CH(V)CH2CH2-Y) is Stereochemistry since the compounds are biologically active attached to the primary hydroxyl that is phosphorylated as the diastereomeric mixture or as a single stereoisomer. in cells. Compounds named in Table 1 are designated by numbers 2) Acyclovir where -P(O)(Y CH(V)CH2CH2-Y) is assigned to the variables of formula VIa using the following 55 attached to the primary hydroxyl. convention: 3) Cytarabine where -P(O)(Y CH(V)CH2CH2-Y) is M.V.L1.L2 where Y is NH and Y is oxygen; M' is a variable attached to the primary hydroxyl. that represents compounds of the formula M-H which have a 4) Gemcitabine where -P(O)(Y CH(V)CH2CH2-Y) is specific hydroxyl group that is phosphorylated with a group attached to the primary hydroxyl P(O)(Y CH(V)CH2CH2-Y) to make compounds of for 60 5) Fludarabine where -P(O)(Y CH(V)CH2CH2-Y) is mula VIa or M' is a variable that represents phosphonic acids attached to the primary hydroxyl of the formula M-PO, which are transformed to compounds 6) Floxuridine where -P(O)(Y CH(V)CH2CH2-Y) is of formula VIa by replacing two oxygens in the PO, group attached to primary hydroxyl withY CH(V)CH2CH2-Y". V is an aryl or heteroaryl group 7) HPMPC where P(O)(Y CH(V)CH2CH2-Y) that has 2 substituents, L1 and L2, at the designated positions. 65 replaces the PO. group. Variable M' is divided into three groups with the structures 8) PMEA where - P(O)(Y CH(V)CH2CH2-Y) assigned to each group listed below: replaces the PO. group. US 8,080,536 B2 47 48 9) PMPA where - P(O)(Y CH(V)CH2CH2-Y) replaces 3) bromo the PO, group. 4) fluoro Variable V: Group V1 5) methyl 1) 2-(L1)-3 (L2) phenyl 6) isopropyl 2) 2-(L1)-4(L2) phenyl 5 7) methoxy 3) 2-(L1)-5(L2) phenyl 8) dimethylamino 4) 2-(L1)-6(L2) phenyl 9) acyloxy 5) 3-(L1)-4-(L2) phenyl 6) 3-(L1)-5(L2) phenyl Preferred compounds are compounds listed in Table 1 7) 3-(L1)-6(L2) phenyl 10 using groups M1 and V1. For example, compound 1.3.6.7 8) 2-(L1)-6(L2)-4-chlorophenyl represents structure 1 of group M1, i.e. 3TC: structure 3 of 9) 3-(L1)-5 (L2) 4-chlorophenyl group V1, i.e. 2-(L1)-5-(L2) phenyl; structure 6 of variable Variable V: Group V2 L1, i.e. isopropyl; and structure 7 of variable L2, i.e. methoxy. 15 The compound 1.3.6.7. therefore is 3TC with the P(O)(Y- CH(V)CH2CH2Y) attached to the primary hydroxyl group being {1-(2-I-propyl-5-methoxyphenyl)-1,3-propylphos phoryl. Preferred compounds are also compounds listed in Table 1 using groups M1 and V2. Preferred compounds are also compounds listed in Table 1 using groups M1 and V3. Variable V: Group V3 Preferred compounds are also compounds listed in Table 1 1) 4-(L1)-6(L2) 3-pyridyl using groups M2 and V1. 2) 5-(L1)-6(L2) 3-pyridyl 2s Preferred compounds are also compounds listed in Table 1 3) 3-(L1)-4(L2) 2-pyridyl using groups M2 and V2. 4) 3-(L1)-5(L2) 2-pyridyl Preferred compounds are also compounds listed in Table 1 5) 3-(L1)-6(L2) 2-pyridyl using groups M2 and V3. 6) 4-(L1)-5(L2) 2-pyridyl Preferred compounds are also compounds listed in Table 1 7) 4-(L1)-6(L2) 2-pyridyl 30 using groups M3 and V1. 8) 3-(L1)-4 (L2)-2-thienyl Preferred compounds are also compounds listed in Table 1 9) 2-(L1)-5(L2) 3-furanyl using groups M3 and V2. Variable L1 Preferred compounds are also compounds listed in Table 1 1) hydrogen using groups M3 and V3. 2) chloro 35 Preferred compounds are represented by all of the above 3) bromo named compounds with the exception that Y is oxygen and Y 4) fluoro 1S NH. 5) methyl Preferred compounds are represented by all of the above 6) isopropyl named compounds with the exception that Y and Y are NH. 7) methoxy 40 Preferred compounds are represented by all of the above 8) dimethylamino named compounds with the exception that Y is NCH3 and Y 9) acyloxy is oxygen. Variable L2 Preferred compounds are represented by all of the above 1) hydrogen named compounds with the exception that Y is oxygen and Y 2) chloro is NCH. TABLE 1 .1.1.1. 1.1.1.2. 1.1.1.3. 1.1.1.4 1.1.1.5 1.1.1.6 1.1.1.7 1.1.1.8. 1.1.1.9 1.1.2.1 .1.2.2 1.1.2.3 1.1.2.4 1.1.2.5 1.1.2.6 1.1.2.7 1.1.2.8 1.1.2.9 1.1.3.1 1.1.3.2 .1.3.3 1.1.3.4 1.1.3.5 1.1.3.6 1.1.3.7 1.1.3.8 1.1.3.9 1.1.4.1 1.1.4.2 1.1.4.3 1.4.4 1.1.4.5 1.1.4.6 1.1.4.7 1.14.8 1.1.4.9 1.1.5.1 1.1.5.2 1.1.5.3 1.1.5.4 15.5 11.5.6 11.57 115.8 11.5.9 11.6.1 11.6.2 11.6.3 1.1.6.4 11.6.5 16.6 116.7 11.6.8 11.6.9 11.7.1. 11.7.2 11.73 11.74 11.7.5 11.7.6 17.7 11.7.8 11.7.9 11.8.1 118.2 1.18.3 1.18.4 11.85 11.8.6 1.18.7 18.8 11.8.9 11.9.1 11.9.2 11.9.3 11.9.4 11.9.5 11.9.6 11.9.7 11.98 .1.9.9 1.2.1.1 1.2.1.2 1.2.1.3 1.2.1.4 1.2.1.5 1.2.1.6 1.2.1.7 1.2.1.8 1.2.1.9 .2.2.1 1.2.2.2 1.2.2.3 1.2.2.4 1.2.2.5 1.2.2.6 1.2.2.7 1.2.2.8 1.2.2.9 1.2.3.1 2.32 1.2.33 12.34 12.3.5 12.3.6 12.3.7 1.238 12.39 12.41 1.24.2 .2.4.3 1.2.4.4 1.2.4.5 1.2.4.6 1.2.47 1.2.4.8 1.2.4.9 1.2.5.1 1.2.5.2 1.2.5.3 2.5.4 12.55 12.5.6 12.57 1.25.8 12.59 12.6.1 12.6.2 1.2.6.3 1.26.4 26.5 126.6 12.6.7. 12.6.8 12.6.9 1.2.7.1. 1.2.7.2 1.2.7.3 1.2.7.4 12.75 2.7.6 1.2.7.7 1.2.7.8 1.2.7.9 12.8.1. 12.8.2 1.28.3 1.2.84 12.85 12.86 2.8.7 12.8.8 12.89 12.9.1 1.29.2 1.2.9.3 1.29.4 12.95 129.6 1.29.7 2.98 12.9.9 1.3.1.1 1.3.1.2 1.3.1.3 13.14 13.15 1.3.1.6 1.3.17 1.3.1.8 3.1.9 1.3.2.1 1.3.2.2 1.3.2.3 1.3.2.4 13.25 13.26 13.27 1328 13.29 3.3.1 1.3.3.2. 13.33 13.34 13.35 13.3.6 1.3.3.7 1.338 13.39 13.4.1 3.42 1.3.4.3 1.3.4.4 13.45 13.46 13.47 13:48 13.49 13.5.1. 13.5.2 3.5.3 13.5.4 13.55 13.5.6 13.5.7. 13.5.8 13.59 1.3.6.1 1.3.6.2 1.3.6.3 36.4 1.3.6.5 1.3.6.6 1.3.6.7 1.3.6.8 1.3.6.9 1.3.7.1. 13.7.2 1.3.7.3 1374 3.75 13.7.6 1.3.7.7 13.7.8 13.7.9 13.8.1. 13.8.2, 13.83 13.8.4 13.85 US 8,080,536 B2 49 50 TABLE 1-continued 38.6 38.7 3.8.8 38.9 3.91 3.9.2 3.93 3.94 3.95 3.96 39.7 39.8 3.9.9 4.1.1 .4.1.2 4.1.3 4.1.4 4.1.5 4.1.6 4.1.7 4.1.8 4.1.9 .4.2.1 4.2.2 .4.2.3 4.2.4 4.2.5 4.2.6 4.2.7 .4.2.8 4.2.9 4.3.1 4.3.2 4.3.3 .4.3.4 4.3.5 4.3.6 4.3.7 4.3.8 4.3.9 .4.4.1 .4.4.2 .4.4.3 .4.4.4 .4.4.5 .4.4.6 .4.4.7 .4.4.8 .4.4.9 .4.5.1 4.5.2 4.5.3 .4.5.4 45.5 45.6 45.7 45.8 45.9 .4.6.1 .4.6.2 .4.6.3 .4.6.4 46.5 .4.6.6 46.7 .4.6.8 .4.6.9 4.7.1 4.7.2 4.7.3 4.7.4 4.7.5 4.7.6 4.7.7 4.7.8 4.7.9 4.8.1 4.8.2 4.8.3 .4.8.4 48.5 4.8.6 4.8.7 4.8.8 48.9 4.9.1 4.9.2 49.3 .4.9.4 4.95 4.9.6 4.9.7 49.8 4.99 5.1.1 5.1.2 51.3 .5.1.4 S.1.5 5.1.6 S.1.7 5.1.8 5.19 5.2.1 5.2.2 S.2.3 .5.2.4 S.2.5 5.2.6 S.2.7 5.2.8 5.2.9 5.3.1 S.3.2 .5.3.3 53.4 S3.5 5.36 53.7 .5.3.8 S.3.9 .5.4.1 .5.4.2 5.43 .5.44 5.45 54.6 S.4.7 54.8 5.4.9 S.S.1 S.S.2 S.S.3 5.54 5.5.5 S.S.6 5.5.7 S.S.8 S.S.9 5.6.1 56.2 56.3. 56.4 S.6.5 56.6 S.6.7 56.8 56.9 S.7.1 S.7.2 .5.7.3 S.7.4 5.7.5 S.7.6 5.7.7 S.7.8 S.7.9 5.8.1 5.8.2 5.83 58.4 S8.5 58.6 S8.7 58.8 5-8.9 5.9.1 59.2 5.93 5.94 S.9.5 59.6 S.9.7 59.8 S.9.9 .6.1.1 .6.1.2 .6.1.3 .6.1.4 6.15 .6.1.6 6.17 .6.1.8 6.1.9 .6.2.1 .6.2.2 6.23 .6.2.4 6.25 .6.2.6 6.2.7 6.2.8 6.2.9 6.3.1 6.3.2 6.3.3 6.3.4 6.3.5 6.3.6 6.3.7 6.3.8 6.3.9 .6.4.1 .6.4.2 6.4.3 .6.4.4 6.45 6.4.6 6.4.7 6.4.8 6.4.9 6.5.1 6.5.2 6.53 6.54 6.S.S 65.6 6.5.7 65.8 6.5.9 .6.6.1 .6.6.2 6.6.3 .6.6.4 6.65 .6.6.6 66.7 6.6.8 6.6.9 .6.7.1 .6.7.2 .6.7.3 .6.7.4 .6.7.5 .6.7.6 .6.7.7 .6.7.8 6.79 .6.8.1 6.82 6.8.3 6.84. 6.8.5 6.86 6.8.7 6.8.8 6.8.9 6.9.1 6.9.2 6.9.3 .6.9.4 6.95 6.9.6 6.9.7 6.98 6.9.9 .7.1.1 .7.1.2 .7.1.3 .7.1.4 .7.1.5 71.6 .7.1.7 .7.1.8 .7.1.9 7.2.1 7.2.2 7.2.3 7.2.4 7.2.5 7.2.6 7.2.7 7.2.8 7.2.9 7.3.1 7.3.2 7.3.3 7.34 7.3.5 7.3.6 7.3.7 7.3.8 7.3.9 .7.4.1 7.4.2 7.43 .7.4.4 7.45 74.6 7.4.7 7.48 7.4.9 7.5.1 7.5.2 7.5.3 7.5.4 7.5.5 7.5.6 7.5.7 7.58 7.5.9 7.6.1 7.6.2 7.6.3 7.6.4 7.6.5 7.6.6 7.6.7 7.6.8 7.6.9 7.7.1 7.7.2 7.7.3 7.7.4 7.75 7.7.6 7.77 77.8 7.7.9 78.1 7.82 7.8.3 78.4 7.8.5 7.86 7.8.7 78.8 7.8.9 7.91 7.9.2 7.9.3 7.94 7.9.5 79.6 79.7 7.9.8 7.9.9 .8.1.1 .8.1.2 8.13 .8.1.4 8.15 .8.1.6 8.17 8.1.8 .8.19 .8.2.1 .8.2.2 8.23 .8.2.4 8.25 82.6 8.2.7 82.8 8.29 8.31 8.32 8.33 .8.3.4 8.3.5 8.36 8.3.7 8.3.8 83.9 .8.4.1 .8.4.2. .8.4.3. .8.44 84.5 .8.4.6 8.4.7 .84.8 8.49 85.1 8.5.2 8.5.3 8.54 8.S.S 85.6 8.5.7 85.8 8.59 .8.6.1 .8.6.2 .8.6.3 .8.6.4 86.5 .8.6.6 .8.6.7 .8.6.8 .8.6.9 8.7.1 8.7.2 8.7.3 8.74 8.7.5 87.6 8.7.7 8.78 8.7.9 88.1 8.82 8.8.3 .8.8.4 8.85 88.6 88.7 88.8 88.9 8.9.1 8.9.2 89.3 8.9.4 89.5 8.9.6 89.7 8.98 8.9.9 .9.1.1 .9.1.2 9.13 .9.14 9.15 91.6 9.17 9.18 91.9 .9.2.1 92.2 .9.23 .9.2.4 9.25 92.6 92.7 9.28 92.9 9.31 9.32 93.3 9.34 9.3.5 9.36 93.7 9.38 93.9 .9.4.1 .9.4.2 9.43 .9.4.4 9.45 .9.4.6 94.7 9.48 94.9 9.51 95.2 9.S.3 9.54 9.S.S 9.5.6 9.S.7 9.58 9.S.9 96.1 9.6.2 96.3 .9.6.4 9.6.5 96.6 96.7 96.8 96.9 9.7.1 9.7.2 9.7.3 9.7.4 9.7.5 9.7.6 9.7.7 9.7.8 9.7.9 98.1 98.2 98.3 9.84 98.5 98.6 98.7 98.8 98.9 99.1 9.9.2 99.3 9.94 99.5 99.6 99.7 99.8 99.9 2.1.1.1 2.1.1.2 2.1.1.3 2.1.1.4 2.1.1.5 2.1.1.6 2.1.1.7 2.1.1.8 2.1.1.9 2.1.2.1 2.1.2.2 2.1.2.3 2.1.2.4 2.1.2.5 2.1.2.6 2.1.2.7 2.1.2.8 2.1.2.9 2.1.3.1 2.1.3.2 2.1.3.3 2.1.3.4 2.1.3.5 2.1.3.6 2.1.3.7 2.13.8 2.1.3.9 2.1.4.1 2.1.4.2 2.1.4.3 2.1.4.4 2.1.45 2.1.4.6 2.14.7 2.1.4.8 2.1.4.9 2.1.5.1 2.1.5.2 2.1.5.3 2.1.5.4 2.1.S.S 2.1.5.6 2.1.5.7 2.1.5.8 2.1.5.9 2.1.6.1 2.1.6.2 2.1.6.3 2.1.6.4 2.1.6.5 2.1.6.6 2.1.6.7 2.1.6.8 2.1.6.9 2.17.1 2.17.2 2.17.3 2.17.4 2.17.5 2.17.6 2.17.7 2.17.8 2.17.9 2.1.8.1 2.18.2 2.18.3 2.1.8.4 2.18.5 2.18.6 2.18.7 2.18.8 2.18.9 2.19.1 2.1.9.2 21.9.3 2.194 21.9.5 2.1.96 21.9.7 2.1.98 2.1.99 2.2.1.1 2.2.1.2 2.2.1.3 2.2.1.4 2.2.1.5 2.2.1.6 2.2.1.7 2.2.1.8 2.2.1.9 2.2.2.1 2.2.2.2 2.2.23 2.2.2.4 2.2.2.5 2.2.2.6 2.2.2.7 2.2.2.8 2.2.2.9 2.2.3.1 2.23.2 22.3.3 2.2.3.4 2.2.3.5 22.3.6 22.3.7 2.23.8 2.23.9 2.2.4.1 2.2.4.2 2.2.4.3 2.2.4.4 2.2.45 2.2.4.6 2.2.4.7 2.2.4.8 2.2.4.9 2.2.5.1 2.2.5.2 2.2.5.3 2.2.5.4 2.2.S.S 2.2.5.6 2.2.5.7 2.2.5.8. 2.2.5.9 2.2.6.1 2.2.6.2 2.2.6.3 2.2.6.4 2.26.5 2.2.6.6 2.26.7 2.26.8 2.26.9 2.2.7.1 2.2.7.2 2.2.7.3 2.2.7.4 2.2.7.5 2.2.7.6 2.2.7.7 2.2.7.8 2.2.7.9 22.8.1 22.8.2 2.28.3 2.2.8.4 2.28.5 22.86 2.28.7 22.8.8 2.28.9 2.29.1 2.29.2 2.29.3 2.2.9.4 2.29.5 2.29.6 2.2.9.7 2.29.8 2.29.9 2.3.1.1 2.3.1.2 2.3.1.3 2.3.1.4 2.3.1.5 2.3.1.6 2.3.1.7 2.3.1.8 2.3.1.9 2.3.2.1 2.3.2.2 2.32.3 2.3.2.4 2.3.25 2.3.2.6 2.3.2.7 2.3.2.8 2.3.2.9 2.3.3.1 2.3.3.2 2.3.3.3 2.3.3.4 2.3.3.5 2.3.3.6 2.3.3.7 2.338 233.9 2.3.4.1 2.3.4.2 2.3.4.3 2.3.4.4 2.3.4.5 2.3.4.6 2.3.4.7 2.348 2.3.4.9 23.5.1 2.3.5.2 2.3.5.3 2.35.4 2.3.S.S 2.35.6 2.3.5.7 2.3.5.8 2.35.9 2.3.6.1 2.3.6.2 2.3.6.3 2.3.6.4 2.3.6.5 2.3.6.6 2.3.6.7 2.3.6.8, 2.3.6.9 2.3.7.1 2.3.7.2 2.3.7.3 2.3.7.4 2.3.7.5 2.3.7.6 2.3.7.7 2.3.7.8 2.3.7.9 23.81 2.38.2 2.38.3 2.38.4 2.38.5 2.38.6 2.38.7 2.38.8 2.38.9 23.91 23.9.2 23.93 2.39.4 2.3.9.5 23.96 2.39.7 23.9.8 2.399 2.4.1.1 2.4.1.2 2.4.1.3 2.4.1.4 2.4.15 2.4.1.6 24.17 2.4.1.8 2.4.1.9 2.4.2.1 2.4.2.2 2.4.2.3 2.4.2.4 24.25 2.4.2.6 24.2.7 2.4.2.8 2.4.2.9 2.4.3.1 2.4.3.2 24.3.3 2.4.3.4 2.4.3.5 2.4.3.6 2.43.7 24.38 243.9 2.4.4.1 2.4.4.2 2.4.4.3 2.4.4.4 2.4.4.5 2.4.4.6 2.4.4.7 2.4.4.8 2.4.4.9 2.45.1 2.45.2 2.45.3 2.45.4 2.45.5 2.45.6 2.45.7 2.45.8 2.45.9 2.4.6.1 2.4.6.2 2.4.6.3 2.4.6.4 2.4.6.5 2.4.6.6 2.4.6.7 2.4.6.8 2.46.9 2.47.1 2.47.2 24.7.3 2.47.4 US 8,080,536 B2 51 52 TABLE 1-continued 2.47.5 2.47.6 2.47.7 2.47.8 2.47.9 2.4.8.1 2.4.8.2 248.3 2.4.8.4 2.48.5 2.4.8.6 2.48.7 2.48.8 2.4.89 2.4.9.1 2.4.9.2 2.4.9.3 2.4.9.4 24.95 2.4.9.6 2.4.9.7 2.4.9.8 2.4.9.9 2.5.1.1 2.5.1.2 2.51.3 2.5.1.4 2.5.1.5 2.51.6 2.5.1.7 2.5.1.8 2.5.19 2.5.2.1 2.5.2.2 2.5.2.3 2.5.2.4 2.5.2.5 2.5.2.6 2.5.2.7 2.5.2.8 2.5.2.9 2.5.3.1 2.5.3.2 2.5.3.3 2.5.3.4 2.5.3.5 2.5.3.6 2.5.3.7 2.53.8 2.5.3.9 25.4.1 2.54.2 2.5.4.3 2.5.44 2.5.45 25.46 2.54.7 2.5.48 25.4.9 2.S.S.1 2.S.S.2 2.S.S.3 2.55.4 2.5.5.5 2.S.S.6 2.5.5.7 2.S.S.8 2.S.S.9 2.56.1 2.5.6.2 2.56.3 2.56.4 2.S.6.5 256.6 2.5.6.7 256.8 2.56.9 2.5.7.1 2.5.7.2 2.5.7.3 2.5.7.4 2.5.7.5 2.5.7.6 2.5.7.7 2.5.7.8 2.5.7.9 2.58.1 2.5.8.2 2.5.8.3 2.58.4 2.5.8.5 2.58.6 2.5.8.7 2.58.8 2.5.8.9 2.59.1 2.59.2 2.S.9.3 25.94 2.5.9.5 2.59.6 2.59.7 2.5-9.8 2.S.99 2.6.1.1 2.6.1.2 2.6.13 2.6.1.4 26.15 2.6.1.6 26.17 2.6.1.8 2.6.19 2.6.2.1 2.6.2.2 2.6.2.3 2.6.2.4 2.6.2.5 2.6.2.6 2.6.2.7 2.6.2.8 2.6.29 2.6.3.1 2.6.32 2.6.3.3 2.6.3.4 2.63.5 2.6.36 2.6.3.7 2.6.38 26.3.9 2.6.4.1 2.6.4.2 2.6.4.3 2.6.4.4 2.64.5 2.6.4.6 26.4.7 2.6.4.8 26.49 26.5.1 26.5.2 26.5.3 26.54 2.6.S.S 2.65.6 2.6.S.7 26.5.8 26.5.9 2.6.6.1 2.6.6.2 2.66.3 2.6.6.4 26.65 26.66 26.6.7 2.66.8 26.6.9 2.6.7.1 2.6.7.2 2.6.7.3 2.6.7.4 2.6.7.5 2.6.7.6 2.6.7.7 2.6.7.8 2.6.7.9 26.8.1 2.68.2 26.83 2.6.8.4 2.68.5 2.6.8.6 2.68.7 2.6.8.8 2.68.9 26.9.1 26.9.2 26.93 26.94 26.9.5 2.69.6 26.9.7 26.98 26.9.9 2.7.1.1 2.7.1.2 2.7.1.3 2.71.4 2.7.1.5 2.7.1.6 2.7.1.7 2.7.1.8 2.7.1.9 27.2.1 27.2.2 2.72.3 27.2.4 2.72.5 27.26 2.72.7 2.72.8 2.7.2.9 2.7.3.1 2.73.2 2.7.3.3 2.73.4 2.73.5 2.73.6 2.73.7 2.7.3.8 2.73.9 2.74.1 2.74.2 27.4.3 2.7.4.4 2.74.5 2.74.6 2.74.7 2.74.8 2.74.9 2.7.5.1 2.7.5.2 2.7.5.3 2.75.4 2.7.5.5 2.75.6 2.75.7 2.75.8 2.7.5.9 2.76.1 2.76.2 2.76.3 27.64 2.76.5 2.76.6 2.76.7 2.76.8 2.76.9 2.77.1 2.77.2 2.7.7.3 2.7.7.4 2.7.75 2.7.7.6 2.77.7 2.77.8 2.7.7.9 2.78.1 2.78.2 2.78.3 2.78.4 2.78.5 2.78.6 2.78.7 2.78.8 2.78.9 2.79.1 2.79.2 2.79.3 2.79.4 2.79.5 2.79.6 2.79.7 2.79.8 2.79.9 2.8.1.1 2.8.1.2 2.8.1.3 2.8.1.4 2.8.1.5 2.8.1.6 2.8.1.7 2.8.1.8 2.8.19 2.8.2.1 2.8.2.2 2.8.2.3 2.8.2.4 2.8.25 2.8.2.6 2.8.2.7 2.8.2.8 2.8.2.9 2.8.31 2.83.2 2.83.3 2.8.34 2.83.5 2.8.36 2.8.3.7 2.83.8 2.83.9 2.84.1 2.8.4.2 2.84.3 2.8.44 2.84.5 2.84.6 2.84.7 2.84.8 2.84.9 2.8.5.1 2.85.2 2.8.5.3 2.85.4 2.8.S.S 2.8.5.6 2.85.7 2.85.8 2.8.5.9 2.8.6.1 2.8.6.2 2.8.6.3 2.8.6.4 2.8.6.5 2.8.6.6 2.8.6.7 2.86.8 2.8.6.9 2.8.7.1 2.87.2 2.8.7.3 2.87.4 2.8.7.5 2.87.6 2.87.7 2.8.78 2.87.9 2.88.1 2.8.8.2 2.8.83 2.88.4 2.8.85 2.8.86 2.88.7 2.88.8 2.88.9 2.89.1 2.89.2 2.89.3 2.89.4 2.89.5 2.89.6 2.89.7 2.89.8 2.89.9 2.9.1.1 2.91.2 2.91.3 2.9.1.4 2.91.5 2.91.6 2.91.7 2.9.1.8 2.91.9 2.92.1 2.9.2.2 2.92.3 2.9.2.4 2.92.5 2.92.6 2.92.7 2.92.8 2.92.9 2.93.1 2.93.2 2.93.3 2.93.4 2.93.5 2.93.6 2.93.7 2.93.8 2.93.9 2.9.4.1 2.9.4.2 2.94.3 2.9.4.4 2.9.45 2.94.6 2.94.7 2.94.8 2.94.9 2.95.1 2.95.2 2.95.3 2.95.4 2.9.S.S 2.956 2.95.7 2.95.8 2.95.9 2.96.1 2.96.2 2.96.3 2.96.4 2.96.5 2.96.6 2.96.7 2.96.8 2.96.9 2.97.1 2.9.7.2 2.97.3 2.97.4 2.97.5 2.97.6 2.9.7.7 2.97.8 2.97.9 2.98.1 2.98.2 2.98.3 2.98.4 2.98.5 2.98.6 2.98.7 2.98.8 2.98.9 2.99.1 29.9.2 2.99.3 2.99.4 2.99.5 2.99.6 2.99.7 2.99.8 2.99.9 3.1.1.1 3.1.1.2 3.11.3 3.1.1.4 3.11.5 3.1.1.6 3.11.7 3.1.1.8 3.11.9 3.1.2.1 3.1.2.2 3.12.3 3.1.2.4 3.12.5 3.12.6 3.1.2.7 3.12.8 3.12.9 3.13.1 3.1.3.2 3.1.3.3 3.13.4 3.1.3.5 3.13.6 3.1.3.7 3.13.8 3.13.9 3.14.1 3.14.2 3.14.3 3.14.4 3.14.5 3.1.4.6 3.14.7 3.14.8 3.14.9 3.15.1 3.15.2 3.15.3 3.15.4 3.1.S.S 3.15.6 3.15.7 3.15.8 3.15.9 3.1.6.1 3.16.2 3.1.6.3 3.1.6.4 3.16.5 3.16.6 3.1.6.7 3.1.6.8 3.1.6.9 3.17.1 3.17.2 3.17.3 3.17.4 3.17.5 3.17.6 3.17.7 3.17.8 3.17.9 3.18.1 3.18.2 3.18.3 3.18.4 3.18.5 3.18.6 3.18.7 3.18.8 3.18.9 3.19.1 3.19.2 3.19.3 3.19.4 3.19.5 3.19.6 3.19.7 3.19.8 3.19.9 3.2.1.1 3.2.1.2 3.21.3 3.2.1.4 3.21.5 3.21.6 3.21.7 3.2.1.8 3.21.9 3.2.2.1 3.2.2.2 3.22.3 3.2.2.4 3.2.2.5 3.22.6 3.2.2.7 3.22.8 3.22.9 3.23.1 3.23.2 3.23.3 3.23.4 3.23.5 3.23.6 3.23.7 3.23.8 3.23.9 3.2.4.1 3.2.4.2 3.24.3 3.2.4.4 3.24.5 3.2.4.6 3.24.7 3.24.8 3.24.9 3.25.1 3.25.2 3.2.5.3 3.25.4 3.2.S.S 3.25.6 3.25.7 3.2.5.8 3.25.9 3.26.1 3.26.2 3.26.3 3.2.6.4 3.26.5 3.26.6 3.2.6.7 3.26.8 3.26.9 3.2.7.1 3.2.7.2 3.2.7.3 3.27.4 3.2.7.5 3.2.7.6 3.2.7.7 3.2.7.8 3.2.7.9 3.28.1 3.28.2 3.283 3.28.4 3.28.5 3.28.6 3.28.7 3.28.8 3.28.9 3.29.1 3.29.2 3.29.3 3.29.4 3.29.5 3.29.6 3.2.9.7 3.29.8 3.29.9 3.3.1.1 3.31.2 3.3.1.3 3.31.4 3.31.5 3.3.1.6 3.31.7 3.3.1.8 3.31.9 3.3.2.1 3.3.2.2 3.32.3 3.32.4 3.3.2.5 3.32.6 3.32.7 3.32.8 3.32.9 3.33.1 3.33.2 3.3.3.3 3.33.4 3.33.5 3.33.6 3.33.7 3.33.8 3.33.9 3.34.1 3.34.2 3.34.3 3.3.4.4 3.34.5 3.34.6 3.34.7 3.34.8 3.34.9 3.35.1 3.35.2 3.35.3 3.35.4 3.3.S.S 3.35.6 3.3.5.7 3.35.8 3.35.9 3.3.6.1 3.3.6.2 3.3.6.3 3.3.6.4 3.3.6.5 3.3.6.6 3.3.6.7 3.3.6.8 3.36.9 3.3.7.1 3.3.7.2 3.3.7.3 3.37.4 3.3.7.5 3.3.7.6 3.3.7.7 3.37.8 3.3.7.9 3.38.1 3.38.2 3.38.3 3.38.4 3.38.5 3.38.6 3.38.7 3.38.8 3.3-8.9 3.39.1 3.39.2 3.39.3 3.39.4 3.39.5 3.39.6 3.39.7 3.39.8 3.39.9 3.4.1.1 3.4.1.2 3.4.13 3.4.1.4 3.4.15 3.4.1.6 3.41.7 3.4.1.8 3.41.9 3.4.2.1 3.4.2.2 3.42.3 3.4.2.4 3.425 3.4.2.6 3.42.7 3.42.8 3.42.9 3.43.1 34.32 34.3.3 3.43.4 3.43.5 3.43.6 3.43.7 3.43.8 3.43.9 3.4.4.1 3.4.4.2 3.4.4.3 3.4.4.4 3.445 3.4.4.6 3.44.7 3.4.4.8 3.44.9 3.45.1 34.52 3.45.3 3.45.4 34.S.S 3.456 3.45.7 3.45.8 3.45.9 3.4.6.1 3.4.6.2 34.6.3 3.4.6.4 3.46.5 3.46.6 3.4.6.7 3.46.8 3.46.9 34.7.1 3.47.2 34.7.3 34.74 3.47.5 3.47.6 3.47.7 3.47.8 34.79 3.48.1 3.48.2 3.48.3 3.4.8.4 3.48.5 3.48.6 3.48.7 3.48.8 3.48.9 34.9.1 3.4.9.2 34.9.3 34.94 3.49.5 34.9.6 3.4.9.7 34.9.8 3.49.9 3.5.1.1 3.5.1.2 3.5.1.3 3.5.1.4 3.5.1.5 3.5.1.6 3.5.1.7 3.5.1.8 3.5.1.9 3.5.2.1 3.5.2.2 3.5.23 3.5.2.4 3.5.2.5 3.5.2.6 3.5.2.7 3.5.2.8 3.5.2.9 3.5.3.1 3.5.3.2 3.5.3.3 3.534 3.5.3.5 3.5.3.6 3.5.3.7 3.5.3.8 3.5.3.9 3.54.1 3.54.2 3.54.3 3.5.44 3.54.5 3.54.6 3.54.7 3.54.8 3.54.9 3.S.S.1 3.S.S.2 3.S.S.3 3.S.S.4 3.5.5.5 3.S.S.6 3.5.5.7 3.S.S.8 3.S.S.9 3.56.1 3.56.2 3.S.6.3 US 8,080,536 B2 53 54 TABLE 1-continued 3.56.4 3.S.6.5 3.56.6 3.S.6.7 3.56.8 3.S.6.9 3.5.7.1 3.5.7.2 3.5.7.3 3.5.7.4 3.5.7.5 3.5.7.6 3.5.7.7 3.5.7.8 3.5.7.9 3.5.8.1 3.5.8.2 3.5.8.3 3.58.4 3.5.8.5 3.5.8.6 3.5.8.7 3.5.8.8 3.5.8.9 3.S.9.1 3.S.9.2 3.59.3 3.59.4 3.59.5 3.S.9.6 3.S.9.7 3.59.8 3.S.9.9 3.6.1.1 3.6.1.2 3.6.13 3.6.1.4 3.6.15 3.6.16 3.6.17 3.6.1.8 3.6.19 3.6.2.1 3.6.2.2 36.23 3.6.2.4 3.6.25 3.6.2.6 3.6.2.7 3.6.2.8 3.6.2.9 36.31 3.6.32 3.6.3.3 3.6.3.4 3.6.3.5 36.36 3.6.3.7 3.638 3.6.3.9 3.6.4.1 3.6.4.2 36.4.3 3.6.4.4 36.45 3.6.46 3.64.7 36.4.8 36.4.9 36.5.1 36.5.2 3.6.5.3 36.5.4 3.6.S.S 36.56 3.6.5.7 3.65.8 3.6.5.9 3.66.1 3.6.6.2 3.6.6.3 3.66.4 3.6.6.5 36.66 3.6.6.7 3.66.8 3.66.9 3.6.7.1 3.6.7.2 3.6.7.3 3.6.7.4 3.6.7.5 3.6.7.6 3.6.7.7 3.6.7.8 3.6.7.9 3.68.1 36.8.2 3.68.3 36.84 36.8.5 3.68.6 3.6.8.7 3.68.8 36.8.9 36.9.1 3.69.2 36.9.3 36.94 3.6.9.5 36.96 3.6.9.7 36.9.8 36.9.9 3.7.1.1 3.7.1.2 3.7.1.3 3.7.1.4 3.7.1.5 3.7.1.6 3.7.1.7 3.7.1.8 3.7.1.9 3.7.2.1 3.7.2.2 3.7.2.3 3.72.4 3.7.2.5 3.72.6 3.72.7 3.7.2.8 3.72.9 3.7.3.1 3.7.32 3.7.3.3 37.3.4 3.7.3.5 3.7.3.6 37.3.7 3.7.3.8 3.7.3.9 3.74.1 3.74.2 3.74.3 37.44 3.74.5 3.74.6 3.74.7 3.748 37.49 3.7.5.1 3.75.2 3.75.3 3.75.4 3.7.5.5 3.75.6 3.75.7 3.75.8 3.75.9 3.7.6.1 3.7.6.2 3.76.3 3.7.6.4 3.7.6.5 3.7.6.6 3.7.6.7 3.76.8 3.7.6.9 3.7.7.1 3.7.7.2 3.7.7.3 3.77.4 3.7.7.5 3.7.76 3.7.7.7 3.77.8 3.7.7.9 3.7.8.1 37.82 3.7.8.3 37.8.4 3.78.5 3.7.8.6 3.7.8.7 3.7.8.8 3.7.8.9 3.7.9.1 3.7.9.2 3.79.3 37.94 3.7.9.5 3.7.9.6 3.7.9.7 3.79.8 3.7.9.9 3.81.1 3.8.1.2 3.8.1.3 3.814 3.8.1.5 3.8.1.6 3.81.7 3.8.1.8 3.8.19 3.8.2.1 3.8.2.2 38.23 3.8.2.4 3.82.5 3.8.2.6 3.8.2.7 3.8.2.8 3.8.2.9 38.31 38.32 38.3.3 38.34 3.8.3.5 38.36 3.8.3.7 38.3.8 3.83.9 3.84.1 3.842 3.84.3 3.84.4 3.84.5 38.4.6 3.84.7 3.84.8 38.4.9 3.85.1 3.8.5.2 3.8.5.3 38.54 3.8.S.S 3.85.6 3.85.7 3.85.8 3.8.5.9 3.8.6.1 3.8.6.2 3.8.6.3 3.86.4 3.8.6.5 3.8.6.6 3.8.6.7 3.8.6.8 3.86.9 3.8.7.1 3.8.7.2 3.8.7.3 3.87.4 3.8.75 3.8.7.6 3.87.7 3.8.7.8 3.8.7.9 3.88.1 38.82 3.88.3 3.884 3.8.8.5 3.88.6 3.88.7 3.88.8 3.88.9 38.9.1 3.89.2 3.89.3 38.9.4 3.89.5 3.89.6 3.89.7 38.98 3.89.9 3.9.1.1 3.9.1.2 3.913 3.9.14 3.9.1.5 3.91.6 3.9.17 39.18 3.9.19 3.9.2.1 3.9.2.2 3.9.23 3.92.4 3.9.25 392.6 3.92.7 3.9.28 3.92.9 39.31 3.93.2 3.93.3 39.34 3.93.5 3.936 3.93.7 3.938 3.93.9 3.94.1 3.94.2 3.94.3 3.944 3.9.45 3.94.6 3.94.7 39.4.8 3.94.9 3.9.5.1 3.9.5.2 3.9.S.3 3.95.4 3.9.S.S 3.9.S.6 3.95.7 3.9.S.8 3.9.5.9 3.96.1 39.6.2 3.96.3 3.96.4 3.96.5 3.96.6 3.96.7 3.96.8 3.96.9 39.7.1 3.9.7.2 3.9.7.3 39.74 3.9.7.5 39.7.6 3.9.7.7 39.7.8 3.97.9 39.8.1 39.8.2 39.83 39.8.4 39.8.5 39.8.6 3.98.7 3.988 39.8.9 3.99.1 3.9.9.2 3.99.3 3.99.4 3.99.5 3.99.6 3.99.7 3.99.8 3.99.9 4.1.1.1 4.1.1.2 4.1.1.3 4.1.1.4 4.1.1.5 4.1.1.6 4.1.1.7 4.1.1.8 4.1.1.9 4.1.2.1 4.1.2.2 4.1.2.3 4.1.2.4 4.1.2.5 4.1.2.6 4.1.2.7 4.1.2.8 4.1.2.9 4.1.3.1 4.1.3.2 4.1.3.3 4.1.3.4 4.1.3.5 4.1.3.6 4.1.3.7 4.1.3.8 4.1.3.9 4.1.4.1 4.1.4.2 4.1.4.3 4.1.4.4 4.1.4.5 4.1.4.6 4.1.4.7 4.1.4.8 4.1.4.9 4.1.5.1 4.1.5.2 4.15.3 4.1.5.4 4.1.5.5 4.1.5.6 4.1.5.7 4.1.5.8 4.1.5.9 4.1.6.1 4.1.6.2 4.1.6.3 4.1.6.4 4.1.6.5 4.1.6.6 4.1.6.7 4.1.6.8 4.1.6.9 4.1.7.1 4.17.2 4.17.3 4.1.7.4 4.17.5 4.17.6 4.17.7 4.1.78 4.17.9 4.1.8.1 4.1.8.2 4.18.3 4.1.8.4 4.18.5 4.1.8.6 4.18.7 4.18.8 4.1.89 4.1.9.1 4.1.9.2 4.1.93 4.1.9.4 4.19.5 4.1.9.6 4.19.7 4.1.98 4.19.9 4.2.1.1 4.2.1.2 4.2.1.3 4.2.1.4 4.2.1.5 4.2.1.6 4.2.1.7 4.2.1.8 4.2.1.9 4.2.2.1 4.2.2.2 4.2.2.3 4.2.2.4 4.2.2.5 4.2.2.6 4.22.7 4.2.2.8 4.2.2.9 4.2.3.1 4.2.3.2 4.2.3.3 4.2.3.4 4.2.35 4.2.3.6 4.2.3.7 4.238 4.23.9 4.2.4.1 4.2.4.2 4.2.4.3 4.2.4.4 4.2.4.5 4.2.4.6 4.2.4.7 4.2.4.8 4.2.4.9 4.2.5.1 4.2.5.2 4.2.5.3 4.2.5.4 4.25.5 4.2.5.6 4.2.5.7 4.25.8 4.2.5.9 4.2.6.1 4.2.6.2 4.2.6.3 4.2.6.4 4.2.6.5 4.2.6.6 4.2.67 4.2.6.8 4.26.9 4.2.7.1 4.2.7.2 4.2.7.3 4.2.7.4 4.2.7.5 4.2.7.6 4.2.7.7 4.2.7.8 4.2.7.9 4.2.8.1 4.2.8.2 4.283 4.2.8.4 4.28.5 4.2.8.6 4.28.7 4.28.8 4.2.89 4.2.9.1 4.2.9.2 4.29.3 4.2.9.4 42.95 4.29.6 4.29.7 42.9.8 4.299 4.3.1.1 4.3.1.2 4.3.1.3 4.3.1.4 4.3.1.5 4.3.1.6 4.3.17 4.3.1.8 4.3.1.9 4.3.2.1 4.3.2.2 4.3.2.3 4.3.2.4 4.3.2.5 4.3.2.6 4.3.2.7 4.3.2.8 4.3.2.9 4.3.3.1 4.3.3.2 4.3.3.3 4.3.3.4 4.3.3.5 4.3.3.6 4.33.7 4.3.3.8 4.33.9 4.3.4.1 4.3.4.2 4.3.4.3 4.3.4.4 4.3.4.5 4.3.4.6 4.3.4.7 4.3.4.8 4.3.4.9 4.35.1 435.2 4.3.5.3 4.3.54 4.3.S.S 4.3.56 4.3.5.7 4.35.8 4.35.9 4.3.6.1 4.3.6.2 4.3.6.3 4.3.6.4 4.3.6.5 4.3.6.6 4.3.6.7 4.3.6.8 4.3.6.9 4.3.7.1 4.3.7.2 4.3.7.3 4.3.7.4 4.3.7.5 4.3.7.6 4.3.7.7 4.3.7.8 4.3.7.9 4.38.1 4.38.2 4.383 4.3.8.4 4.38.5 4.38.6 4.38.7 4.38.8 4.38.9 4.39.1 4.3.9.2 4.3.93 4.3.94 4.3.9.5 4.3.96 4.3.9.7 4.3.98 4.3.99 4.4.1.1 4.4.1.2 4.4.1.3 4.4.1.4 4.4.1.5 4.4.1.6 4.4.1.7 4.4.1.8 4.4.1.9 4.4.2.1 4.4.2.2 4.4.2.3 4.4.2.4 4.4.2.5 4.4.2.6 4.4.2.7 4.4.2.8 4.4.2.9 4.4.3.1 4.4.3.2 4.4.3.3 4.4.3.4 4.43.5 4.4.3.6 4.43.7 4.4.3.8 4.43.9 4.4.4.1 4.4.4.2 4.4.4.3 4.4.4.4 4.4.4.5 4.4.4.6 4.4.4.7 4.4.4.8 4.4.4.9 4.4.5.1 4.4.5.2 4.45.3 4.4.5.4 4.45.5 4.4.5.6 4.45.7 4.458 4.459 4.4.6.1 4.4.6.2 4.4.6.3 4.4.6.4 4.4.6.5 4.4.6.6 4.4.6.7 4.4.6.8 4.4.6.9 4.4.7.1 4.4.7.2 4473 4.4.7.4 44.75 4.47.6 447.7 4.478 447.9 4.4.8.1 4.4.8.2 4.4.8.3 4.4.8.4 4.48.5 4.4.8.6 4.48.7 4.4.8.8 4.4.89 4.4.9.1 4.4.9.2 4.4.9.3 4.4.9.4 4.4.9.5 4.4.9.6 4.4.9.7 4.4.9.8 4.4.9.9 4.5.1.1 4.5.1.2 4.51.3 4.5.1.4 4.51.5 4.51.6 4.51.7 4.5.1.8 4.5.19 4.5.2.1 4.5.2.2 4.5.23 4.5.2.4 45.25 4.5.2.6 4.5.2.7 4.5.2.8 4.5.2.9 4.5.3.1 4.5.3.2 4.5.3.3 4.5.3.4 4.5.3.5 4.5.3.6 4.5.3.7 4.5.3.8 4.5.3.9 4.5.4.1 4.5.4.2 45.4.3 4.5.44 45.45 4.5.4.6 4.54.7 4.5.48 4.54.9 45.5.1 4.55.2 4.S.S.3 45.54 4.S.S.S 45.5.6 4.S.S.7 4.S.S.8 4.S.S.9 4.56.1 45.6.2 4.56.3 4.5.6.4 45.6.5 4.56.6 45.6.7 4.56.8 45.6.9 4.5.7.1 4.5.7.2 4.5.7.3 45.74 45.75 4.5.7.6 4.5.7.7 4.5.7.8 4.5.7.9 4.5.8.1 4.5.8.2 4.583 4.5.8.4 4.5.8.5 4.5.8.6 4.5.8.7 4.5.8.8 4.5.8.9 4.59.1 45.92 4.59.3 45.94 45.9.5 4.59.6 45.9.7 4.59.8 4.59.9 4.6.1.1 4.6.1.2 4.6.1.3 4.6.1.4 4.6.15 4.6.1.6 4.6.17 4.6.1.8 4.6.1.9 4.6.2.1 4.6.2.2 4.6.2.3 4.6.2.4 4.6.2.5 4.6.2.6 4.6.2.7 4.6.2.8 4.6.29 4.6.3.1 4.6.3.2 46.3.3 4.6.3.4 46.35 46.36 46.3.7 46.38 46.3.9 4.6.4.1 4.6.4.2 4.6.4.3 4.6.4.4 4.6.4.5 4.6.4.6 46.4.7 4.6.4.8 4.6.4.9 46.51 46.5.2 US 8,080,536 B2 55 56 TABLE 1-continued 46.53 4.6.5.4 46.5.5 465.6 46.57 46.5.8 46.5.9 4.6.6.1 4.6.6.2 46.6.3 4.6.6.4 46.65 4.6.6.6 4.66.7 46.6.8 46.6.9 4.6.7.1 4.6.7.2 4.6.7.3 4.6.7.4 4.6.7.5 4.6.7.6 4.6.7.7 4.6.7.8 46.79 4.6.8.1 4.6.8.2 46.83 4.6.8.4 46.85 46.86 46.87 46.8.8 4.68.9 4.6.9.1 46.9.2 46.93 4.6.9.4 46.95 46.96 46.9.7 46.98 46.9.9 4.7.1.1 4.7.1.2 4.7.1.3 4.7.1.4 4.7.15 4.7.1.6 4.7.1.7 4.7.1.8 4.7.1.9 4.7.2.1 4.7.2.2 4.7.23 4.7.2.4 4.7.2S 4.7.26 4.7.2.7 4.7.2-8 4.7.2.9 4.7.3.1 4.7.32 4.7.3.3 4.7.34 4.7.3.5 4.7.36 4.7.3.7 4.7.38 4.7.3.9 4.7.4.1 4.7.4.2 47.43 4.7.4.4 4.7.45 4.7.46 4.7.4.7 4.7.48 4.7.49 4.7.51 4.7.5.2 4.7.5.3 4.75.4 4.7.S.S 4.7.5.6 4.7.5.7 4.7.58 4.7.5.9 4.7.6.1 4.7.62 4.7.6.3 4.7.6.4 4.7.6.5 4.7.6.6 4.7.6.7 4.76.8 4.7.6.9 4.7.7.1 4.7.7.2 4.7.7.3 4.77.4 4.7.7.5 4.7.7.6 4.7.7.7 4.7.7.8 4.7.7.9 4.7.8.1 4.7.82 4.7.83 4.7.84 4.7.8.5 4.7.86 4.7.8.7 4.7.8.8 4.7.8.9 4.7.9.1 4.7.9.2 4.7.9.3 4.7.94 4.7.9S 4.7.9.6 4.7.9.7 4.7.9.8 4.7.9.9 4.8.1.1 4.8.1.2 4.8.1.3 4.8.1.4 4.8.1.5 4.8.1.6 4.8.1.7 4.8.1.8 4.8.19 4.8.2.1 4.8.2.2 4.82.3 4.8.2.4 48.25 4.8.2.6 4.8.2.7 4.8.2.8 4.8.2.9 48.31 4.83.2 48.33 4.8.3.4 4.835 48.36 4.83.7 48.38 48.39 4.8.4.1 4.8.4.2 4.84.3 4.8.44 4.84.5 4.8.4.6 48.4.7 4.84.8 48.4.9 48.5.1 4.852 48.5.3 48.54 48.S.S 4.856 48.5.7 48.58 48.59 4.8.6.1 4.8.6.2 4.8.6.3 4.8.6.4 486.5 4.8.6.6 4.8.6.7 4.8.6.8 4.8.6.9 4.8.7.1 4.8.7.2 4.8.7.3 4.8.74 4.8.7.5 4.87.6 4.8.7.7 4.8.78 487.9 48.8.1 4.882 48.83 4.8.8.4 4.885 48.8.6 4.88.7 48.88 4.88.9 4.89.1 48.9.2 4.89.3 48.94 48.9.5 4.89.6 48.9.7 48.98 48.99 4.9.1.1 4.9.1.2 4.9.13 4.9.14 4.91.5 4.9.1.6 4.9.17 4.9.18 4.91.9 4.9.2.1 4.9.2.2 4.9.23 4.9.2.4 4.9.25 4.9.26 4.9.2.7 4.9.28 4.92.9 4.93.1 4.93.2 4.93.3 4.93.4 4.93.5 4.93.6 4.93.7 49.38 4.93.9 4.9.4.1 4.9.4.2 4.94.3 4.9.4.4 4.9.45 4.9.4.6 4.94.7 4.94.8 49.4.9 4.951 4.95.2 4.95.3 4.95.4 4.9.S.S 4.956 4.95.7 4.95.8 4.95.9 4.9.6.1 4.9.62 4.96.3 4.9.6.4 4.9.6.5 4.96.6 4.96.7 4.96.8 4.96.9 4.9.7.1 4.9.7.2 4.9.7.3 4.97.4 4.97.5 4.97.6 4.97.7 4.97.8 49.79 4.98.1 49.82 49.83 49.84 49.8.5 4.98.6 49.87 4.98.8 4.9-8.9 4.99.1 4.9.9.2 4.99.3 4.99.4 4.99.5 4.99.6 4.99.7 4.9.98 4.99.9 5.1.1.1 5.1.1.2 5.1.1.3 5.1.1.4 5.1.1.5 5.1.16 5.1.1.7 5.1.1.8 5.1.1.9 5.1.2.1 5.1.2.2 5.1.2.3 5.12.4 S.12.5 5.12.6 S.12.7 5.1.2.8 5.1.2.9 5.1.3.1 5.1.3.2 S.1.3.3 5.1.3.4 S.1.3.5 5.13.6 S.1.3.7 5.13.8 S.1.3.9 5.1.4.1 5.14.2 5.14.3 5.1.4.4 5.14.5 5.14.6 5.14.7 5.14.8 5.14.9 S.1.5.1 S.1.5.2 S.15.3 5.1.5.4 5.1.5.5 S.1.5.6 5.1.5.7 S.1.5.8 S.1.5.9 5.1.6.1 5.1.6.2 5.1.6.3 5.1.6.4 S.1.6.5 5.1.6.6 S.1.6.7 5.1.6.8 5.1.6.9 S.17.1 S.17.2 S.17.3 5.17.4 5.17.5 S.17.6 5.17.7 S.17.8 S.17.9 5.1.8.1 5.18.2 5.1.83 5.18.4 S.18.5 5.1.86 S.18.7 5.18.8 5.18.9 5.19.1 5.19.2 5.19.3 5.194 S.1.9.5 5.19.6 5.19.7 5.19.8 5.19.9 5.2.1.1 5.2.1.2 5.2.1.3 5.2.1.4 5.2.1.5 5.2.1.6 5.2.1.7 5.2.1.8 5.2.1.9 5.2.2.1 5.2.2.2 S.2.2.3 5.2.2.4 S.2.2.5 5.22.6 S.2.2.7 5.2.2.8 5.2.2.9 5.2.3.1 S.23.2 S.2.3.3 5.23.4 S.2.3.5 5.2.3.6 S.23.7 S.23.8 S.23.9 5.24.1 5.24.2 5.24.3 5.2.4.4 5.245 5.24.6 S.2.47 5.24.8 5.24.9 S.2.5.1 S.2.5.2 S.2.5.3 5.2.5.4 5.2.5.5 S.2.5.6 5.2.5.7 S.2.5.8 S.2.5.9 5.2.6.1 5.2.6.2 5.2.6.3 5.2.6.4 S.26.5 5.2. 6.6 S.2.6.7 5.2.6.8 5.2.6.9 S.2.7.1 S.2.7.2 S.2.7.3 S.2.7.4 5.2.7.5 S.2.7.6 5.2.7.7 S.2.7.8 S.2.7.9 5.2.8.1 5.2.8.2 5.2.8.3 5.2.84 5.2.8.5 5.2.8.6 5.2.8.7 5.2.8.8 5.2.89 5.2.9.1 5.2.9.2 S.29.3 5.2.9.4 S.2.9S 5.2.96 S.2.9.7 5.2.98 S.2.9.9 5.3.1.1 5.3.1.2 5.3.1.3 5.3.14 5.3.1.5 5.3.1.6 5.3.1.7 5.3.1.8 5.3.1.9 5.3.2.1 5.3.2.2 5.3.2.3 5.3.2.4 5.3.2.5 5.3.2.6 5.3.2.7 5.3.2.8 5.3.2.9 5.3.3.1 5.3.3.2 5.3.3.3 5.334 5.33.5 5.3.3.6 5.3.3.7 5.3.3.8 5.33.9 53.4.1 5.3.4.2 53.43 53.44 5.3.4.5 53.4.6 5.3.4.7 5.3.4.8 5.34.9 5.35.1 5.3.5.2 5.35.3 5.35.4 53.5.5 5.3.S.6 53.5.7 5.35.8 5.35.9 5.3.6.1 5.3.6.2 5.3.6.3 5.3.6.4 5.3.6.5 5.3.6.6 5.3.6.7 5.3.6.8 5.3.6.9 5.3.7.1 5.3.7.2 5.3.7.3 5.3.7.4 5.3.7.5 5.3.7.6 5.3.7.7 5.3.7.8 5.3.7.9 5.38.1 5.38.2 5.38.3 5.38.4 5.38.5 5.38.6 5.38.7 5.38.8 5.38.9 5.3.9.1 5.3.9.2 5.3.9.3 5.3.94 5.3.9.5 5.3.96 5.3.9.7 5.3.9.8 5.3.9.9 5.4.1.1 5.4.1.2 5.4.13 5.4.1.4 5.4.15 5.41.6 54.17 5.4.1.8 5.4.19 5.4.2.1 5.4.2.2 54.23 5.4.2.4 5.42.5 54.2.6 S.4.2.7 542.8 5.42.9 5.4.3.1 543.2 5.4.3.3 5.43.4 5.43.5 5.43.6 5.43.7 5.43.8 5.43.9 5.4.4.1 5.4.4.2 5.44.3 5.4.4.4 5.445 5.4.4.6 5.44.7 5.448 5.4.4.9 5.45.1 5.45.2 5.45.3 5.454 S.4.S.S 5.45.6 5.45.7 5.45.8 S.4.5.9 5.4.6.1 5.4.6.2 5.4.6.3 5.4.6.4 5.4.6.5 5.46.6 5.4.6.7 5.46.8 5.46.9 54.71 S.47.2 54.7.3 54.7.4 S47.5 54.7.6 5.47.7 S47.8 S.4.7.9 54.8.1 54.82 54.83 54.8.4 S.48.5 54.8.6 54.8.7 5.488 54.8.9 5.4.9.1 5.4.9.2 5.4.9.3 5.4.9.4 5.4.9.5 5.4.9.6 5.4.9.7 5.4.9.8 5.4.9.9 S.S.1.1 S.S.1.2 S.S. 1.3 5.5.1.4 5.5.1.5 S.S.1.6 5.5.1.7 S.S.1.8 S.S.1.9 S.S.2.1 S.S.2.2 S.S.23 5.5.2.4 5.5.2.5 S.S.2.6 5.5.2.7 S.S.2.8 S.S.2.9 S.S.3.1 S.S.3.2 S.S.3.3 S.S.34 5.5.3.5 S.S.3.6 5.5.3.7 S.S.3.8 S.S.3.9 554.1 554.2 S.S.4.3 5.5.44 S.S.4.5 5.5.46 S.S.4.7 S.S.4.8 S.S.4.9 5.5.5.1 5.5.5.2 5.5.5.3 S.S.S.4 5.5.5.5 S.S.S.6 5.5.5.7 5.55.8 5.5.5.9 S.S.6.1 S.S.6.2 S.S.6.3 5.56.4 S.S.6.5 S.S.6.6 5.5.6.7 S.S. 6.8 S.S.6.9 5.5.7.1 5.5.7.2 5.5.7.3 S.S. 7.4 5.5.7.5 5.5.7.6 5.5.7.7 5.5.7.8 5.5.7.9 S.S.8.1 S.S.8.2 S.S.8.3 S.S.84 5.5.8.5 S.S.8.6 5.5.8.7 S.S.8.8 S.S.8.9 S.S.9.1 S.S.9.2 S.S.9.3 S.S.9.4 5.59.5 S.S.9.6 5.59.7 S.S.9.8 S.S.9.9 5.6.1.1 5.6.1.2 56.13 5.6.1.4 S.6.1.5 5.6.16 S.6.17 56.1.8 5.6.19 5.6.2.1 5.6.2.2 56.23 5.6.2.4 S.6.2.5 5.6.2.6 S.6.2.7 5.6.2.8 5.6.29 5.6.3.1 5.6.32 S.6.3.3 5.6.3.4 S.6.3.5 5.6.36 S.6.3.7 5.638 S.6.3.9 56.4.1 56.4.2 56.4.3 5.6.4.4 56.4.5 56.4.6 56.4.7 56.4.8 5.64.9 S.6.5.1 S. 6.5.2 S. 6.5.3 56.5.4 S.6.S.S S. 6.5.6 56.5.7 S. 6.5.8 S.6.5.9 56.6.1 56.6.2 56.6.3 56.64 S.6.6.5 56.6.6 S.6.6.7 56.68 56.6.9 S.6.7.1 S.6.7.2 S.6.7.3 5.6.7.4 5.6.7.5 S.6.7.6 5.6.7.7 S.6.7.8 S.6.7.9 5.6.8.1 56.8.2 56.83 56.8.4 S. 6.8.5 5.6.86 S.6.8.7 5.68.8 5.6.8.9 56.9.1 56.9.2 S.6.9.3 56.94 S.6.9.5 56.96 S. 6.9.7 56.9.8 56.9.9 5.7.1.1 5.7.1.2 5.7.1.3 5.7.1.4 5.7.1.5 5.7.1.6 5.7.1.7 5.7.1.8 5.7.1.9 5.7.2.1 5.7.2.2 5.7.2.3 5.7.2.4 5.7.2.5 5.7.26 5.7.2.7 S. 7.2.8 5.7.2.9 5.7.3.1 5.7.3.2 5.7.3.3 5.7.34 5.7.3.5 5.7.3.6 5.7.3.7 5.7.3.8 S. 7.3.9 5.74.1 US 8,080,536 B2 57 58 TABLE 1-continued 5.7.4.2 5.7.4.3 5.7.44 5.74.5 57.4.6 5.74.7 5.74.8 5.7.4.9 5.7.5.1 5.7.5.2 5.7.5.3 5.75.4 5.7.5.5 5.7.5.6 5.7.57 5.7.5.8 5.7.5.9 5.7.6.1 S. 7.6.2 5.7.6.3 57.6.4 5.7.6.5 5.7.6.6 5.7.6.7 5.7.6.8 5.7.6.9 5.7.7.1 57.7.2 57.7.3 5.7.7.4 57.7.5 5.7.7.6 5.7.7.7 57.7.8 5.7.7.9 5.78.1 5.78.2 5.7.8.3 5.7.84 5.78.5 5.78.6 5.78.7 5.78.8 5.78.9 5.7.9.1 S.79.2 S.79.3 5.7.9.4 5.7.9.5 5.7.9.6 5.7.9.7 S.79.8 5.7.9.9 5.8.1.1 5.8.1.2 5.8.1.3 5.8.1.4 5.8.1.5 5.8.1.6 5.8.1.7 5.8.1.8 5.8.19 5.8.2.1 5.8.2.2 5.8.2.3 5.8.2.4 5.8.2.5 5.8.2.6 5.8.2.7 5.8.2.8 5.8.2.9 5.83.1 5.8.3.2 5.83.3 58.34 5.835 5.83.6 5.8.3.7 5.83.8 5.83.9 58.4.1 5.842 5.84.3 5.84.4 5.84.5 5.84.6 5.84.7 58.48 58.49 5.8.5.1 5.8.5.2 5.8.5.3 5.8.5.4 5.8.5.5 5.8.S.6 5.8.5.7 5.85.8 5.8.5.9 5.8.6.1 5.8.6.2 5.8.6.3 5.8.6.4 5.8.6.5 5.8.6.6 5.8.6.7 5.8.6.8 5.8.6.9 5.8.7.1 5.87.2 5.8.7.3 5.87.4 5.8.7.5 5.87.6 5.8.7.7 5.8.7.8 5.87.9 5.88.1 58.8.2 5.88.3 58.84 5.8.8.5 5.88.6 5.8.8.7 58.88 5.8.8.9 5.89.1 5.89.2 5.8.9.3 5.894 5.8.9.5 5.89.6 5.89.7 5.8.9.8 5.89.9 5.9.1.1 5.9.1.2 S.91.3 5.9.14 S.91.5 5.91.6 S.9.17 5.9.1.8 5.9.19 59.2.1 5.9.2.2 S.9.2.3 5.92.4 S.9.2.5 5.92.6 5.9.2.7 59.28 S.92.9 5.9.3.1 S.93.2 5.9.3.3 5.93.4 5.93.5 S.9.3.6 S.93.7 5.9.3.8 S.93.9 5.94.1 5.94.2 5.94.3 5.944 S.9.45 5.94.6 S.94.7 5.948 59.49 S.9.5.1 5.95.2 5.9.5.3 S.9.S.4 5.9.5.5 S.9.S.6 5.95.7 S.9.S.8 S.9.S.9 59.6.1 59.6.2 S.96.3 59.6.4 S.9.6.5 59.6.6 S.96.7 5.96.8 59.6.9 S.9.7.1 5.9.7.2 S.9.7.3 5.9.7.4 5.9.7.5 S.9.7.6 5.9.7.7 S.9.7.8 S.97.9 59.8.1 5.98.2 5.9.83 59.8.4 S.98.5 59.8.6 S.98.7 5.9.8.8 5.9-8.9 5.99.1 S.9.9.2 5.99.3 5.99.4 5.99.5 5.99.6 5.99.7 5.9-9.8 5.99.9 6.1.1.1 6.1.1.2 6.1.1.3 6.1.1.4 6.1.1.5 6.1.1.6 6.11.7 6.1.1.8 6.1.1.9 6.1.2.1 6.1.2.2 6.1.2.3 6.1.2.4 6.1.25 6.1.2.6 6.1.2.7 6.1.2.8 6.12.9 6.1.3.1 6.13.2 6.1.3.3 6.1.3.4 6.13.5 6.13.6 6.13.7 6.13.8 6.13.9 6.1.4.1 6.1.4.2 6.1.4.3 6.1.4.4 6.14.5 6.1.4.6 6.14.7 6.1.4.8 6.1.4.9 6.1.5.1 6.15.2 6.15.3 6.1.5.4 6.1.S.S 6.1.5.6 6.1.5.7 6.1.58 6.1.5.9 6.1.6.1 6.1.6.2 6.16.3 6.1.6.4 6.16.5 6.1.6.6 6.16.7 6.1.6.8 6.16.9 6.17.1 6.17.2 6.17.3 6.17.4 6.17.5 6.17.6 6.17.7 6.1.78 6.17.9 6.1.8.1 6.18.2 6.18.3 6.1.8.4 6.18.5 6.18.6 6.18.7 6.18.8 6.1.89 6.19.1 6.192 6.1.93 6.1.9.4 6.19.5 6.19.6 6.19.7 6.1.98 6.19.9 6.2.1.1 6.2.1.2 6.2.1.3 6.2.1.4 6.2.1.5 6.2.1.6 6.2.1.7 6.2.1.8 6.2.19 6.2.2.1 6.2.2.2 6.2.23 6.2.2.4 6.2.2.5 6.2.2.6 6.2.2.7 6.2.2.8 6.2.2.9 6.2.3.1 6.23.2 6.2.3.3 6.2.3.4 6.2.35 6.2.3.6 6.2.3.7 6.2.38 6.2.3.9 6.2.4.1 6.2.4.2 6.2.4.3 6.2.4.4 6.2.45 6.2.4.6 6.2.47 6.2.4.8 6.2.49 6.2.5.1 6.25.2 6.2.5.3 6.25.4 6.2.S.S 6.2.5.6 6.2.5.7 6.25.8 6.2.5.9 6.2.6.1 6.2.6.2 6.2.6.3 6.2.6.4 6.26.5 6.26.6 6.2. 6.7 6.26.8 6.2.6.9 6.2.7.1 6.2.7.2 6.2.7.3 6.2.7.4 6.2.7.5 6.2.7.6 6.2.7.7 6.2.7.8 6.2.7.9 6.2.8.1 6.2.8.2 6.2.83 6.2.8.4 6.2.85 6.2.86 6.28.7 6.2.8.8 6.2.8.9 6.2.9.1 6.2.92 6.2.9.3 6.2.94 6.2.9.5 6.29.6 6.2.9.7 6.29.8 6.2.99 6.3.1.1 6.3.1.2 6.3.1.3 6.3.1.4 6.3.1.5 6.3.1.6 6.3.1.7 6.3.1.8 6.3.1.9 6.3.2.1 6.3.2.2 6.3.2.3 6.3.2.4 6.3.2.5 6.3.2.6 6.3.2.7 6.3.2.8 6.3.2.9 6.3.3.1 6.3.3.2 6.3.3.3 6.3.3.4 6.3.3.5 6.3.3.6 6.3.3.7 6.3.3.8 6.3.3.9 6.3.4.1 6.3.4.2 6.3.4.3 6.3.4.4 6.3.4.5 6.3.4.6 6.3.4.7 6.3.4.8 6.3.4.9 6.35.1 6.35.2 6.35.3 6.35.4 6.3.S.S 6.35.6 6.3.5.7 6.35.8 6.35.9 6.3.6.1 6.3.6.2 6.3.6.3 6.3.6.4 6.3.6.5 6.3.6.6 6.3.6.7 6.3.6.8 6.3.6.9 6.3.7.1 6.3.7.2 6.3.7.3 6.3.7.4 6.3.7.5 6.3.7.6 6.3.7.7 6.3.7.8 6.3.7.9 6.3.8.1 6.38.2 6.3.8.3 6.38.4 6.3.85 6.38.6 6.3.8.7 6.38.8 6.3-8.9 6.39.1 6.3.9.2 6.3.93 6.3.9.4 6.3.9.5 6.3.96 6.3.9.7 6.3.98 6.3.9.9 6.4.1.1 6.4.1.2 6.4.1.3 6.4.1.4 6.4.15 6.4.1.6 6.4.17 6.4.1.8 6.4.1.9 6.4.2.1 6.4.2.2 6.4.2.3 6.4.2.4 6.4.25 6.4.2.6 6.4.2.7 6.4.2.8 6.42.9 6.4.3.1 6.4.3.2 6.4.3.3 6.4.3.4 6.4.35 6.4.36 6.4.3.7 6.4.38 6.4.3.9 6.4.4.1 6.4.4.2 6.4.4.3 6.4.4.4 6.4.4.5 6.4.4.6 6.4.4.7 6.4.4.8 6.4.4.9 6.45.1 6.45.2 6.45.3 6.4.5.4 6.45.5 6.456 6.45.7 6.458 6.45.9 6.4.6.1 6.4.6.2 6.4.6.3 6.4.6.4 6.4.6.5 6.4.6.6 6.4.6.7 6.4.6.8 6.4.6.9 6.47.1 6.47.2 6.4.7.3 6.4.7.4 6.4.75 6.47.6 6.4.7.7 6.47.8 6.47.9 6.4.8.1 6.4.8.2 6.4.83 6.4.8.4 6.4.8.5 6.4.86 6.4.8.7 6.4.8.8 6.4.89 6.4.9.1 6.4.9.2 6.4.9.3 6.4.9.4 6.4.9.5 6.4.9.6 6.4.9.7 6.4.9.8 6.4.9.9 6.5.1.1 6.5.1.2 6.5.13 6.5.1.4 6.5.1.5 6.5.1.6 6.5.1.7 6.5.1.8 6.5.19 6.5.2.1 6.5.2.2 6.5.23 6.5.2.4 6.5.2.5 6.5.2.6 6.5.2.7 6.5.2.8 6.5.2.9 6.5.3.1 6.5.3.2 6.5.3.3 6.5.3.4 6.5.3.5 6.5.3.6 6.5.3.7 6.5.3.8 6.5.3.9 6.54.1 6.5.4.2 6.543 6.5.44 6.5.45 6.5.46 6.5.4.7 6.5.48 6.5.4.9 6.S.S.1 6.S.S.2 6.S.S.3 6.55.4 6.S.S.S 6.S.S.6 6.5.5.7 6.S.S.8 6.S.S.9 6.5.6.1 6.5-6.2 6.56.3 6.56.4 6.5-6.5 6.5.6.6 6.5.6.7 6.5-6.8 6.5.6.9 6.5.7.1 6.5.7.2 6.5.7.3 6.5.7.4 6.5.7.5 6.5.7.6 6.5.7.7 6.5.7.8 6.5.7.9 6.5.8.1 6.5.8.2 6.5.8.3 6.5-8.4 6.5.8.5 6.5.8.6 6.5.8.7 6.5.8.8 6.5-8.9 6.59.1 6.5.9.2 6.5.93 6.5.94 6.5.9.5 6.59.6 6.5.9.7 6.5-9.8 6.5.99 6.6.1.1 6.6.1.2 6.6.13 6.6.1.4 6.6.15 6.6.1.6 6.6.17 6.6.1.8 6.6.19 6.6.2.1 6.6.2.2 6.6.23 6.6.2.4 6.6.25 6.6.2.6 6.6.2.7 6.6.2.8 6.6.29 6.63.1 6.6.32 6.6.3.3 6.6.3.4 6.63.5 6.63.6 6.6.3.7 6.638 6.63.9 6.6.4.1 6.6.4.2 6.6.43 6.6.4.4 6.64.5 6.6.4.6 6.64.7 6.6.4.8 6.64.9 6.6.5.1 6.6.5.2 66.53 6.6.54 6.6.S.S 6.65.6 6.6.5.7 6.658 6. 6.5.9 6.6.6.1 6.6.6.2 6.6.6.3 6.6.6.4 6. 6.65 6.6. 6.6 6.66.7 6.6. 6.8 6.66.9 6.6.7.1 6.6.7.2 6.6.7.3 6.6.7.4 6.6.7.5 6.6.7.6 6.6.7.7 6.6.7.8 6.6.7.9 6.68.1 6. 6.8.2 6.68.3 6.6.84 6.6.8.5 6.6.86 6. 6.8.7 6.6.8.8 6.68.9 6.6.9.1 6.6.9.2 6.6.93 6.69.4 6.6.95 6.6.9.6 6.6.9.7 6.6.98 6.6.9.9 6.7.1.1 6.7.1.2 6.7.1.3 6.7.1.4 6.7.1.5 6.7.1.6 6.7.1.7 6.7.1.8 6.7.1.9 6.7.2.1 6.7.2.2 6.7.2.3 6.7.24 6.7.2S 6.7.26 6.7.2.7 6.7.2.8 6.7.2.9 6.7.3.1 6.7.3.2 6.7.3.3 6.7.34 6.73.5 6.7.36 6.73.7 6.738 6.7.3.9 6.74.1 6.7.4.2 6.7.4.3 6.7:44 6.7.45 6.74.6 6.7.4.7 6.74.8 6.74.9 6.7.5.1 6.7.5.2 6.7.5.3 6.75.4 6.7.5.5 6.7.5.6 6.7.5.7 6.7.58 6.75.9 6.7.6.1 6.7.6.2 6.7.63 6.7.6.4 6.7.6.5 6.7.6.6 6.7.6.7 6.76.8 6.7.6.9 6.77.1 6.7.7.2 6.7.7.3 6.7.74 6.7.7.5 6.7.7.6 6.7.7.7 6.7.7.8 6.7.7.9 6.7.8.1 6.7.82 6.7.83 6.7.84 6.7.8.5 6.7.86 6.7.8.7 6.7.8.8 6.7.8.9 6.79.1 6.79.2 6.79.3 6.79.4 6.7.9.5 6.79.6 6.7.9.7 6.79.8 6.7.9.9 6.8.1.1 6.8.1.2 6.8.1.3 6.8.1.4 6.8.1.5 6.8.1.6 6.8.1.7 6.8.1.8 6.8.19 6.8.2.1 6.8.2.2 6.82.3 6.8.2.4 6.82.5 6.82.6 6.8.2.7 6.8.2.8 6.8.2.9 US 8,080,536 B2 59 60 TABLE 1-continued 6.8.31 6.8.32 6.8.3.3 6.834 6.8.35 6.83.6 6.8.3.7 68.38 6.83.9 6.8.4.1 6.8.4.2 6.84.3 6.8.4.4 6.84.5 6.846 6.84.7 6.84.8 6.8.49 6.85.1 6.8.5.2 6.8.5.3 6.854 6.8.S.S 6.85.6 6.8.5.7 6.85.8 6.85.9 6.8.6.1 6.8.6.2 6.8.6.3 6.8.6.4 6.8.6.5 6.8.6.6 6.8.6.7 6.8.6.8 6.8.6.9 6.8.7.1 6.8.7.2 6.8.7.3 6.8.74 6.8.7.5 6.87.6 6.8.7.7 6.8.78 6.8.79 6.88.1 6.882 6.8.83 6.884 6.8.85 6.8.8.6 6.88.7 6.8.8.8 6.88.9 6.8.9.1 6.89.2 6.89.3 68.9.4 6.895 6.8.96 68.9.7 6.8.98 6.8.9.9 6.9.1.1 6.9.1.2 6.9.13 6.9.14 6.9.15 6.91.6 6.9.17 6.9.18 6.9.19 6.9.2.1 6.9.2.2 6.9.23 6.92.4 6.9.25 6.92.6 6.92.7 6.9.28 6.92.9 6.9.31 6.93.2 6.933 6.9.34 6.93.5 6.93.6 6.93.7 6.93.8 6.93.9 6.9.4.1 6.94.2 6.94.3 6.9.4.4 6.9.45 6.94.6 6.94.7 6.9.4.8 6.94.9 6.951 6.95.2 6.95.3 6.95.4 6.9.S.S 6.956 6.95.7 6.95.8 6.95.9 6.9.6.1 6.9.62 6.96.3 6.96.4 6.9.6.5 6.96.6 6.96.7 6.96.8 6.96.9 6.9.7.1 6.9.72 6.9.7.3 6.9.7.4 6.9.7.5 6.97.6 6.97.7 6.97.8 6.9.7.9 6.98.1 6.9.8.2 6.9.83 6.9.84 6.9.8.5 6.98.6 6.98.7 6.98.8 6.98.9 6.99.1 6.9.9.2 6.99.3 6.9.94 6.9.95 6.99.6 6.9.9.7 6.9.9.8 6.99.9 7.1.1.1 7.1.1.2 7.1.1.3 7.1.1.4 7.1.1.5 7.1.16 7.1.1.7 7.1.1.8 7.1.1.9 7.1.2.1 7.1.2.2 7.1.2.3 7.1.24 7.1.2.5 7.1.2.6 7.1.2.7 7.1.2.8 7.1.2.9 7.1.3.1 7.1.3.2 7.1.3.3 7.1.3.4 7.1.3.5 7.13.6 7.1.3.7 7.1.3.8 7.1.3.9 7.14.1 7.14.2 7.14.3 7.14.4 7.14.5 7.14.6 7.14.7 7.14.8 7.14.9 7.1.5.1 7.1.5.2 7.1.5.3 71.54 7.1.5.5 7.1.5.6 7.1.5.7 7.1.5.8 7.1.5.9 7.1.6.1 7.16.2 7.1.6.3 7.16.4 7.1.6.5 7.16.6 7.1.6.7 7.1.6.8 7.1.6.9 7.1.7.1 7.17.2 7.17.3 7.17.4 7.17.5 7.17.6 7.17.7 7.17.8 7.17.9 7.18.1 7.18.2 7.18.3 7.18.4 7.18.5 7.18.6 7.18.7 7.18.8 7.18.9 7.19.1 71.9.2 7.19.3 71.94 7.1.9.5 7.19.6 7.1.9.7 71.98 7.19.9 7.2.1.1 7.2.1.2 7.2.1.3 7.2.1.4 7.2.1.5 7.2.1.6 7.2.1.7 7.2.1.8 7.2.19 7.2.2.1 7.2.2.2 7.2.2.3 7.2.2.4 7.2.2.5 7.2.2.6 7.2.2.7 7.2.2.8 7.2.2.9 7.2.3.1 7.2.3.2 7.2.3.3 7.2.34 7.2.3.5 7.2.3.6 7.2.3.7 7.2.3.8 7.2.3.9 7.2.4.1 7.24.2 7.2.43 7.2.4.4 7.2.45 7.2.4.6 7.2.4.7 7.2.48 7.2.49 7.2.5.1 7.2.5.2 7.2.5.3 7.2.5.4 7.2.5.5 7.2.5.6 7.2.5.7 7.2.5.8 7.2.5.9 7.2.6.1 7.2.6.2 7.2.6.3 7.2.64 7.2.6.5 7.26.6 7.2.6.7 7.2. 6.8 7.2.6.9 7.2.7.1 7.2.7.2 7.2.7.3 7.2.7.4 7.2.7.5 7.2.7.6 7.2.7.7 7.2.7.8 7.2.7.9 7.28.1 7.28.2 7.2.8.3 7.28.4 7.2.8.5 7.28.6 7.2.8.7 7.28.8 7.28.9 7.2.9.1 7.2.9.2 7.2.9.3 7.2.9.4 7.2.9.5 7.2.9.6 7.2.9.7 7.2.9.8 7.2.9.9 7.3.1.1 7.3.1.2 7.3.1.3 7.3.14 7.3.1.5 7.3.1.6 7.3.1.7 7.3.1.8 7.3.1.9 7.3.2.1 7.3.2.2 7.3.2.3 7.3.2.4 7.3.2.5 7.3.2.6 7.3.2.7 7.3.2.8 7.3.2.9 7.3.3.1 7.3.3.2 7.3.3.3 7.3.3.4 7.3.3.5 7.3.3.6 7.3.3.7 7.3.3.8 7.3.3.9 7.3.4.1 7.3.4.2 7.3.4.3 7.3.4.4 7.3.4.5 7.3.4.6 7.3.4.7 7.3.4.8 7.34.9 7.35.1 7.3.5.2 7.3.5.3 7.35.4 7.3.5.5 7.35.6 7.3.5.7 7.35.8 7.35.9 7.3.6.1 7.3.6.2 7.3.6.3 7.3.6.4 7.3.6.5 7.3.6.6 7.3.6.7 7.3.6.8 7.3.6.9 7.3.7.1 7.3.7.2 7.3.7.3 7.3.7.4 7.3.7.5 7.3.7.6 7.3.7.7 7.3.7.8 7.3.7.9 7.3.8.1 7.38.2 7.38.3 7.38.4 7.3.85 7.38.6 7.38.7 7.38.8 7.3.8.9 7.39.1 7.3.9.2 7.3.9.3 7.39.4 7.3.9.5 7.3.96 7.3.9.7 7.3.9.8 7.3.9.9 7.4.1.1 74.1.2 7.4.13 7.4.1.4 74.15 7.4.1.6 74.17 7.4.1.8 74.1.9 7.4.2.1 7.4.2.2 7.4.23 7.4.2.4 742.5 7.4.2.6 7.4.2.7 742.8 7.42.9 7.4.3.1 7.4.3.2 7.4.3.3 743.4 7.43.5 7.4.36 7.43.7 7.4.38 7.43.9 7.4.4.1 7.4.4.2 7.4.4.3 7.4.4.4 7:44.5 7:44.6 7.447 7:44.8 7.4.4.9 745.1 7.4S.2 7.4.5.3 7.454 74.S.S 7.456 7.45.7 745.8 7.45.9 7.4.6.1 7.4.6.2 7.4.6.3 7.46.4 7.46.5 7.46.6 7.4.6.7 7.46.8 7.46.9 74.7.1 7.47.2 74.7.3 74.7.4 74.7.5 74.7.6 74.7.7 74.7.8 74.7.9 74.8.1 7:48.2 7:48.3 74.84 7.48.5 748.6 7.4.8.7 748.8 7.4.89 7.4.9.1 7.4.9.2 7.4.9.3 7.4.9.4 7.4.9.5 7.4.9.6 7.4.9.7 7.4.9.8 7.4.9.9 7.5.1.1 7.5.1.2 7.5.1.3 7.5.1.4 7.5.1.5 7.5.1.6 7.5.1.7 7.5.1.8 7.5.1.9 7.5.2.1 7.5.2.2 7.5.2.3 7.5.2.4 7.5.2.5 7.5.2.6 7.5.2.7 7.5.2.8 7.5.2.9 7.5.3.1 7.5.3.2 7.5.3.3 7.5.3.4 7.5.3.5 7.5.3.6 7.5.3.7 7.5.3.8 7.5.3.9 754.1 7.5.4.2 7.5.4.3 7.5.44 7.5.45 7.5.46 7.5.4.7 7.5.48 7.5.4.9 7.5.5.1 7.5.5.2 7.5.5.3 7.S.S.4 7.5.5.5 7.5.5.6 7.5.5.7 7.5.5.8 7.5.5.9 7.5.6.1 7.5.6.2 7.5.6.3 7.56.4 7.5.6.5 7.5.6.6 7.5.6.7 7.5.6.8 7.5.6.9 7.5.7.1 7.5.7.2 7.5.7.3 7.5.7.4 7.5.7.5 7.5.7.6 7.5.7.7 7.5.7.8 7.5.7.9 7.5.8.1 7.5.8.2 7.5.8.3 7.5.8.4 7.5.8.5 7.5.8.6 7.5.8.7 7.5.8.8 7.5.8.9 7.5.9.1 7.5.9.2 7.5.9.3 7.5.94 7.5.9.5 7.59.6 7.5.9.7 7.5.9.8 7.5.9.9 7.6.1.1 7.6.1.2 7.6.13 7.6.1.4 7.6.1.5 7.6.16 7.6.17 7.6.1.8 7.6.19 7.6.2.1 7.6.2.2 7.6.2.3 7.6.2.4 7.6.2.5 7.6.2.6 7.6.2.7 7.6.2.8 7.6.29 7.6.3.1 7.6.3.2 7.6.3.3 7.6.3.4 7.6.3.5 7.6.36 7.6.3.7 7.6.3.8 7.6.3.9 7.6.4.1 7.6.4.2 7.6.43 7.6.44 7.6.45 7.6.46 7.6.4.7 7.6.4.8 7.6.49 7.6.5.1 7.6.5.2 7.6.5.3 7.654 7.6.5.5 7.6.5.6 7.6.5.7 7.6.5.8 7.6.5.9 7.6.6.1 7.6.6.2 7.6.6.3 7.6.6.4 7.6.6.5 7.6.6.6 7.6.6.7 7.6.68 7.6.6.9 7.6.7.1 7.6.7.2 7.6.7.3 7.6.7.4 7.6.7.5 7.6.7.6 7.6.7.7 7.6.7.8 7.6.7.9 7.6.8.1 7.6.82 7.6.8.3 7.6.8.4 7.6.8.5 7.6.8.6 7.6.8.7 7.6.8.8 7.6.8.9 7.6.9.1 7.6.9.2 7.6.9.3 7.6.94 7.6.9.5 7.6.96 7.6.9.7 7.6.98 7.6.9.9 7.7.1.1 7.7.1.2 7.7.1.3 7.7.1.4 7.7.1.5 7.7.1.6 7.7.1.7 7.7.1.8 7.7.1.9 7.7.2.1 7.7.2.2 7.7.2.3 7.7.2.4 7.7.2.5 7.7.26 7.72.7 7.7.2.8 7.7.2.9 7.7.3.1 7.7.3.2 7.7.3.3 7.73.4 7.7.3.5 7.7.3.6 7.7.3.7 7.7.3.8 7.7.3.9 7.7.4.1 7.74.2 7.7.4.3 77.4.4 7.74.5 7.7.46 7.74.7 7.7.4.8 7.74.9 7.7.5.1 7.7.5.2 7.7.5.3 7.75.4 7.75.5 7.7.5.6 7.75.7 7.75.8 7.7.5.9 7.7.6.1 7.7.6.2 7.7.6.3 7.7.6.4 7.76.5 7.7.6.6 7.7.6.7 7.76.8 7.7.6.9 7.77.1 7.77.2 7.77.3 7.7.7.4 7.77.5 7.7.7.6 7.7.7.7 7.77.8 7.77.9 7.7.8.1 7.7.8.2 7.7-8.3 7.7.84 7.7.8.5 7.7.8.6 7.7.8.7 7.7.8.8 7.7-8.9 7.7.9.1 7.79.2 7.79.3 7.7.9.4 7.79.5 7.7.9.6 7.79.7 7.7.9.8 7.7.9.9 7.81.1 7.8.1.2 7.8.1.3 7.8.1.4 7.8.1.5 7.8.1.6 7.8.1.7 7.8.1.8 7.8.19 7.8.2.1 7.8.2.2 7.8.2.3 78.24 7.8.2.5 7.8.2.6 7.8.2.7 7.8.2.8 7.8.2.9 7.8.3.1 78.3.2 7.83.3 78.34 78.35 7.8.3.6 78.3.7 7.83.8 7.83.9 7.84.1 78.4.2 78.4.3 7.84.4 7.84.5 78.4.6 7.8.4.7 7.84.8 7.84.9 7.8.5.1 7.85.2 7.8.5.3 7.8.5.4 7.8.5.5 7.8.5.6 7.8.5.7 7.85.8 7.85.9 7.8.6.1 7.8.6.2 7.8.6.3 7.8.6.4 7.8.6.5 7.8.6.6 7.8.6.7 78.68 7.8.6.9 78.7.1 7.8.7.2 78.7.3 7.87.4 7.8.7.5 78.7.6 78.7.7 7.8.7.8 78.7.9 7.88.1 7.88.2 7.88.3 7.88.4 7.88.5 78.86 7.88.7 7.88.8 7.88.9 7.89.1 7.8.9.2 7.89.3 7.89.4 7.8.9.5 7.89.6 7.8.9.7 7.8.9.8 7.89.9 7.9.1.1 7.9.1.2 7.9.13 7.91.4 7.91.5 7.91.6 7.91.7 7.9.1.8 US 8,080,536 B2 61 62 TABLE 1-continued 7.9.19 7.9.2.1 7.9.2.2 7.9.23 7.9.24 7.9.2.5 7.92.6 7.9.2.7 7.92.8 7.9.2.9 7.9.3.1 7.93.2 7.9.3.3 7.93.4 7.9.3S 7.9.3.6 79.3.7 7.9.3.8 7.93.9 7.94.1 7.942 7.94.3 7.944 7.9.45 7.946 7.94.7 7.94.8 7.94.9 7.95.1 7.9.S.2 79.5.3 7.9S4 79.5.5 79.5.6 79.5.7 79.5.8 7.95.9 79.6.1 79.6.2 7.9.6.3 79.6.4 7.9.6.5 79.6.6 79.6.7 79.6.8 79.6.9 7.97.1 7.9.7.2 7.9.7.3 7.9.7.4 7.9.7.5 7.97.6 7.9.7.7 7.97.8 7.9.7.9 7.98.1 7.9.8.2 7.9.8.3 7.9.84 7.9.8.5 7.9.8.6 7.98.7 7.9.8.8 7.98.9 7.99.1 7.9.9.2 7.99.3 7.99.4 7.99.5 7.99.6 7.9.9.7 7.99.8 7.99.9 8.1.1.1 8.1.1.2 8.1.1.3 8.1.1.4 8.1.1.5 8.1.1.6 8.11.7 8.1.1.8 8.1.19 8.1.2.1 8.1.2.2 8.1.2.3 8.1.2.4 8.1.25 8.12.6 8.1.2.7 8.1.2.8 8.12.9 8.1.3.1 8.1.3.2 8.1.3.3 8.13.4 8.1.3.5 8.13.6 8.1.3.7 8.13.8 8.1.3.9 8.1.4.1 8.1.4.2 8.14.3 8.1.4.4 8.14.5 8.1.4.6 8.14.7 81.48 8.14.9 8.15.1 8.15.2 8.15.3 8.15.4 8.1.S.S 8.15.6 8.15.7 8.15.8 8.15.9 8.1.6.1 8.1.6.2 8.1.6.3 8.1.6.4 8.1.6.5 8.1.6.6 8.1.6.7 8.1.6.8 8.1.6.9 8.17.1 8.17.2 8.17.3 8.17.4 8.1.75 8.17.6 8.17.7 8.17.8 8.17.9 8.18.1 8.18.2 81.83 8.18.4 8.18.5 8.18.6 8.18.7 8.18.8 8.18.9 8.19.1 8.192 8.19.3 8.1.94 8.1.9.5 8.19.6 8.1.97 8.19.8 8.19.9 8.2.1.1 8.2.1.2 8.2.1.3 8.2.1.4 8.2.1.5 8.2.1.6 8.2.1.7 8.2.1.8 8.2.19 8.2.2.1 8.2.2.2 8.223 8.2.2.4 8.2.2.5 8.22.6 8.2.2.7 8.2.2.8 8.22.9 8.23.1 8.23.2 82.3.3 8.23.4 8.23.5 82.3.6 82.3.7 8.23.8 82.3.9 8.2.4.1 8.2.4.2 8.24.3 8.2.4.4 8.24.5 8.2.4.6 82.47 8.24.8 82.49 8.2.5.1 8.252 8.2.5.3 8.25.4 8.2.S.S 8.256 8.2.5.7 8.25.8 8.25.9 8.2.6.1 82.6.2 8.26.3 8.2.6.4 8.26.5 8.26.6 8.26.7 8.26.8 82.6.9 8.2.7.1 8.2.7.2 8.2.7.3 8.2.7.4 8.2.7.5 8.2.7.6 8.2.7.7 8.2.7.8 8.2.7.9 82.8.1 82.82 8.283 82.84 8.285 8.28.6 82.87 8.2.8.8 8.28.9 8.29.1 82.9.2 8.29.3 8.29.4 8.29.5 8.29.6 8.2.9.7 8.29.8 8.299 8.3.1.1 8.3.1.2 8.3.1.3 8.314 8.3.1.5 8.3.1.6 8.3.1.7 8.3.1.8 8.3.1.9 8.3.2.1 8.3.2.2 8.3.2.3 8.3.2.4 8.3.2.5 8.3.2.6 8.3.2.7 8.3.2.8 8.3.2.9 8.33.1 8.33.2 8.33.3 8.33.4 8.33.5 8.33.6 8.33.7 83.38 8.33.9 8.3.4.1 8.3.4.2 8.3.4.3 8.3.4.4 8.345 8.3.4.6 8.3.4.7 8.3.4.8 8.3.4.9 8.35.1 8.3.5.2 8.35.3 8.3.54 8.3.S.S 8.35.6 8.3.5.7 8.35.8 8.3.5.9 8.3.6.1 8.3.6.2 8.3.6.3 8.3.6.4 8.3.6.5 8.3.6.6 8.3.6.7 8.3.6.8 8.3.6.9 8.3.7.1 8.3.7.2 8.3.7.3 8.3.7.4 8.3.7.5 8.3.7.6 8.3.7.7 8.3.7.8 8.3.7.9 8.38.1 8.38.2 83.8.3 8.38.4 8.3.8.5 8.38.6 8.38.7 8.38.8 8.3-8.9 8.39.1 8.392 8.3.93 8.39.4 8.3-9.5 8.3.96 8.39.7 8.3-9.8 8.39.9 8.4.1.1 8.4.1.2 8.4.1.3 8.4.1.4 84.15 8.4.1.6 8.4.17 84.1.8 8.4.19 8.4.2.1 8.4.2.2 84.23 8.4.2.4 8.4.25 8.4.2.6 8.4.2.7 8.42.8 8.429 8.4.31 8.432 8.4.3.3 8.4.3.4 8.43.5 8.436 8437 8.4.38 8.43.9 8.4.4.1 8.4.4.2 8.4.4.3 8.4.4.4 84.45 8.4.4.6 8.447 8.4.4.8 8.4.4.9 8.45.1 84.52 8.45.3 8.45.4 8.4.S.S 8.45.6 8.45.7 8.458 8.45.9 8.4.6.1 8.4.6.2 8.4.6.3 8.4.6.4 8:46.5 8.4.6.6 8.4.6.7 8.4.6.8 8.46.9 847.1 8.4.72 8.4.7.3 84.74 8.4.7.5 8.47.6 8.47.7 847.8 84.79 8.4.8.1 848.2 8.4.83 8.4.8.4 8.48.5 8.4.86 848.7 8.48.8 8.4.89 84.91 8.4.9.2 8.4.9.3 84.94 8.4.9.5 84.96 8.4.9.7 8.4.9.8 84.99 8.5.1.1 8.5.1.2 8.5.13 85.14 8.5.1.5 8.5.1.6 8.5.1.7 8.5.1.8 8.5.19 8.5.2.1 8.5.2.2 8.5.23 8.5.2.4 8.5.2.5 8.5.2.6 8.5.2.7 8.5.2.8 8.5.2.9 8.5.3.1 8.5.3.2 8.5.3.3 8.5.3.4 8.5.3.5 85.36 8.5.3.7 8.5.3.8 8.5.3.9 8.54.1 8.54.2 8.54.3 8.5.44 8.54.5 8.54.6 8.S.4.7 8.54.8 8.5.4.9 8.S.S.1 8.S.S.2 8.S.S.3 8.S.S.4 8.5.5.5 8.S.S.6 8.5.5.7 8.S.S.8 8.S.S.9 8.5.6.1 85.6.2 8.5.6.3 8.56.4 8.S.6.5 85.6.6 8.5.6.7 8.5.6.8 85.6.9 8.5.7.1 8.5.7.2 8.5.7.3 8.5.7.4 8.5.7.5 8.5.7.6 8.5.7.7 8.5.7.8 8.5.7.9 8.5.8.1 8.5.8.2 8.5.8.3 8.5.8.4 8.5.8.5 8.5.8.6 8.5.8.7 8.5.8.8 8.5.8.9 8.5.9.1 8.5.9.2 8.5.93 8.5.94 8.59.5 8.59.6 8.5.9.7 8.S.9.8 8.5.9.9 8.6.1.1 8.6.1.2 86.13 8.6.1.4 8.6.15 8.6.16 8.6.17 86.1.8 8.6.19 8.6.2.1 8.6.2.2 8.6.23 8.6.2.4 8.6.25 8.6.2.6 8.6.2.7 8.6.2.8 8.6.29 8.6.3.1 8.6.32 8.6.3.3 8.6.3.4 86.35 8.6.36 8.6.3.7 8.638 8.6.3.9 8.6.4.1 8.6.4.2 8.6.43 8.6.4.4 8.6.45 8.6.46 8.64.7 8.6.4.8 8.64.9 8.6.5.1 8.6.5.2 8.6.53 8.6.5.4 8.6.S.S 8.6.5.6 8.6.5.7 8.6.58 8.6.5.9 8.6.6.1 8.6.6.2 8.6.6.3 8.66.4 86. 6.5 86.66 8.6.67 86. 6.8 8.6.6.9 8.6.7.1 8.6.7.2 8.6.7.3 8.6.7.4 8.6.7.5 8.6.7.6 8.6.7.7 8.6.7.8 8.6.7.9 8.6.8.1 8.6.8.2 86.83 8.6.84 8.68.5 8.6.86 86.8.7 86.88 8.6.8.9 86.9.1 86.92 8.6.9.3 86.94 8.6.95 86.9.6 8.6.9.7 86.9.8 86.9.9 8.7.1.1 8.7.1.2 8.7.1.3 8.7.1.4 8.7.1.5 8.7.1.6 8.7.1.7 8.7.1.8 8.7.1.9 8.7.2.1 8.7.2.2 8.7.2.3 8.7.24 8.7.2.5 87.26 8.7.2.7 8.7.2.8 8.7.2.9 8.7.3.1 8.73.2 8.7.3.3 8.73.4 8.73.5 8.7.3.6 8.73.7 8.7.3.8 8.73.9 8.74.1 8.7.4.2 8.7.43 8.7:44 8.7:45 8.74.6 8.7.4.7 8.74.8 8.74.9 8.7.5.1 8.7.5.2 8.7.5.3 8.7.54 8.7.5.5 8.7.5.6 8.7.5.7 8.7.5.8 8.75.9 8.7.6.1 8.76.2 8.76.3 87.6.4 8.76.5 8.76.6 8.7.6.7 8.76.8 8.76.9 8.77.1 8.7.7.2 8.77.3 8.7.74 8.7.7.5 87.76 8.7.7.7 87.7.8 8.7.7.9 8.7.8.1 87.82 8.7.83 8.78.4 8.7.8.5 87.8.6 8.7.8.7 8.7.8.8 8.7.8.9 8.7.91 8.7.9.2 8.79.3 8.7.94 8.79.5 8.7.96 8.7.9.7 8.7-9.8 8.7.9.9 88.1.1 8.8.1.2 8.8.1.3 88.14 8.8.1.5 88.1.6 8.8.1.7 88.1.8 88.19 8.8.2.1 8.8.2.2 8.8.23 8.8.2.4 8.82.5 8.8.2.6 8.82.7 8.8.2.8 8.8.2.9 88.31 8.8.32 88.3.3 8.834 8.8.35 8.83.6 8.8.3.7 8.83.8 8.83.9 88.4.1 8.842 8.84.3 8.8.4.4 8.84.5 8.8.4.6 8.84.7 8.84.8 8.84.9 8.85.1 88.5.2 8.8.5.3 8.8.54 8.8.S.S 8.85.6 8.8.5.7 8.85.8 8.8.59 8.8.6.1 8.8.6.2 8.8.6.3 8.8.6.4 8.8.6.5 8.8.6.6 8.8.6.7 8.8.6.8 8.8.6.9 8.8.7.1 8.87.2 8.8.7.3 88.7.4 8.8.75 8.8.7.6 8.8.7.7 8.8.78 8.8.7.9 88.8.1 88.8.2 88.8.3 8.884 88.8.5 88.8.6 8.8.8.7 88.8.8 8.88.9 88.9.1 8.89.2 8.8.93 88.9.4 8.895 88.9.6 8.89.7 8.8.98 88.9.9 89.1.1 8.9.1.2 89.13 8.9.14 89.15 8.91.6 89.17 89.1.8 8.9.19 8.9.2.1 8.9.2.2 8.9.23 8.92.4 8.9.25 8.92.6 8.9.2.7 89.28 8.92.9 8.9.31 89.32 8.933 8.93.4 8.93.5 8.93.6 8.93.7 89.38 8.93.9 8.94.1 8.94.2 89.43 8.944 8.9.45 8.94.6 8.94.7 89.48 8.94.9 89.5.1 89.5.2 8.9.5.3 89.54 8.9.S.S 89.5.6 8.95.7 8.9.S.8 89.5.9 8.96.1 89.6.2 8.96.3 8.96.4 89.6.5 8.96.6 8.96.7 8.96.8 8.96.9 89.7.1 8.9.7.2 89.7.3 89.7.4 8.9.7.5 89.7.6 8.97.7 8.9.7.8 89.7.9 8.98.1 8.98.2 8.98.3 8.98.4 8.98S 8.98.6 8.98.7 8.98.8 8.98.9 8.99.1 8.9.9.2 8.99.3 8.9.94 8.9.9.5 8.99.6 8.9.9.7 US 8,080,536 B2 63 64 TABLE 1-continued 89.98 8.999 9.1.1.1 9.1.1.2 9.1.1.3 9.1.1.4 9.1.15 91.1.6 9.11.7 9.1.1.8 9.11.9 9.1.2.1 9.1.2.2 9.1.2.3 9.1.2.4 91.25 9.1.2.6 9.1.2.7. 9.1.2.8, 9.1.2.9 91.3.1 9.1.3.2 9.1.3.3 91.3.4. 9.1.3.5 91.3.6 9.1.3.7 91.3.8 91.3.9 9.14.1 9.14.2 9.14.3 9.144. 9.14.5 9.14.6 9.14.7 9.14.8 9.14.9 9.1.5.1 9.15.2 9.15.3 9.15.4 9.15.5 91.56 9.1.5.7. 9.15.8 9.15.9 9.1.6.1 9.1.6.2 9.1.6.3 9.1.6.4 9.1.6.5 9.1.6.6 9.1.6.7. 9.1.6.8, 9.1.6.9 9.17.1 9.17.2 91.73 9.17.4 9.17.5 9.17.6 9.17.7 91.78 9.17.9 91.8.1 9.18.2 9.18.3 91.84 9.18.5 9.18.6 9.18.7 9.18.8 9.18.9 9.19.1 91.9.2 9.19.3 9.19.4 91.9.5 9.1.96 91.9.7 9.19.8 9.19.9 92.1.1 9.2.1.2 9.2.1.3 9.2.1.4 9.2.1.5 9.2.1.6 9.2.1.7 9.2.1.8 9.2.1.9 9.2.2.1 9.2.2.2 9.2.2.3 9.2.2.4 9.2.2.5 9.2.2.6 9.2.2.7. 9.2.2.8 9.2.2.9 92.3.1 9.23.2 92.33 92.34 92.3.5 9.23.6 92.3.7 9.238 9.23.9 9.2.4.1 9.24.2 9.243 9.244. 9.2.45 9.24.6 9.24.7 92.48 92.49 9.2.5.1 9.2.5.2 9.2.5.3 9.2.5.4 9.2.5.5 9.2.5.6 9.2.5.7. 9.25.8 9.2.5.9 92.6.1 9.2.6.2 92.6.3 92.64 9.26.5 92-6.6 9.2.6.7. 92.6.8, 9.26.9 92.7.1 9.2.7.2 9.2.7.3 92.7.4 9.2.7.5 9.2.7.6 9.2.7.7 9.2.78 927.9 92.8.1 9.2.8.2 9.283 9.2.84 9.28.5 9.2.86 9.28.7 92.88 9.2.89 92.9.1 9.2.9.2 92.93 92.94 9.2.95 92.9.6 9.2.9.7 9.2.98 92.99 9.3.1.1 9.3.1.2 9.3.1.3 93.14 93.15 9.3.1.6 93.17 9.3.1.8 9.3.1.9 9.3.2.1 9.3.2.2 9.3.2.3 9.324 93.25 93.26 9.3.2.7 9.3.2.8, 9.329 9.3.3.1 93.3.2 9.333 93.3.4. 9.3.3.5 93.3.6 93.3.7 93.38 93.3.9 9.3.4.1 9.3.4.2 9.343 9.344 93.4.5 9.34.6 93.47 93.48 9.34.9 93.5.1 93.5.2 9.3.5.3 93.54 9.3.S.S 93.5.6 93.5.7 93.5.8 93.5.9 9.3.6.1 9.3.6.2 9.3.6.3 9.3.6.4 93.6.5 9.3.6.6 9.3.6.7. 9.3.6.8, 9.3.6.9 93.7.1 9.3.7.2 9.3.7.3 9.3.7.4 9.3.7.S 93.7.6 9.3.7.7 93.7.8 9.3.7.9 93.8.1 93.8.2 9.3.83 93.8.4 93.85 9.38.6 93.8.7 93.88 93.8.9 9.39.1 93.9.2 93.93 9.394 93.9.5 9.3.96 9.3.9.7 93.98 9.399 94.1.1 9.4.1.2 9.4.13 94.14 9.4.15 9.4.1.6 9.4:1.7 9.4.18 94.19 9.4.2.1 9.4.22 9.4.23 94.2.4 94.25 9.4.2.6 94.2.7. 9.4.2.8, 9.42.9 94.3.1 9.4.3.2 94.3.3 9.434 94-3.5 94.3.6 9.43.7 943.8 9.4.39 9.4.4.1 9.4.4.2 9.4.4.3 9.4.4.4 9.4.4.5 9.4.4.6 9.4.4.7 9.4.4.8 9.4.4.9 9.451 9.45.2 94.5.3 9.454 94.5.5 9.45.6 94.5.7. 94.5.8 9.45.9 94.6.1 9.4.6.2 9.4.6.3 94.64 9.4.6.5 9.4.6.6 9.4.6.7. 94.6.8, 9.4.6.9 9.4.71 94.7.2 9.4.7.3 9.47.4 94.75 9.47.6 94.77 94.78 94.7.9 94.8.1 9.48.2 9.48.3 9.4.84 9.48.5 94.86 9.4.8.7 9.48.8 9.4.89 94.9.1 94.9.2 9.4.9.3 94.9.4 94.9.5 9.4.9.6 94.9.7 9.4.9.8 94.9.9 9.5.1.1 9.5.1.2 95.13 9.5.1.4 9.S. 1.5 9.5.1.6 9.5.1.7 9.5.1.8 9.5.1.9 9.5.2.1 9.5.2.2 9.5.2.3 9.5.2.4 9.5.2.5 9.S.2.6 9.5.2.7. 9.5.2.8, 9.5.2.9 9.5.3.1 9.5.3.2 9.5.3.3 9.5.3.4. 9.5.3.S 9.5.3.6 9.5.3.7 9.5.3.8 9.5.3.9 95.4.1 9.5.4.2 95.4.3 9.5.44. 95.45 9.546 9.5.4.7 9.S.4.8 9.S.4.9 9.S.S.1 9.S.S.2 9.S.S.3 9.S.S.4 9.S.S.S 9.S.S.6 9.S.S.7 9.S.S.8 9.S.S.9 9.S.6.1 9.S.6.2 9.S.6.3 9.S.6.4 9.S.6.5 9.S.6.6 9.S.6.7. 9.S.6.8, 9.S.6.9 9.5.7.1 9.5.7.2 9.5.7.3 9.5.7.4 9.5.7.5 9.5.7.6 9.5.7.7 9.5.7.8 9.5.7.9 9.5.8.1 9.5.8.2 9.5.8.3 9.5.8.4 9.5.8.S 9.5.8.6 9.5.8.7 9.5.8.8 9.5.8.9 9.S.9.1 9.S.9.2 95.9.3 9.5.94 9.5.9.5 95.96 9.5.9.7 95.9.8 95.99 96.1.1 9.6.1.2 9.6.13 9.6.1.4 9.6.1.5 9.6.16 9.6.17 9.6.18, 9.6.19 9.6.2.1 9.6.2.2 9.6.2.3 9.6.2.4 9.6.2.5 9.6.2.6 9.6.2.7. 96.2.8, 9.6.29 96.31 9.6.32 96.3.3 96.34 9.6.35 96.3.6 9.6.3.7. 96.38 96.39 9.6.4.1 96.4.2 9.6.4.3 96.44 96.45 9.6.46 96.47 9.6.4.8 96.49 96.51 9.6.5.2 96.5.3 96.54 96.5.5 96.56 9.6.5.7 96.58 9.6.5.9 9.6.6.1 96.6.2 9.6.6.3 96.6.4 9.6.6.5 96.6.6 96.6.7. 96.68 9.6.6.9 9.6.7.1 9.6.7.2 9.6.7.3 9.6.7.4 9.6.7.5 9.6.7.6 9.6.7.7 9.6.7.8 9.6.7.9 96.8.1 9.6.8.2 9.6.8.3 96.84 96.85 96.8.6 9.68.7 96.8.8 96.89 96.9.1 96.9.2 9.6.9.3 96.94 96.9.5 9.6.9.6 96.9.7 9.69.8 96.9.9 9.7.1.1 9.7.1.2 9.7.1.3 9.7.1.4 9.7.1.S 9.7.1.6 9.7.1.7 9.7.1.8 9.7.1.9 9.7.2.1 9.7.2.2 9.7.2.3 9.7.2.4 9.7.2.S 9.7.2.6 9.7.2-7 9.7.2.8, 9.7.2.9 9.7.3.1 9.7.3.2 9.7.3.3 9.7.34 9.7.3.5 9.7.3.6 9.7.3.7 9.7.3.8 9.7.3.9 9.7.4.1 9.7.4.2 9.7.4.3 97.44 9.7.4.5 9.7.46 9.74.7 9.7.4.8 9.74.9 9.7.5:1. 9.7.5.2 9.7.5.3 97.5.4 9.7.S.S 9.7.5.6 9.7.5.7. 9.7.5.8 9.7.5.9 9.7.6.1 9.7.6.2 9.7.6.3 9.7.6.4 9.7.6.S 9.7.6.6 9.7.6.7 9.7.6.8, 9.7.6.9 9.77.1 9.7.7.2 9.7.7.3 97.7.4 9.77.5 9.7.7.6 97.7.7 9.7.7.8 9.7.7.9 9.7.8.1 9.7.8.2 9.7.8.3 9.7.84 9.7.8.5 9.7.8.6 9.7.8.7 9.7.8.8 9.7.8.9 9.7.9.1 9.7.9.2 9.7.9.3 97.94 9.7.9.S 97.9.6 9.7.9.7 97.9.8 97.9.9 9.8.1.1 9.8.1.2 9.8.1.3 9.8.1.4 9.8.1.5 9.8.1.6 9.8.1.7 9.8.18, 9.8.19 9.8.2.1 9.8.2.2 9.8.2.3 9.8.2.4 9.8.2.5 9.8.2.6 9.8.2.7. 9.8.2.8, 9.8.2.9 98.3.1 98.32 9.8.33 98.3.4. 9.83.5 9.8.3.6 98.37 9.8.38 9.83.9 9.84.1 98.4.2 9.84.3 98.44 98.45 9.84.6 9.8.47 9.84.8 98.4.9 98.5.1 9.85.2 98.5.3 98.54 98.55 98.5.6 9.85.7 9.85.8 98.59 9.8.6.1 9.8.6.2 9.8.6.3 9.8.6.4 9.8.6.5 9.8.6.6 9.8.6.7. 9.8.6.8, 9.8.6.9 98.7.1 9.8.7.2 9.8.7.3 9.8.74 98.7.S 98.76 9.8.7.7 98.7.8 9.87.9 9.88.1 98.82 98.83 98.84 9.88.5 98.8.6 9.88.7 9.8.88 9.88.9 9.89.1 9.8.9.2 98.93 98.94 98.9.5 9.89.6 98.9.7 98.98 98.99 99.1.1 99.1.2 9.91.3 99.14 99.1.5 99.1.6 9.91.7 99.18, 9.9.19 9.9.2.1 99.2.2 9.9.23 9.9.2.4 99.25 9.9.2.6 99.27 9.9.2.8, 9.9.2.9 99.31 9.93.2 99.33 99.34 9.93.5 99.36 9.93.7. 99.38 9.93.9 99.4.1 9.94.2 9.94.3 99.44 9.94.5 9.94.6 9.94.7 9.94.8 9.94.9 9.95.1 99.5.2 9.95.3 99.54 99.5.5 9.95.6 9.95.7 9.95.8 9.95.9 99.6.1 9.96.2 99.6.3 99.64 99.65 99.6.6 99.6.7. 99.6.8, 9.96.9 99.7.1 99.7.2 9.97.3 99.74 99.75 9.97.6 99.77 9.97.8 9.97.9 99.8.1 9.98.2 9.98.3 9.984 99.8.5 9.98.6 9.98.7 99.8.8 9.98.9 9.99.1 99.9.2 9.99.3 99.94 9.99.5 99.96 99.97 99.98 99.99

Another group of preferred compounds are named in Table The compounds are shown without depiction of stereochem 2 and designated by numbers assigned to the variables of 65 istry since the compounds are biologically active as the dias formula I using the following convention: M'.Y/Y".V/Z/W. tereomeric mixture or as a single stereoisomer. M' is a vari US 8,080,536 B2 65 66 able that represents compounds of the formula M-H which Variable V/Z/W: Group V/Z/W2 have a specific hydroxyl group that is phosphorylated with a 1) V phenyl: Z=NHAc; W=hydrogen group P(O)IY CH(V)CH(Z)CH(W) Y to make com 2) V=3,5-dichlorophenyl: Z=NHAc; W=hydrogen 3) V=4-pyridyl; Z=NHAc; W=hydrogen pounds of formula I or M' is a variable that represents phos 4) V-phenyl: Z-hydrogen; W-methyl phonic acids of the formula M-PO, which are transformed to 5) V-3,5-dichlorophenyl: Z-hydrogen; W-methyl compounds of formula I by replacing two oxygens in the 6) V=4-pyridyl: Z-hydrogen; W-methyl PO, group with Y CH(V)CH(Z)CH(W) Y. 7) V-phenyl: Z-hydroxy; W-hydrogen The structures for variable M' are the same as described 8) V-3,5-dichlorophenyl: Z-hydroxy; W=hydrogen above. 9) V=4-pyridyl: Z-hydroxy; W=hydrogen 10 Variable V/Z/W: Group V/Z/W3 1) V-hydrogen; Z=CH2OH; W=hydrogen V H 2) V-hydrogen; Z=CH2OC(O)CH3; W=hydrogen O Y 3) V-hydrogen; Z=CH2OC(O)OCH3; W=hydrogen V / 4) V-methyl; Z=CH2OH; W=hydrogen M-P Z 15 5) V-methyl; Z=CH2OC(O)CH3; W=hydrogen V 6) V-methyl; Z=CH2OC(O)OCH3; W=hydrogen Yi H 7) Z-hydrogen; V and W= CH2-CH(OH)CH2 W. W 8) Z-hydrogen; V and W= CH2-CH(OAc)CH2 9) Z=hydrogen; V and W= CH2-CH(OCO2CH2CH3) Variable YAY CH2 1)Y=NH;Y'-oxygen Preferred compounds are compounds listed in Table 2 2)Y=oxygen;Y'=NH using groups M1 and V/Z/W1. For example, compound 3)Y=NH;Y=NH 1.1.3 represents structure 1 of group M1, i.e. 3TC: structure 4)Y=N CH:Y'-oxygen 1 of the variable Y/Y', i.e. Y=NH and Y'-oxygen; structure 3 25 of group V/Z/W1, i.e. V=4-pyridyl, Z-methyl and 5)Y=oxygen;Y-NCH3 W-hydrogen. The compound 1.1.3. therefore is 3TC with the 6)Y=N CH2CH3;Y'-oxygen P(O)(N(H) CH(4-pyridyl)CH(CH3)CH2O) attached to the 7)Y=N-phenyl: Y'-oxygen primary hydroxyl. 8)Y-Ni-propyl: Y'-oxygen Preferred compounds are also compounds listed in Table 2 9)Y=oxygen;Y'=N CH2CH3 30 using groups M1 and V/Z/W2. Preferred compounds are Variable V/Z/W: Group V/Z/W1 also compounds listed in Table 2 using groups M1 and 1)V-phenyl: Z-methyl; W=hydrogen VIZ/W3. Preferred compounds are also compounds listed in 2) V-3,5-dichlorophenyl: Z-methyl; W-hydrogen Table 2 using groups M2 and V/Z/W1. Preferred compounds 3) V=4-pyridyl: Z-methyl; W-hydrogen are also compounds listed in Table 2 using groups M2 and 4) V-phenyl: Z-methoxy; W-hydrogen 35 V/Z/W 2. Preferred compounds are also compounds listed in 5) V-3,5-dichlorophenyl: Z-methoxy; W-hydrogen Table 2 using groups M2 and V/Z/W 3. Preferred com 6) V=4-pyridyl: Z-methoxy; W-hydrogen pounds are also compounds listed in Table 2 using groups 7) V-phenyl: Z-hydrogen; W- phenyl M3 and V/Z/W1. Preferred compounds are also compounds 8) V-3,5-dichlorophenyl: Z-hydrogen; W-3,5-dichlo listed in Table 2 using groups M3 and V/Z/W 2. Preferred rophenyl compounds are also compounds listed in Table 2 using groups 9) V=4-pyridyl: Z-hydrogen; W=4-pyridyl M3 and V/Z/W 3. TABLE 2

1.1. 1.1.2 1.13 1.1.4 1.15 1.1.6 11.7 1.1.8 11.9 1.2. 1.2.2 1.23 1.2.4 1.25 1.2.6 12.7 12.8 12.9 1.3. 13.2 1.3.3 1.3.4 1.3.5 13.6 1.3.7 1.38 13.9 1.4. 1.4.2 1.4.3 1.4.4 14.5 1.4.6 14.7 1.4.8 14.9 1.5. 15.2 1.5.3 15.4 1.5.5 1.5.6 1.5.7 1.S.8 1.5.9 1.6. 1.6.2 1.63 1.6.4 1.65 16.6 16.7 1.6.8 16.9 1.7. 1.7.2 1.7.3 1.7.4 1.7.5 1.7.6 17.7 1.7.8 1.7.9 1.8. 18.2 18.3 1.8.4 18.5 18.6 18.7 18.8 18.9 19. 1.9.2 19.3 1.94 1.9S 1.96 19.7 1.98 1.99 2.1. 2.1.2 2.13 2.1.4 2.1.5 2.1.6 2.1.7 2.1.8 2.19 2.2. 2.2.2 2.23 2.2.4 2.2.5 22.6 2.2.7 2.2.8 2.2.9 23. 23.2 2.3.3 2.3.4 2.3.5 2.36 2.3.7 23.8 23.9 2.4. 2.4.2 2.4.3 2.4.4 2.45 2.4.6 24.7 2.4.8 2.49 2.S. 2.5.2 2.5.3 2.54 2.5.5 2.56 2.5.7 2.S.8 2.5.9 2.6. 2.6.2 2.6.3 2.6.4 26.5 26.6 2.6.7 2.68 26.9 2.7. 2.7.2 2.7.3 27.4 2.7.5 2.7.6 2.7.7 2.7.8 2.79 2.8. 2.8.2 2.8.3 2.8.4 2.8.5 2.8.6 2.8.7 2.8.8 2.8.9 2.9. 2.9.2 2.93 2.94 2.95 2.96 2.9.7 2.98 2.99 3.1. 3.1.2 3.13 3.1.4 3.1.5 3.1.6 3.1.7 3.1.8 3.19 3.2. 3.2.2 3.23 3.2.4 3.25 3.26 3.2.7 3.28 3.29 3.3. 3.32 3.3.3 3.34 3.35 3.36 3.3.7 3.38 3.3.9 3.4. 3.4.2 3.43 3.4.4 34.5 34.6 34.7 3.48 3.49 3.S. 3.5.2 3.5.3 3.54 3.5.5 3.S.6 3.5.7 3.S.8 3.S.9 3.6. 36.2 3.6.3 36.4 3.6.5 3.6.6 3.6.7 3.68 36.9 3.7. 3.7.2 3.7.3 3.74 3.7.5 3.7.6 3.7.7 3.7.8 3.7.9 38. 38.2 3.83 3.84 3.85 38.6 38.7 3.88 38.9 3.9. 3.9.2 3.9.3 3.94 3.95 3.96 39.7 3.98 3.9.9 4.1. 4.1.2 4.1.3 4.1.4 4.15 4.1.6 4.17 4.1.8 4.1.9 4.2. 4.2.2 4.2.3 4.2.4 4.25 4.2.6 4.2.7 4.2.8 42.9 4.3. 4.3.2 4.3.3 4.3.4 4.3.5 4.3.6 4.3.7 4.38 4.3.9 4.4. 4.4.2 4.4.3 4.4.4 4.45 4.4.6 44.7 4.4.8 4.4.9 4.5. 4.52 4.5.3 45.4 4.S.S 45.6 45.7 45.8 4.59 4.6. 4.6.2 46.3 4.6.4 46.5 4.6.6 46.7 46.8 46.9 4.7. 4.7.2 4.7.3 4.7.4 4.7.5 4.7.6 4.7.7 4.7.8 4.7.9 4.8. 4.8.2 48.3 4.8.4 48.5 4.86 4.8.7 48.8 48.9 4.9. 4.9.2 4.9.3 4.9.4 4.95 4.9.6 4.9.7 49.8 4.99 5.1. 5.1.2 S.1.3 5.14 5.1.5 5.1.6 5.1.7 S.1.8 S.1.9 S.2. S.2.2 S.23 US 8,080,536 B2 67 68 TABLE 2-continued S.2.4 S.2.S. S.2.6 S.2.7 S.2.8 S.2.9 5.3.1 5.3.2 5.33 S.34 53.5 5.3.6 5.3.7 S.38 S.3.9 541 S.4.2 S.43 S.4.4 S.4.S. S.4.6 S.4.7 54.8 5.4.9 5.5. 5.5.2 5.5.3 5.5.4 5.5.5 5.5.6 5.5.7. 5.5.8 5.5.9 5.6. 56.2 S.6.3 S.6.4 S.6.S. S.6.6 S.6.7. S.6.8 S.6.9 S.71 S.7.2 5.7.3 5.7.4 5.75 5.7.6 5.7.7 5.7.8 S. 7.9 5.8.1 5.8.2, 5.8.3 5.8.4 5.8.5 5.8.6 5.8.7 5.8.8 5.8.9 S.9. 5.9.2 5.9.3 5.9.4 5.9S 5.9.6 5.9.7 5.9.8 S.99 6.1. 6.1.2 6.13 6.1.4 6.1.5 6.1.6 6.17 6.1.8 6.1.9 6.2.1 6.2.2 6.23 6.2.4 6.25 6.2.6 6.2.7 6.2.8, 6.2.9 6.3.1, 6.32 6.3.3 6.3.4 6.35 6.36 6.3.7 6.3.8 6.3.9 6.4. 6.4.2 6.4.3 6.4.4 6.45 6.4.6 6.4.7 6.4.8 6.49 6.5. 6.5.2 6.5.3 6.5.4 6.55 6.5.6 6.5.7 6.58 6.5.9 6.6.1 6.6.2. 6.6.3 66.4 6.6.5 6.6.6 6.6.7 6.6.8, 6.6.9 6.7.1 6.7.2 6.7.3 6.7.4 6.75 6.7.6 6.7.7 6.7.8 6.79 6.8. 6.82 6.8.3 6.8.4 6.85 6.8.6 6.8.7 6.88 6.8.9 6.9. 6.9.2 6.9.3 6.9.4 6.9.5 6.9.6 6.9.7 6.9.8 6.9.9 7.1.1 7.1.2 7.1.3 7.1.4 7.1.5 71.6 7.1.7 7.1.8 7.1.9 7.2.1 7.2.2 7.2.3 7.2.4 7.2.S 7.2.6 7.2.7 7.2.8 7.2.9 7.3. 7.32 7.3.3 7.34 7.3.S 7.3.6 7.3.7 7.3.8 7.39 7.4. 7.4.2 7.43 7.44 7.4.S 7.46 74.7 74.8 7.4.9 7.5.1 7.5.2 7.5.3 75.4 7.5.5 7.5.6 7.5.7 7.58 7.5.9 7.6.1 7.6.2 7.6.3 7.6.4 7.6.S 7.6.6 7.6.7 7.6.8 7.6.9 7.7. 7.7.2 7.7.3 7.7.4 7.7.5 7.7.6 7.7.7 7.7.8 7.7.9 7.8. 7.8.2 7.83 78.4 7.8.S 7.8.6 7.8.7 7.8.8 7.8.9 7.9.1 7.9.2 7.9.3 7.9.4 79.5 7.9.6 7.9.7 7.9.8 7.9.9 8.1.1. 8.1.2, 8.13 8.14 8.15 8.1.6 81.7 81.8 8.19 82. 8.22 82.3 8.2.4 8.25 8.2.6 82.7 828 8.2.9 8.3. 8.32 8.3.3 83.4 8.35 8.3.6 83.7 83.8 8.3.9 841 8.4.2 8.43 8.44 84.5 8.4.6 84.7 8.48 8.49 8.5.1. 85.2 8.5.3 85.4 8.5.5 8.5.6 85.7 85.8 8.5.9 86. 8.6.2 8.6.3 86.4 86.5 86.6 86.7 8.6.8, 8.6.9 87. 8.7.2 8.7.3 8.7.4 8.7S 8.7.6 8.7.7 8.7.8 8.7.9 8.8.1. 8.82 8.83 88.4 8.85 8.86 88.7 8.88 88.9 89.1. 89.2 8.9.3 89.4 8.95 8.96 89.7 8.98 8.9.9 9.1. 9.1.2 9.1.3 91.4 915 91.6 91.7 9.18, 9.19 92. 9.2.2 92.3 9.2.4 9.25 9.2.6 92.7 9.2.8, 9.2.9 93.1 93.2 9.33 93.4 93.5 9.36 93.7 93.8 93.9 94.1 94.2 9.4.3 94.4 94.5 9.4.6 94.7 94.8 94.9 9.S. 9.S.2 9.5.3 95.4 9.S.S 9.S.6 95.7 9.S.8 9.S.9 9.6. 9.6.2 96.3 96.4 9.6.5 9.6.6 9.6.7. 96.8 96.9 9.7.1 9.7.2 9.7.3 97.4 9.7.5 9.7.6 9.7.7 9.7.8 9.7.9 9.8.1 98.2 9.8.3 98.4 98.5 9.8.6 98.7 98.8 98.9 99. 9.9.2 9.93 99.4 9.95 99.6 99.7 9.98 9.99

Syntheses of Compounds of Formula I -continued Synthesis of the compounds encompassed by the present invention includes: I). Synthesis of prodrugs; and II). Synthe sis of Substituted 1.3-hydroxyamines and 1,3-diamines. I) Syntheses of Prodrugs: 35 The following procedures on the preparation of prodrugs illustrate the general procedures used to prepare the prodrugs of the invention which apply to all phosphate, phosphonate and phosphoramidate containing drugs. Prodrugs can be 40 introduced at different stages of synthesis of a drug. Most I.1.a. Synthesis of Activated P(V) Intermediates: often they are made at a later stage, because of the general Most preferred synthesis of phosphoramidate nucleotide sensitivity of these groups to various reaction conditions. prodrugs is the reaction of MH hydroxy group with 4-nitro Optically pure prodrugs containing single isomer at phospho phenylphosphoramidate (where L is nitrophenyl) in presence rus center can be made either by separation of the diastere 45 of a strong base such as Sodium hydride or lithium hydride. omers by a combination of column chromatography and/or The activated precursor is prepared by reaction of commer crystallization, or by enantioselective synthesis of chiral acti cially available 4-nitrophenyl phosphorochloridate with a vated phosphoramide intermediates. Substituted 1.3-aminoalcohol or 1,3-diamine. In general, Syn The preparation of prodrugs is further organized into 1) thesis of phosphoroamidate prodrug is achieved by coupling synthesis via activated P(V) intermediates, 2) synthesis via 50 the alcohol with the corresponding activated phosphoroami activated P(III) intermediates, 3) synthesis via phosph(on)ate date precursor for example, Chlorophosphoroamidate diacid, and 4) miscellaneous methods. Some of the general (L-chloro) addition on to 5'-hydroxy of nucleoside is a well synthetic methods described inhere are utilized for phosphate known method for preparation of phosphoroamidates and phosphonate ester synthesis. However, these methods are (Stepanov et al., Tetrahedron Lett., 1989, 30, 5125). The equally applicable in phosphoramidate case also when the 55 activated precursor can be prepared by several well known nitrogen in the prodrug moiety is appropriately protected methods. Chlorophosphoroamidates useful for synthesis of and/or substituted. the prodrugs are prepared from the Substituted-1,3-pro I.1. Synthesis Via Activated P(V) Intermediate: panediamine or 1.3-aminoalcohol (Kirsten, et al., J. Org. Chem., 1997, 62, 6882). Chlorophosphoroamidate may also 60 be made by oxidation of the corresponding P(III) intermedi V V ates (Ozaki, et al. Bull. Chem. Soc. Jpn., 1989, 62. 3869) HY O y which are obtained by reaction of appropriately protected Z -- s Z -- and/or substituted 1,3-aminoalcohol or 1,3-diamine with / \ phosphorus trichloride. Alternatively, the chlorophosphoroa HY Yi 65 midate agent is made by treating Substituted-1,3-aminoalco W W W W hol or 1,3-diamine with phosphorusoxychloride (Welch, et al., J. Org. Chem., 1990, 55,5991). Chlorophosphoroamidate US 8,080,536 B2 69 70 species may also be generated in situ from corresponding to couple by transition metal catalysis (Nagamatsu, et al., phosphites (Jung, et al., Nucleosides and Nucleotides, 1994, Tetrahedron Lett., 1987, 28, 2375). 13, 1597). Phosphoroflouridate intermediate prepared from Reaction of the optically pure diastereomer of phosphora phosphoroamidic acid may also act as a precursor in prepa midate intermediate with the hydroxyl of drug in the presence ration of cyclic prodrugs (Watanabe et al., Tetrahedron lett., of an acid produces the optically pure phosphate prodrug by 1988, 29,5763). direct S2(P) reaction (Nakayama, et al., J. Am. Chem. Soc., Phosphoramidates (LNRR") can also be used as interme 1990, 112,6936). Alternatively, reaction of the optically pure diates in the case of phosphoramidate prodrugs as in the phosphoramidate precursor with a fluoride source, preferably synthesis of phosphate esters where the nitrogen is Substi cesium fluoride or tetrabutylammonium fluoride, produces tuted or in the protected form. Monoalkyl or dialkylphospho 10 the more reactive phosphorofluoridate which reacts with the ramidate (Watanabe, etal, Chem Pharm Bull., 1990,38,562), hydroxyl of the drug to give the optically pure prodrug con triazolophosphoramidate (Yamakage, et al., Tetrahedron, figuration at the phosphorus atom (Ogilvie, et al., J. Am. 1989, 45, 5459) and pyrrolidinophosphoramidate (Na Chem. Soc., 1977, 99, 1277). kayama, et al., J. Am. Chem. Soc., 1990, 112, 6936) are some I.2 Synthesis Via Phosphoramidite Intermediate: of the known intermediates used for the preparation of phos 15 phate esters. Another effective phosphorylating procedure is a metal catalyzed addition of cyclic chlorophosphoroamidate V V adduct of 2-oxazolone. This type of intermediate attains high HY Y selectivity in phosphorylation of primary hydroxy group in M 1. MH Z -- L-P Z -- presence of secondary hydroxyl group (Nagamatsu, et al. V 2. O Tetrahedron Lett., 1987, 28, 2375). These agents are obtained HY Yi by reaction of a chlorophosphoroamidate with the amine or W W W Wi alternatively by formation of the corresponding phosphora V midite followed by oxidation. Y I.b. Synthesis of Chiral Activated Phosphoramidate: 25 On/P Z Phosphorylation of an enantiomerically pure substituted M1 V 1.3-aminoalcohol or 1,3-diamine with for example, a com Yi mercially available phosphorodichloridate R OP(O)Cl. where RO is a leaving group, preferably aryl substituted with W W electron withdrawing groups, such as a nitro or a chloro, 30 produces two diastereomeric intermediates that can be sepa I.2.a. Synthesis of Activated P(III) Intermediates: rated by a combination of column chromatography and/or Phosphorylation of hydroxy and amino groups is achieved crystallization. Such a method may also be utilized in prepar using cyclic phosphorylating agents where the agent is at the ing chiral chlorophosphoroamidate. Chiral phosphoramidate P(III) oxidation state. One preferred phosphorylating agent is intermediates can be obtained by utilization of optically pure 35 a chloro phosphoramidite (L' chloro). Cyclic chlorophos amine as the chiral auxiliary. This type of intermediate are phoramidites are prepared under mild conditions by reaction known to undergo stereospecific Substitution in the phosphate of phosphorus trichloride with substituted 1,3-aminoalcohols formation (Nakayama, et al. J. Am. Chem. Soc., 1990, 112, or 1,3-diamines (Wada, et al. Tetrahedron Lett., 1990, 31, 6936). The relative configuration of the phosphorus atom can 6363). Alternatively phosphoramidites can be used as the be determined by comparison of the 'P NMR spectra. The 40 phosphorylating agent (Beaucage, et al., Tetrahedron, 1993, chemical shift of the equatorial phosphoryloxy moiety (trans 49, 6123). Appropriately substituted phoshoramidites can be isomer) is always more upfield than the one of the axial prepared by reacting cyclic chlorophosphoramidite with isomer (cis-isomer) (Verkade, et al., J. Org. Chem., 1977, 42, N,N-dialkylamine (Perich, et al., Aust. J. Chem., 1990, 43. 1549). 1623. Perich, et al., Synthesis, 1988, 2, 142) or by reaction of 45 commercially available dialkylaminophosphorochloridate with substituted propyl-1,3-aminoalcohols or 1,3-diamines. I.2.b. Synthesis of Chiral Activated P(III) Intermediate: In the cases where unsymmetrical 1.3-aminoalcohols or O-7'N o-7's 1,3-diamines are used, the cyclic phosphoramidites are v7ro N/ NA v7) N/ no H H 50 expected to form a mixture of chiral isomers. When an opti cally active pure amino alcohol or diamine is used a chro cis trans matographically separable mixture of two stable diastere omers with the leaving group (NRR) axial and equatorial on I.1.c. Synthesis of Prodrugs. Using Activated Phosphorami the phosphorous atom is expected (Brown et al., Tet., 1990, dates: 55 46, 4877). Pure diasteromers can usually be obtained by Coupling of activated phosphoramidates with alcohols chromatographic separation. These intermediates may also (MH) is accomplished in the presence of an organic base. For be prepared through chirality induction by an optically active example, Chlorophosphoramidates Synthesized as described amine. in the earlier section react with an alcohol in the presence of I.2.c. Synthesis of Prodrugs Using Activated Phosphoramid a base Such as pyridines or N-methylimidazole. In some cases 60 ites: phosphorylation is enhanced by in situ generation of Appropriately substituted chlorophosphoramidites are iodophosphoramidates from chloro (Stomberg, et al., used to phosphorylate alcohols on nucleosides in the presence Nucleosides & Nucleotides., 1987, 5: 815). Phosphoroflouri of an organic base (e.g., triethylamine, pyridine). Alterna date intermediates have also been used in phosphorylation tively, the phosphoramidite can be obtained by coupling the reactions in the presence of a base such as CsP or n-Bulli to 65 nucleoside with a phosphoramidate in the presence of a cou generate cyclic prodrugs (Watanabe et al., Tetrahedron lett., pling promoter Such as tetrazole or benzimidazolium triflate 1988, 29. 5763). Phosphoramidate intermediates are shown (Hayakawa et al., J. Org. Chem., 1996, 61, 7996). Since US 8,080,536 B2 71 72 condensation of alcohols with chloro phosphoramidites or 3495) or by chirality induction (Taapken, et al., Tetrahedron phosphoramidites is an S2(P) reaction, the product is Lett., 1995, 36, 6659; J. Org. Chem., 1998, 63, 8284). expected to have an inverted configuration. This allows for the 1.4. Miscellaneous Methods: Stereoselective synthesis of cyclic phosphoramidites of pro Phosphorylation of an alcohol is also achieved under Mit drugs. Isomeric mixtures of phosphorylation reactions can 5 Sunobu reaction conditions using the cyclic 1',3'-propanyl also be equilibrated (e.g. thermal equilibration) to a more phosphoramidic acid in the presence of triphenylphosphine thermodynamically stable isomer. and diethylazodicarboxylate (Kimura et al., Bull. Chem. Soc. The resulting phosphoramidites of prodrugs are Subse Jpn., 1979, 52, 1191). Furthermore, phosphate prodrugs can quently oxidized to the corresponding phosphoramidate pro be made by conversion of nucleoside to the dichloridate inter drugs using an oxidant such as molecular oxygen or t-butyl 10 mediate with phosphorylchloride in presence of triethylphos hydroperoxide (Ozaki et al., Tetrahedron. Lett., 1989, 30, phite and quenching with Substituted-1,3-aminoalcohols 5899). Oxidation of optically pure P(III) intermediate is (Farquhar et al., J. Org. Chem., 1983, 26, 1153). expected to stereoselectively provide optically active pro Phosphorylation can also beachieved by making the mixed drugs (Mikolajczyk, et al., J. Org. Chem., 1978, 43, 2132. anhydride of the cyclic diester of phosphoramidic acid and a Cullis, P. M. J. Chem. Soc., Chem. Commun., 1984, 1510, 15 sulfonyl chloride, preferably 8-quinolinesulfonyl chloride, Verfurth, et al., Chem. Ber:, 1991, 129, 1627). and reacting the hydroxyl of the drug in the presence of a base, I.3 Synthesis of Phosphoramidate Prodrugs Via Diacids: preferably methylimidazole (Takaku, et al., J. Org. Chem., 1982, 47, 4937). In addition, starting from a chiral cyclic V phosphoramidic acid, obtained by chiral resolution (Wyn berg, et al., J. Org. Chem., 1985, 50,4508) is also a source for HY chiral phosphoramidate prodrugs. Z III. Synthesis of Substituted 1.3-Hydroxyamines and 1,3- HY Diamines: O, OH O L 25 A large number of synthetic methods are available for the N/ N/ W Wi P -e- P -e- preparation of substituted 1,3-hydroxyamines and 1,3-di /N MN M OH M. L. amines due to the ubiquitous nature of these functionalities in V naturally occurring compounds. Following are some of these Y methods organized into: On/ 30 1. Synthesis of Substituted 1.3-hydroxy amines; 2. Synthesis M12P V Z of substituted 1,3-diamines and 3. synthesis of chiral substi Yi tuted 1,3-hydroxyamines and 1,3-diamines. III.1. Synthesis of Substituted 1.3-Hydroxy Amines: W W A general synthetic procedure for 3-aryl-3-hydroxy-pro 35 pan-1-amine type of prodrug moiety involves aldol type con Prodrugs of formula I are synthesized by reaction of the densation of aryl esters with alkyl nitriles followed by reduc corresponding phosphodichloridate and an 1.3-aminoalcohol tion of resulting substituted benzoylacetonitrile (Shih et al., or 1,3-diamine (Khamnei, et al., J. Med. Chem., 1996, Heterocycles, 1986, 24, 1599). The procedure can also be 39:4109). For example, the reaction of a phosphodichloridate adapted for formation 2-substituted aminopropanols by using with substituted 1,3-aminoalcohols or diamines in the pres 40 Substituted alkylnitrile. In another approach, 3-aryl-3-amino ence of base (such as pyridine, triethylamine, etc) yields propan-1-ol type of prodrug groups are synthesized from aryl compounds of formula I. These conditions can also be applied aldehydes by condensation of malonic acid in presence of in the synthesis of cyclophosphamide analogs where com ammonium acetate followed by reduction of resulting Substi mercially available bis(2-chloroethyl)phosphoramidic tuted b-amino acids. Both these methods enable to introduce dichloride is coupled with corresponding substituted 1,3- 45 wide variety of substitution of aryl group (Shih, et al., Het amino alcohols or 1,3-diamines. erocycles., 1978, 9, 1277). In an alternate approach, -substi Such reactive dichloridate intermediates, can be prepared tuted organolithium compounds of 1-amino-1-aryl ethyl from the corresponding acids and the chlorinating agents e.g. dianion generated from Styrene type of compounds undergo thionyl chloride (Starrett, et al., J. Med Chem., 1994, 1857), addition with carbonyl compounds to give variety of W. W. oxalyl chloride (Stowell, et al., Tetrahedron Lett., 1990, 31: 50 substitution by variation of the carbonyl compounds (Barlu 3261), and phosphorus pentachloride (Quast, et al., Synthesis, enga, et al., J. Org. Chem., 1979, 44, 4798). 1974, 490). Alternatively, these dichlorophosphonates can Large number of synthetic methods are known for the also be generated from disilyl esters (Bhongle, et al., Synth. preparation of racemic or chiral 1.3-diols. These methods Commun., 1987, 17: 1071) and dialkyl esters (Still, et al., may be utilized in the synthesis of corresponding Substituted Tetrahedron Lett., 1983, 24: 4405; Patois, et al., Bull. Soc. 55 1.3-aminoalcohols or 1.3 diamines by converting hydroxy Chim. Fr., 1993, 130: 485). functionality to a leaving group and treating with anhydrous Alternatively, acid coupling reagents including, but not ammonia or required primary or secondary amines (Corey, et limited to, carbodiimides (Alexander, et al., Collect. Czech. al., Tetrahedron Lett., 1989, 30, 5207: Gao, et al., J. Org. Chem. Commun., 1994, 59: 1853; Casara, et al., Bioorg. Med. Chem., 1988, 53,4081). A similar transformation may also be Chem. Lett., 1992, 2: 145; Ohashi, et al., Tetrahedron Lett., 60 achieved directly from alcohols in Mitsunobu type of reaction 1988, 29: 1189; Hoffman, M., Synthesis, 1988, 62), and ben conditions (Hughes, D. L., Org. React., 1992, 42). Zotriazolyloxytris-(dimethylamino)phosphonium salts A variety of synthetic methods are known to prepare the (Campagne, et al., Tetrahedron Lett., 1993, 34: 6743) are also following types of 1,3-diols: a) 1-substituted; b) 2-substi used in the synthesis of compounds of formula I starting from tuted; and c) 1.2- or 1.3-annulated in their racemic or chiral phosphate or phosphonate diacids. 65 form. Substitution of V, W, Z groups of formula I, can be Chiral phosphonoamidate prodrugs can be synthesized by introduced or modified either during synthesis of aminoalco either resolution (Pogatnic, et al., Tetrahedron Lett., 1997,38, hols or after the synthesis of prodrugs. US 8,080,536 B2 73 74 III.1a) 1-Substituted 1,3-Diols: III.2. Synthesis of Substituted 1,3-Diamines: 1,3-Dihydroxy compounds can be synthesized by several Substituted 1,3-diamines are synthesized starting from well known methods in literature. Aryl Grignard additions to variety of substrates. Arylglutaronitriles can be transformed 1-hydroxy propan-3-all give 1-aryl-substituted propan-1,3- to 1-substituted diamines by hydrolysis to amide and Hoff diols. This method will enable conversion of various substi man rearrangement conditions (Bertochio, et al., Bull. Soc. tuted aryl halides to 1-aryl substituted-1,3-propane diols Chim. Fr; 1962, 1809). Whereas, malononitrile substitution (Coppi, et al., J. Org. Chem., 1988, 53,911). Aryl halides can will enable variety of Z substitution by electrophile introduc also be used to synthesize 1-substituted propanediols by Heck tion followed by hydride reduction to corresponding coupling of 1,3-diox-4-ene followed by reduction and diamines. In another approach, cinnamaldehydes react with 10 or Substituted hydrazines to give corresponding hydrolysis (Sakamoto, et al., Tetrahedron Lett., 1992, 33, pyrazolines which upon catalytic hydrogenation result in Sub 6845). Substituted 1,3-diols can be generated enanatioselec stituted 1,3-diamines (Weinhardt, et al., J. Med. Chem., 1985, tive reduction of vinylketone and hydroboration or by kinetic 28,694). High trans-diastereoselectivity of 1.3-substitution is resolution of allylic alcohol. Variety of aromatic aldehydes also attainable by aryl Grignard addition on to pyrazolines can be converted to 1-substituted-1,3-diols by vinyl Grignard 15 followed by reduction (Alexakis, et al., J. Org. Chem., 1992, addition followed by hydroboration. Substituted aromatic 576, 4563). 1-Aryl-1,3-diaminopropanes are also prepared aldehydes are also utilized by lithium-t-butylacetate addition by diborane reduction of 3-amino-3-arylacrylonitriles which followed by ester reduction (Turner, J. Org. Chem., 1990, 55 in turn are made from nitrile Substituted aromatic compounds 4744). In another method, commercially available cinnamyl (Dornow, et al., Chem. Ber, 1949, 82, 254). Reduction of alcohols can be converted to epoxy alcohols under catalytic 1,3-diimines obtained from corresponding 1.3-carbonyl asymmetric epoxidation conditions. These epoxy alcohols compounds are another source of 1,3-diamine prodrug moi are reduced by Red-Al to result in enantiomerically pure ety which allows a wide variety of activating groups V and/or 1,3-diols (Gao, et al., J. Org. Chem., 1980, 53. 4081). Alter Z (Barluenga, et al., J. Org. Chem., 1983, 48, 2255). natively, enantiomerically pure 1,3-diols can be obtained by III.3. Synthesis of Chiral Substituted 1.3-Hydroxyamines and chiral borane reduction of hydroxyethyl aryl ketone deriva 25 1,3-Diamines: tives (Ramachandran, et al., Tetrahedron Lett., 1997,38761). Enantiomerically pure 3-aryl-3-hydroxypropan-1-amines Pyridyl, quinoline, isoquinoline propan-3-ol derivatives can are synthesized by CBS enantioselective catalytic reaction of be oxygenated to 1-substituted-1,3-diol by N-oxide forma B-chloropropiophenone followed by displacement of halo tion followed by rearrangement in acetic anhydride condi group to make secondary or primary amines as required (Co tions (Yamamoto, et al., Tetrahedron, 1981, 37, 1871). Aldol 30 rey, et al., Tetrahedron Lett., 1989, 30, 5207). Chiral 3-aryl condensation is another well described method for synthesis 3-amino propan-1-ol type of prodrug moiety may be obtained of the 1.3-oxygenated functionality (Mukaiyama, Org. by 1,3-dipolar addition of chirally pure olefin and substituted React., 1982, 28, 203). Chral substituted diols can also be nitrone of arylaldehyde followed by reduction of resulting made by enantioselective reduction of carbonyl compounds, isoxazolidine (Koizumi, et al., J. Org. Chem., 1982, 47. by chiral aldol condensation or by enzyme promoted kinetic 35 4005). Chiral induction in 1,3-polar additions to form substi resolution. tuted isoxazolidines is also attained by chiral phosphine pal III.1b) 2-Substituted 1,3-Diols: ladium complexes resulting in enatioselective formation of Various 2-substituted-1,3-diols can be made from com 8-amino alcohol (Hori, et al., J. Org. Chem., 1999, 64, 5017). mercially available 2-(hydroxymethyl)-1,3-propane diol. Alternatively, optically pure 1-aryl Substituted amino alco Pentaerythritol can be converted to triol via decarboxylation 40 hols are obtained by selective ring opening of corresponding of diacid followed by reduction (Werle, et al., Liebigs. Ann. chiral epoxy alcohols with desired amines (Canas et al., Tet Chem., 1986,944) or diol-monocarboxylic acid derivatives rahedron Lett., 1991, 32, 6931). can also be obtained by decarboxylation under known condi Several methods are known for diastereoselective synthe tions (Iwata, et al., Tetrahedron lett. 1987, 28, 3131). Nitrot sis of 1,3-disubstituted aminoalcohols. For example, treat riol is also known to give triol by reductive elimination (La 45 ment of (E)-N-cinnamyltrichloroacetamide with hypochlo tour, et al., Synthesis, 1987, 8, 742). The triol can be rus acid results in trans-dihydrooxazine which is readily derivatised by mono acetylation or carbonate formation by hydrolysed to erythro-3-chloro-O-hydroxy-6-phenylpropan treatment with alkanoyl chloride, or alkylchloroformate amine in high diastereoselectivity (Commercon et al., Tetra (Greene and Wuts, Protective groups in Organic synthesis, hedron Lett., 1990, 31, 3871). Diastereoselective formation John Wiley, New York, 1990). Aryl substitution can be 50 of 1.3-aminoalcohols is also achieved by reductive amination affected by oxidation to aldehyde and aryl Grignard addi of optically pure 3-hydroxy ketones (Haddad et al., Tetrahe tions. Aldehydes can also be converted to Substituted amines dron Lett., 1997, 38,5981). In an alternate approach, 3-ami by reductive amination reaction. noketones are transformed to 1,3-disubstituted aminoalco III.1c) Cyclic-1,3-diols: hols in high stereoselectivity by a selective hydride reduction Compounds of formula I where V—Z or V W are fused 55 (Barluenga et al., J. Org. Chem., 1992, 57, 1219). by four carbons are made from Cyclohexane derivatives. All the above mentioned methods can also be applied to Commercially available cis,cis-1,3,5-cyclohexane triol can prepare corresponding V—Z or V-W annulated chiral ami be used as is or modified as described in case of 2-substituted noalcohols. Furthermore. Such optically pure amino alcohols propan-1,3-diols to give various analogues. These modifica are also a source to obtain optically pure diamines by the tions can either be made before or after ester formation. 60 procedures described earlier in the section. Various 1.3-cyclohexane diols can be made by Diels-Alder Formulations methodology using pyrone as diene (Posner, et al., Tetrahe Compounds of the invention are administered orally in a dron Lett., 1991, 32, 5295). Cyclohexyl diol derivatives are total daily dose of about 0.1 mg/kg/dose to about 100 mg/kg/ also made by nitrile oxide-olefin additions (Curran, et al., J. dose, preferably from about 0.3 mg/kg/dose to about 30 Am. Chem. Soc., 1985, 107, 6023). Alternatively, cyclohexyl 65 mg/kg/dose. The most preferred dose range is from 0.5 to 10 precursors are also made from commercially available quinic mg/kg (approximately 1 to 20 nmoles/kg/dose). The use of acid (Rao, et al., Tetrahedron Lett., 1991, 32, 547.) time-release preparations to control the rate of release of the US 8,080,536 B2 75 76 active ingredient may be preferred. The dose may be admin oil, sesame oil or coconut oil, or in a mineral oil such as liquid istered in as many divided doses as is convenient. When other paraffin. The oral Suspensions may contain a thickening methods are used (e.g. intravenous administration), com agent, such as beeswax, hard paraffin or cetyl alcohol. Sweet pounds are administered to the affected tissue at a rate from ening agents, such as those set forth above, and flavoring 0.3 to 300 nmol/kg/min, preferably from 3 to 100 nmoles/kg/ agents may be added to provide a palatable oral preparation. min. Such rates are easily maintained when these compounds These compositions may be preserved by the addition of an are intravenously administered as discussed below. antioxidant Such as ascorbic acid. For the purposes of this invention, the compounds may be Dispersible powders and granules of the invention suitable administered by a variety of means including orally, parenter for preparation of an aqueous suspension by the addition of ally, by inhalation spray, topically, or rectally informulations 10 water provide the active ingredient in admixture with a dis containing pharmaceutically acceptable carriers, adjuvants persing or wetting agent, a Suspending agent, and one or more and vehicles. The term parenteral as used here includes sub preservatives. Suitable dispersing or wetting agents and Sus cutaneous, intravenous, intramuscular, and intraarterial injec pending agents are exemplified by those disclosed above. tions with a variety of infusion techniques. Intraarterial and Additional excipients, for example Sweetening, flavoring and intravenous injection as used herein includes administration 15 coloring agents, may also be present. through catheters. Oral administration is generally preferred. The pharmaceutical compositions of the invention may Pharmaceutical compositions containing the active ingre also be in the form of oil-in-water emulsions. The oily phase dient may be in any form suitable for the intended method of may be a vegetable oil. Such as olive oil or arachis oil, a administration. When used for oral use for example, tablets, mineral oil. Such as liquid paraffin, or a mixture of these. troches, lozenges, aqueous or oil Suspensions, dispersible Suitable emulsifying agents include naturally-occurring powders or granules, emulsions, hard or soft capsules, syrups gums, such as gum acacia and gum tragacanth, naturally or elixirs may be prepared. Compositions intended for oral occurring phosphatides, such as soybean lecithin, esters or use may be prepared according to any method known to the partial esters derived from fatty acids and hexitol anhydrides, art for the manufacture of pharmaceutical compositions and Such as Sorbitan monooleate, and condensation products of Such compositions may contain one or more agents including 25 these partial esters with ethylene oxide. Such as polyoxyeth Sweetening agents, flavoring agents, coloring agents and pre ylene Sorbitan monooleate. The emulsion may also contain serving agents, in order to provide a palatable preparation. Sweetening and flavoring agents. Tablets containing the active ingredient in admixture with Syrups and elixirs may be formulated with Sweetening non-toxic pharmaceutically acceptable excipient which are agents, such as glycerol, Sorbitol or Sucrose. Such formula suitable for manufacture of tablets are acceptable. These 30 tions may also contain a demulcent, a preservative, a flavoring excipients may be, for example, inert diluents, such as cal or a coloring agent. cium or sodium carbonate, lactose, calcium or sodium phos The pharmaceutical compositions of the invention may be phate; granulating and disintegrating agents, such as maize in the form of a sterile injectable preparation, such as a sterile starch, oralginic acid; binding agents, such as starch, gelatin injectable aqueous or oleaginous Suspension. This Suspen or acacia, and lubricating agents, such as magnesium Stearate, 35 sion may be formulated according to the known art using Stearic acid or talc. Tablets may be uncoated or may be coated those Suitable dispersing or wetting agents and Suspending by known techniques including microencapsulation to delay agents which have been mentioned above. The sterile inject disintegration and adsorption in the gastrointestinal tract and able preparation may also be a sterile injectable solution or thereby provide a Sustained action over a longer period. For Suspension in a non-toxic parenterally acceptable diluent or example, a time delay material Such as glyceryl monostearate 40 Solvent, such as a solution in 1,3-butane-diol or prepared as a or glyceryl distearate alone or with a wax may be employed. lyophilized powder. Among the acceptable vehicles and Sol Formulations for oral use may be also presented as hard vents that may be employed are water, Ringer's Solution and gelatin capsules where the active ingredient is mixed with an isotonic sodium chloride solution. In addition, sterile fixed inert Solid diluent, for example calcium phosphate or kaolin, oils may conventionally be employed as a solventor Suspend or as Soft gelatin capsules wherein the active ingredient is 45 ing medium. For this purpose any bland fixed oil may be mixed with water oran oil medium, Such as peanut oil, liquid employed including synthetic mono- or diglycerides. In addi paraffin or olive oil. tion, fatty acids Such as oleic acid may likewise be used in the Aqueous Suspensions of the invention contain the active preparation of injectables. materials in admixture with excipients suitable for the manu The amount of active ingredient that may be combined facture of aqueous Suspensions. Such excipients include a 50 with the carrier material to produce a single dosage form will Suspending agent, such as Sodium carboxymethylcellulose, vary depending upon the host treated and the particular mode methylcellulose, hydroxypropyl methylcellulose, sodium of administration. For example, a time-release formulation alginate, polyvinylpyrrolidone, gum tragacanthandgum aca intended for oral administration to humans may contain 20 to cia, and dispersing or wetting agents such as a naturally 2000 umol (approximately 10 to 1000 mg) of active material occurring phosphatide (e.g., lecithin), a condensation product 55 compounded with an appropriate and convenient amount of of an alkylene oxide with a fatty acid (e.g., polyoxyethylene carrier material which may vary from about 5 to about 95% of Stearate), a condensation product of ethylene oxide with a the total compositions. It is preferred that the pharmaceutical long chain aliphatic alcohol (e.g., heptadecaethyleneoxycet composition be prepared which provides easily measurable anol), a condensation product of ethylene oxide with a partial amounts for administration. For example, an aqueous solu ester derived from a fatty acid and a hexitol anhydride (e.g., 60 tion intended for intravenous infusion should contain from polyoxyethylene Sorbitan monooleate). The aqueous Suspen about 0.05 to about 50 umol (approximately 0.025 to 25 mg) sion may also contain one or more preservatives such as ethyl of the active ingredient per milliliter of solution in order that or n-propyl p-hydroxy-benzoate, one or more coloring infusion of a suitable volume at a rate of about 30 mL/hr can agents, one or more flavoring agents and one or more Sweet OCCU. ening agents. Such as Sucrose or saccharin. 65 As noted above, formulations of the present invention Suit Oil Suspensions may be formulated by Suspending the able for oral administration may be presented as discrete units active ingredient in a vegetable oil. Such as arachis oil, olive Such as capsules, cachets or tablets each containing a prede US 8,080,536 B2 77 78 termined amount of the active ingredient, as a powder or examples which illustrate some of the processes by which granules; as a solution or a Suspension in an aqueous or these compounds are prepared. These examples should not non-aqueous liquid; or as an oil-in-water liquid emulsion or a however be construed as specifically limiting the invention water-in-oil liquid emulsion. The active ingredient may also and variations of the compounds, now known or later devel be administered as a bolus, electuary or paste. A tablet may be made by compression or molding, option oped, are considered to fall within the scope of the present ally with one or more accessory ingredients. Compressed invention as hereinafter claimed. tablets may be prepared by compressingina Suitable machine Compounds of formula I are prepared using procedures the active ingredient in a free flowing form Such as a powder detailed in the following examples. or granules, optionally mixed with a binder (e.g., povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert 10 Example 1 diluent, preservative, disintegrant (e.g., sodium starch glyco late, cross-linked povidone, cross-linked sodium carboxym General Procedure for Phosphoramidate Prodrugs of ethyl cellulose) Surface active or dispersing agent. Molded Phosphonic Acid Containing Drugs tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert 15 A suspension of 5-Isobutyl-2-methyl-4-(2-(5-phosphono) liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled furanyl)thiazole (200 mg, 0.6 mmol) in 2.5 mL of thionyl release of the active ingredient therein using, for example, chloride was heated at reflux temperature for 4h. The reaction hydroxypropyl methylcellulose in varying proportions to mixture was cooled and evaporated to dryness followed by provide the desired release profile. Tablets may optionally be azeotroping with toluene. To the solution of the resulting provided with an enteric coating, to provide release in parts of residue in 4 ml of methylene chloride was added a solution of the gut other than the stomach. This is particularly advanta amino alcohol (82 mg 0.54 mmol) and pyridine (0.5 ml, 6 geous with the compounds of formula I when such com mmol) in 1 mL of methylene chloride. pounds are Susceptible to acid hydrolysis. After stirring at 25°C. for 4h the reaction was evaporated Formulations suitable for topical administration in the 25 and coevaporated with toluene. The crude product was chro mouth include lozenges comprising the active ingredient in a flavored base, usually Sucrose and acacia or tragacanth; pas matographed by eluting with 10% methanol-dichlo tilles comprising the active ingredient in an inert base such as romethane to give 52 mg (20.8%) of a less polarisomer and 48 gelatin and glycerin, or Sucrose and acacia; and mouthwashes mg (19.2%) of a more polar isomer. comprising the active ingredient in a Suitable liquid carrier. The following compounds were prepared in this manner: Formulations for rectal administration may be presented as 30 1.1: 5-Isobutyl-2-methyl-4-2-5-(1-phenyl-1,3-propyl) a Suppository with a Suitable base comprising for example phosphorimidol-furanylthiazole, less polar isomer. cocoa butter or a salicylate. Rf=0.76 in 10% MeOHACH2Cl2. Anal. Cald. for Formulations suitable for vaginal administration may be C21H25N2O3PS+0.25H2O+0.1 HC1: C, 59.40; H, 6.08; presented as pessaries, tampons, creams, gels, pastes, foams N, 6.60. Found: C, 59.42; H, 5.72: N, 6.44. or spray formulations containing in addition to the active 35 1.2: 5-Isobutyl-2-methyl-4-2-5-(1-phenyl-1,3-propyl) ingredient such carriers as are known in the art to be appro priate. phosphorimidol-furanylthiazole, more polar isomer. Formulations suitable for parenteral administration Rf=0.7 in MeOHACH2C12. Anal. Cald. for include aqueous and non-aqueous isotonic sterile injection C21H25N2O3PS+0.25H2O: C, 59.91; H, 6.1: N, 6.65. Solutions which may contain antioxidants, buffers, bacteri 40 Found: C, 60.17; H, 5.81; N, 6.52. ostats and solutes which render the formulation isotonic with 1.3: 2-Amino-5-isobutyl-4-2-5-(1-phenyl-1,3-propyl) the blood of the intended recipient; and aqueous and non phosphoramidofuranylthiazole. Rf 0.53 (silica, 9:1 aqueous sterile Suspensions which may include Suspending CH2Cl2/MeOH). Anal. cald. for C20 H24 N3 O3 P agents and thickening agents. The formulations may be pre S+0.2CH2C12+0.25H2O: C, 55.27; H, 5.72: N, 9.57. sented in unit-dose or multi-dose sealed containers, for 45 Found: C, 55.03; H, 5.42: N, 9.37. example, ampoules and vials, and may be stored in a freeze 1.4: 2-Amino-5-isobutyl-4-2-5-(1-phenyl-1,3-propyl) dried (lyophilized) condition requiring only the addition of phosphoramidofuranylthiazole, less polar isomer. the Sterile liquid carrier, for example water for injections, Rf-0.57 (silica, 9:1 CH2Cl2/MeOH). Anal. cald. for C20 immediately prior to use. Injection solutions and Suspensions H24 N3 O3 PS+0.15 CH2C12: C, 56.26; H, 5.69; N, 9.77. may be prepared from Sterile powders, granules and tablets of 50 Found: C, 56.36; H, 5.46; N, 9.59. the kind previously described. 1.5: 2-Amino-5-methylthio-4-2-5-(1-phenyl-1,3-propyl) Preferred unit dosage formulations are those containing a phosphoramido-)furanylthiazole, less polar isomer. daily dose or unit, daily Sub-dose, or an appropriate fraction Rf-0.59 (silica, 9:1 CH2Cl2/MeOH). Anal. cald. for C17 thereof, of a drug. H18 N3 O3 PS2+0.4HC1: C, 48.38: H, 4.39; N, 9.96. It will be understood, however, that the specific dose level 55 Found: C, 48.47; H, 4.21: N, 9.96. for any particular patient will depend on a variety of factors 1.6: 2-Amino-5-methylthio-4-2-5-(1-phenyl-1,3-propyl) including the activity of the specific compound employed; the phosphoramidofuranylthiazole, more polar isomer. age, body weight, general health, sex and diet of the indi Rf 0.56 (silica, 9:1 CH2Cl2/MeOH). Anal. cald. for C17 vidual being treated; the time and route of administration; the H18 N3 O3 PS2: C, 50.11; H, 4.45; N, 10.31. Found: C, rate of excretion; other drugs which have previously been 60 49.84; H, 4.19; N, 10.13. administered; and the severity of the particular disease under 1.7: 2-Amino-5-methylthio-4-2-5-(N-methyl-1-phenyl-1, going therapy, as is well understood by those skilled in the art. 3-propyl)phos-phoramidofuranylthiazole. Rf-0.56 (silica, 9:1 CH2Cl2/MeOH). Anal. cald. for C18H20 N3 EXAMPLES O3 PS2+0.25HC1 C: 50.21; H, 4.74: N, 9.76. Found: C, 65 50.31; H, 4.46; N, 9.79. The prodrug compounds of this invention, their intermedi 1.8: 2-Amino-5-isobutyl-4-2-5-(1-phenyl-1,3-propyl)-N- ates, and their preparation can be understood further by the acetylphosph-oramidofuranylthiazole. Rf 0.62 (silica, US 8,080,536 B2 79 80 9:1 CH2Cl2/MeOH). Anal. cald. for C22H26 N3 O4 P 2.2 3'-Azido-3'-deoxy-5'-O-(4-phenyl-2-oxo-1,3,2-oxaza S+1.25H2O: C, 54.82; H, 5.96: N, 8.72. Found: C, 55.09: phosphorin-2-y-1)-thymidine. mp 85-105. Rf-0.5 (silica H, 5.99; N, 8.39. gel, 1/9 methanol/dichloromethane). Anal. cald. for 1.9: 2-Amino-5-isobutyl-4-2-5-(1-2,4-dichlorophenyl CHNOP: C, 49.35; H, 5.01; N, 18.17. Found: C, propan-1,3-yl)phosphoramidofuranylthiazole. mp 235 49.32; H, 5.17; N, 17.89. 237 C (decomp). Rf 0.35 (silica, 3:2 EtOAc/CH2Cl2). Anal. cald. for C20H22C12 N3O3 PS: C, 49.39; H, 4.56: Example 3 N, 8.64. Found: C, 49.04; H, 4.51; N, 8.37. 1.10: 188-1899-2-1-phenyl-1-aza-3-oxa-2-phosphorina Step A nyl-2-methyloxyethyladenine. mp 188-189 C. Rf=0.25 10 (silica, 9:1 CHCl2/MeOH). Anal. cald. for C17H21 N6 O3 Same as step A of example 2. P+0.5H2O: C, 51.38; H, 5.58: N, 21.15. Found: C, 51.05; H, 5.28; N, 20.77. Step B 1.11: 2-Amino-5-isobutyl-4-2-5-(1-(3-chlorophenyl)-pro pan-1,3-yl)phosphoradia-midofuranylthiazole. Light 15 Lithium hydride (15 mg, 1.52 mmol) was added to a solu yellow solid; Rf-0.45 (Silica, 5:95 Methanol/dichlo tion of 9-3-D-arabinofuranosyl-adenine (100 mg, 0.37 romethane); H NMR (200 MHz, DMSO-d6) & 7.35 (m, mmol) and fast eluting (4-nitrophenoxy)-4-phenyl-1,3,2-ox 4H); 7.1 (m. 1H), 6.6 (m, 1H);5.55 (m. 1H), 2.8 (d. 2H), 0.9 aZaphosphorinan-2-one (250 mg. 0.74 mmol) in dimethylfor (d. 6H). mamide at room temperature. After 6 hours the yellow het erogeneous mixture was neutralized with acetic acid. The Example 2 clear yellow solution was concentrated and the residue was purified by column chromatography (silica gel, methanol/ Step A dichloromethane). The isolated product was further purified 25 by preparative TLC and recrystallized from ethanol to give A solution of 3-amino-3-phenyl-1-propanol (1 g, 6.6 the prodrug: 18 mg (11% yield); mmol) and triethylamine (3 ml, 21.8 mmol) in tetrahydrofu The following compound is prepared in this manner: ran (60 ml) was added dropwise over 20 minutes into a 3.15'-O-(2-oxo-4-Phenyl-1,3,2-oxazaphosphorin-2-yl)-9-B- solution of 4-nitrophenyl phosphorodichloridate in tetrahy D-arabinofuranosyl-adenine. mp-250. Rf-0.25 (silica gel, drofuran at 0° C. A white precipitate quickly forms. The 30 2/8 methanol/dichloromethane). Anal. cald. for yellow heterogeneous mixture was stirred at 0°C. for 1 hour CHNOP: C, 49.35; H, 5.01: N, 18.17. Found: C, then warmed slowly tort. After stirring at rt for 60 hours, the 49.22; H, 4.89; N, 18.03. precipitate was dissolved with water (40 ml). The clear yel The following compounds are made in a similar manner low solution was concentrated to ca 40 ml. The resulting with the exception that the reactions were neutralized with 1N mixture was diluted with a saturated aqueous Solution of 35 Solution of hydrochloric acid during work up. sodium bicarbonate and extracted twice with ethyl acetate. 3.2 9-(2-(4-Phenyl-2-oxo-1,3,2-oxazaphosphorinan-2- The combined organic extracts were washed with a saturated oxy)-ethoxy)methyl-guanin: mp 151-165; Rf-0.2 (silica aqueous solution of sodium bicarbonate and brine. The aque ous washes were back extracted and the organic extracts gel, 2/8 methanol/dichloromethane); Anal. cald. for combined. The combined organic extracts were dried over 40 CHNOP+2H2O: C, 44.74; H, 5.52: N, 18.41. Found: Sodium sulfate, concentrated and purified by column chroma C, 44.45; H, 5.40; N, 18.14. tography (silica gel, hexanes/ethylacetate) to give fasteluting 3.3 2'-Deoxy-5-fluoro-5'-O-(4-phenyl-2-oxo-1,3,2-oxaza trans isomer (995 mg, 45%) and slow eluting cis isomer (1 g, phosphorin-2-yl)-uridine. mp 188-195 Dec. Rf 0.25 45%) and); (silica gel, 1/9 methanol/dichloromethane). MS cald. for 45 CHFNOP+Na": 464. Found: 464; MS cald. for Step B CHFN.O.P H: 440. Found: 440. A mixture of 2',3'-dideoxy-adenosine (50 mg 0.21 mmol), Example 4 cesium fluoride (323 mg, 2.1 mmol) and fast eluting (4-Ni trophenoxy)-4-phenyl-1,3,2-oxazaphosphorinan-2-one (142 50 General Procedure for Formation of Nucleotide mg, 0.4 mmol) in tent-butanol was heated at 80°C. After 48 Prodrugs by P(III) Intermediates hours, more (4-Nitrophenoxy)-4-phenyl-1,3,2-oxazaphos phorinan-2-one (70 mg) was added and stirring was contin To a solution of 1(aryl)-1-(1-alkyl or acylamino) propanol ued at 80°C. After 5 days, the cooled yellow heterogeneous (1 mmol) in dichloromethane (10 mL) is added phosphorus mixture was diluted with 50/50 methanol/dichloromethane, 55 trichloride at 0°C. The reaction is warmed to room tempera filtered through a pad of silica and rinsed with 50/50 metha ture and allowed to stir for 3 h. Reaction mixture is concen nol/dichloromethane. The combined filtrates were concen trated, azeotroped with toluene (2x10 mL) and dried. Crude trated and the residue was purified by column chromatogra chlorophosphoramidite is used in the next step without fur phy (silica gel, methanol/dichloromethane) to give the ther purification. prodrug which was re crystallized from dichloromethane and 60 To a solution of nucleoside (1 mmol) in DMF (10 mL) is hexanes: 42 mg (47% yield). added diisopropylethylamine (2 mmol) at -40°C. To this The following compounds are prepared in this manner: mixture is added crude cyclic chlorophoramidite (1 mmole) 2.1 2',3'-Dideoxy-5'-O-(2-oxo-4-phenyl-1,3,2-oxazaphos in 2 mL of DMF. The mixture is warmed to room temperature phorin-2-yl)-adenosine. mp 119-122. Rf-0.25 (silica gel, and stirred for 2 h. The reaction is cooled back to -40°C. and 1/9 methanol/dichloromethane). Anal. Cald. for 65 5-6Mt-butylhydroperoxide in decane (2 mmol) is added and CHNOP+0.9 mol dichloromethane: C, 47.16; H, left at room temperature. After overnight stirring, the reaction 4.93: N, 16.58. Found: C, 47.03; H, 4.98: N, 16.61. is concentrated and crude mixture is chromatographed. US 8,080,536 B2 81 82 Example 5 Example 7

General Procedure for Preparation of General Procedure for Preparation of 3-Aryl-3-Aminoprolpanols from Aryl Aldehydes 3-Aryl-Propylenediamines from Cinnamyl Aldehydes (Shih, et al., Heterocycles, 1978, 9, 1277) (Weinhardt, et al., J. Med Chem., 1985, 28, 694) Step A: Step A: A mixture of an appropriate aryl aldehyde (10 mmol), To a solution of cinnamaldehyde (10 mmol) in 100 mL of malonic acid (10 mmol), and ammonium acetate (10 mmol) 10 EtOH is added substituted (10 mmol). The reaction in 50 mL of 95% ethanol is heated in a water bath at 80° C. is stirred at room temperature for 45 min. Solvents are Carbon dioxide begins to evolve at 55° C. and the reaction is removed on a rotatory evaporator and saturated NaHCO, was complete in 5-7 h. The white solid is collected by suction and added to the residue. The crude product is extracted into ether. recrystallized from aqueous ethanol to obtain the 3-aryl-3- The organic layer is washed, dried and evaporated. Crude aminopropionic acid. 15 products from chromatography results in a pure pyrazoline Step B: adduct. Step B: To a suspension of lithium aluminum hydride (50 mmol) in A solution of 3-phenylpyrazoline (10 mmol) in 50 mL of anhydrous tetrahydrofuran (40 mL) is added a 3-aryl-3-ami AcOH and 10 mL of 10% HCl is hydrogenated at atmospheric nopropionic acid (20 mmol) with cooling and stirring. The pressure over 500mg of 5% Pt/C. The reaction is stirred under mixture is refluxed for 3 h, allowed to stand overnight, and hydrogen for 6 h. The catalyst is removed by filtration and the treated with moist ether and then with water to decompose the filtrate is concentrated. The crude product is purified by col excess of lithium aluminum hydride. The organic layer is umn to give pure 3-Aryl-propylenediamines derivative. decanted, and thematerial left in the flaskis extracted withhot ethylacetate (3x50 mL). The organic solutions are combined 25 Example 8 and rotary evaporated. The residue is vacuum distilled or recrystallized to obtain 3-aryl-3-aminopropanol. General Procedure for 1,3-Amino Alcohols for 2-Substituted Prodrugs (V. Wand W=H and Example 6 Z= CHROH) 30 Step A: General Procedure for Preparation of To a solution of commercially available 2-(hydroxym 3-Aryl-3-Hydroxypropylamines from Epoxy ethyl)-1,3-propane diol (1 mmol) in pyridine (5 mL) is added Cinnamyl Alcohols methanesulfonyl chloride (1 mmol) and stirred at room tem 35 perature until the reaction is complete. The mixture is con (Gao, et al., J. Org. Chem., 1988, 53, 4084) centrated, extracted and product is purified by column chro matography. Step A: Step B: To a solution of commercial (-)-(2S,3S)-2,3-epoxycin A solution of 2-(methylsulfonyloxymethylene)-1,3-pro namyl alcohol (10.0 mmol) in dimethoxyethane (50 mL) is 40 pane diol (1 mmol) and ammonium hydroxide (10 mL, 30% added a 3.4 M solution of sodium bis(2-methoxyethoxy) in water) in THF (10 mL) is heated at 100° C. in a sealed aluminum hydride (Red-Al) in toluene (10.5 mmol) dropwise reactor. After cooling, the mixture is concentrated, extracted under nitrogen at 0°C. After stirring at room temperature for and product is purified by column chromatography to give 3 h, the solution is diluted with ether and quenched with 5% 2-substituted-1,3-amino alcohol. Appropriate amines may be HCl solution. After further stirring at room temperature for 30 45 used in step B to vary —NR substitution. 2-substituted-1,3- min, the white precipitate formed is removed by filtration and diamines may also be synthesized using Step A and B from boiled with ethyl acetate and filtered again. The combined 2-(hydroxymethyl)-1,3-propane diol using appropriate sto organic extracts are dried with magnesium sulfate and con ichiometry of reagents. centrated to give (R)-3-phenyl-1,3-dihydroxypropane. Step C: Step B: 50 Prodrugs of 2-substituted-1,3-amino alcohols or 2-substi To a solution of (R)-3-phenyl-1,3-dihydroxypropane (17.8 tuted-1,3-diamines are synthesized by following coupling mmol) and triethylamine (25.6 mmol) in ether (90 mL) is procedures as described in example 1 or example 2 (Steps. A added dropwise MsCl (18.7 mmol) under nitrogen at -10°C. and B) or example 3 (step B) or example 4 depending on the After stirring at -10 to 0°C. for 3 h, the mixture is poured into parent compound. ice water (30 mL), organic layer was washed with 20% 55 HSO, saturated aqueous NaHCO, and brine, and dried Example 9 over magnesium sulfate. The crude product is purified by chromatography on a silica gel column. General Procedure for Cyclic 1.3-Amino Alcohols (Together V and W are Connected) Step C: 60 A solution of (R)-3-phenyl-3-(hydroxy)-propyl methane Step A: sulfonate (3 mmol) and methylamine (10 mL, 40% in water) Commercially available cis,cis-1,3,5-cyclohexane triol is in THF (10 mL) is heated at 65° C. for 3 h. After cooling, the converted to a mono mesylate as described in example 8, step solution is diluted with ether, washed with saturated aqueous A. sodium bicarbonate and brine, and dried with anhydrous 65 Step B: potassium carbonate. Concentration of extract provides (R)- cis,cis-1-(methylsulfonyloxy)cyclohexane-3,5-diol is 3-phenyl-3-(hydroxy)-propyl amine. transformed to corresponding amino alcohol as described in US 8,080,536 B2 83 84 example 8, Step B. Appropriate amines may be used in step B through filtration to give 2-amino-5-isobutyl-4-2-(5-dieth to vary —NR substitution. 1-Hydroxy-3,5-diamino cyclo ylphosphono) furanylthiazole. hexane may also be synthesized using Step A and B from cis, Step G: cis-1,3,5-cyclohexane triol using appropriate Stoichiometry A solution of 2-amino-5-isobutyl-4-2-(5-dieth of reagents. ylphosphono)-furanylthiazole (1 mmole) in methylene chlo Step C: ride was treated with bromotrimethylsilane (10 mmole) at 25° Prodrugs of cyclohexyl-1,3-amino alcohols or 1,3-di C. for 8 h. The reaction mixture was evaporated to dryness amines are synthesized by following procedures as described and the residue was suspended in water. The resulting Solid in example 1 or example 2 (steps A and B) or example 3 (step was collected through filtration to give 2-amino-5-isobutyl B) or example 4 depending on the parent compound. 10 4-2-(5-phosphono)furanylthiazole as an off-white solid. 10.1 2-amino-5-isobutyl-4-2'-(5'-phosphono)furanylthiaz Example 10 ole mp>250° C. Anal. calcd. for CHNOPS+ 1.25HBr: C, 32.75: H, 4.06; N, 6.94. Found: C, 32.39: H, Preparation of 2-substituted 4.33: N, 7.18. 4-2'-(5'-phosphono) furanylthiazoles 15 The following compounds were prepared according to similar procedure: Step A: 10.2 2-Methyl-5-isobutyl-4-2'-(5'-phosphono) furanylthia A solution of furan (1.3 mmole) in toluene was treated with Zole. Anal. calcd. for CHNOPS+HBr--0.1CH.Cl: C, 4-methylpentanoic acid (1 mmole), trifluoroacetic anhydride 37.20; H, 4.44; N, 3.58. Found: C, 37.24; H, 4.56: N, 3.30. (1.2 mmole) and borontrifluoride etherate (0.1 mmole) at 56° 10.3 2-Amino-5-methylthio-4-2'-(5-phosphono) furanyl C. for 3.5h. The cooled reaction mixture was quenched with thiazole. mp 181-184°C. Anal. calcd. for CHNOPS2+ aqueous sodium bicarbonate (1.9 mmole), filtered through a 0.4HO: C, 32.08; H, 3.30; N, 9.35. celitepad. Extraction, evaporation and distillation gave 2-(4- Found: C, 32.09: H, 3.31: N, 9.15. methyl-1-oxo)pentylfuran as a brown oil (bp 65-77°C., 0.1 mmHg). 25 Example 11 Step B: A solution of 2-(4-methyl-1-oxo)pentylfuran (1 mmole) General Procedure for the Synthesis of in benzene was treated with ethylene glycol (2.1 mmole) and Cyclophosphamide Analogs p-toluenesulfonic acid (0.05 mmole) at reflux for 60h while removing water via a Dean-Stark trap. Triethyl orthoformate 30 (Boyd, et al., J. Med. Chem. 1980, 23, 372) (0.6 mmole) was added and resulting mixture was heated at A solution of bis(2-chloroethyl)phosphoramidic dichlo reflux for an additional hour. Extraction and evaporation gave ride (10 mmol) in ethyl acetate (100 mL) is added dropwise to 2-(2-furanyl)-2-(3-methyl)butyl-1,3-dioxolane as an a solution of commercial 3 methylamino-1-phenyl-1-pro orange liquid. panol (10 mmol) and triethylamine (20 mmol) in ethylacetate Step C: 35 (100 mL) at rt. After stirring at rt for 3 days, the salts are A solution of 2-(2-furanyl)-2-(3-methyl)butyl-1,3-diox filtered off and the combined filtrates are concentrated under olane (1 mmole) in THF was treated with TMEDA (1 mmole) vacuum. The crude product is purified by column chromatog and nBuLi (1.1 mmole) at -45°C., and the resulting reaction raphy on silica. mixture was stirred at -5 to 0° C. for 1 h. The resulting reaction mixture was cooled to -45° C., and cannulated into 40 Example 12 a solution of diethyl chlorophosphate in THF at -45° C. The reaction mixture was gradually warmed to ambient tempera Phosphoramide Prodrugs of ture over 1.25 h. Extraction and evaporation gave 2-2-(5- 1-Methylamino-1-phenyl-3-propanol diethylphosphono)furanyl-2-(3-methyl)butyl-1,3-diox olane as a dark oil. 45 Step A: Step D: The reagent N-methyl-2-(4-nitrophenoxy)-2-oxo-6-phe A solution of 2-2-(5-diethylphosphono) furanyl-2-(3- nyl-1,3,2-oxazaphosphorinane was made using the same pro methyl)butyl-1,3-dioxolane (1 mmole) in methanol was cedure as the one described for step A of example 2: treated with 1 N hydrochloric acid (0.2 mmole) at 60° C. for Fast eluting isomer: White solid, mp 135° C.; Rf-0.4 18 h. Extraction and distillation gave 5-diethylphosphono-2- 50 (Silica, Ethyl acetate/Hexanes 1/1). Slow eluting isomer: (4-methyl-1-oxo)pentylfuran as a light orange oil (bp 152 White solid, mp 128-129: Rf 0.15 (Silica, Ethyl acetate/ 156°C., 0.1 mmHg). Hexanes 1/1). Step E: Step B: A Solution of compound gave 5-diethylphosphono-2-(4- The coupling step with the nucleoside was accomplished in methyl-1-oxo)pentylfuran (1 mmole) in ethanol was treated 55 a manner similar to the one described in step B of example 3 with copper (II) bromide (2.2 mmole) at reflux for 3 h. The except that the reaction mixture was stirred at 100.degree. C. cooled reaction mixture was filtered and the filtrate was for 5 hours. evaporated to dryness. The resulting dark oil was purified by The following compound was prepared in this manner: chromatography to give 5-diethylphosphono-2-(2-bromo-4- 12.1: 9-(2-(N-Methyl-2-oxo-6-phenyl-1,3,2-oxazaphos methyl-1-oxo)pentylfuran as an orange oil. 60 phorin-2-oxy)-ethoxy)methyl-guanine: pale yellow Step F: amorphous solid; Rf-0.45 (Silica, Methanol/dichlo A solution of 5-diethylphosphono-2-(2-bromo-4-methyl romethane 2/8); H NMR (200 MHz, DMSO d) & 7.8 (bs, 1-oxo)pentylfuran (1 mmole) and thiourea (2 mmole) in 1H); 7.3 (bs, 5H), 6.75 (bs, 2H, exchangeable with DO); ethanol was heated at reflux for 2 h. The cooled reaction 5.35 (bs, 3H). mixture was evaporated to dryness and the resulting yellow 65 The following compound was prepared in this manner: foam was Suspended in Saturated Sodium bicarbonate and 12.2: 5-(N-Methyl-2-oxo-6-phenyl-1,3,2-oxazaphosphorin water (pH-8). The resulting yellow solid was collected 2-yl)-9-3-D-arabino-furanosyladenine: Off white amor US 8,080,536 B2 85 86 phous solid; Rf-0.2 (Silica, Methanol/dichloromethane Example B 1/9); H NMR (200 MHz, DMSO d) & 7.4-7.1 (m, 5H): 5.75 (s, 1H), 5.25 (m. 1H); 4.8 (m, 1H). Stability of Phosphoramidate Prodrugs to Esterases, Phosphatases, Adenosine Deaminase, and Plasma Example 13 Methods: Carboxylesterase (porcine liver) and alkaline Phosphoramide Prodrugs of phosphatase (calf intestinal mucose) are purchased from 3-amino-1-hydroxy-1-phenyl propane Sigma Chemical Co. (St. Louis, Mo.). Carboxylesterase activity is measured in 0.1 M Tris-HCl buffer at pH 8.0. Step A: 10 Activity towards p-nitrophenyl acetate, a known Substrate A mixture of 3-phenyl-3-hydroxypropylmethanesulfonate and positive control in the reactions is measured as described (1.71 g, Gao, et al., J. Org. Chem., 1988, 53. 4081), tetrahy for example by Matsushima M., et al. FEBS Lett. (1991)293 drofuran (10 mL) and 28% ammonium hydroxide (30 mL) (1-2): 37-41. Alkaline phosphatase activity is measured in a was heated in a bomb at 65°C. for 16 hours. After cooling, the 0.1 M diethanolamine buffer, pH 9.8, containing 0.5 mM volatiles were eliminated to leave the aqueous solution which 15 MgCl2. Activity towards p-nitrophenyl phosphate, a known was extracted with ether. The aqueous layer was concentrated Substrate and positive control in the reactions, is measured as under reduced pressure, azeotroped twice with anhydrous described e.g. Brenna O., et at (1975) Biochem J. 151(2): acetonitrile and dried under vacuum to give 3-amino-1-phe 291-6. Adenosine deaminase activity is measured as nyl-1-propanol (1.04 g) as a white solid: mp 92-94° C. described by e.g. Gustin NC, et al. Anal. Biochem. (1976) Rf-0.1 (silica, methanol/ethyl acetate 1/1). 71: 527-532. Adenosine is used as a positive control in these Step B: reactions. Plasma is prepared by centrifugation from fresh, Thereagent 2-(4-nitrophenoxy)-2-oxo-6-phenyl-1,3,2-ox heparinized rat or human blood. Prodrugs are incubated at a aZaphosphorinane (13.1) was made using the same procedure concentration of for example, 25uM in appropriate reaction as the one described for step A of example 2: Fast eluting 25 mixtures containing carboxylesterase, alkaline phosphatase, isomer: Rf-0.6 (Silica, Ethyl acetate), "H NMR (200 MHz, adenosine deaminase, or rat or human plasma. In the case of DMSO d) & 8.22 (m, 2H); 7.48 (m, 2H), 7.35 (m, 5H); 5.52 the esterase, phosphatase, and adenosine deaminase assays, (m. 1H). parallel reactions are run with known substrates of the Step C: enzymes as described. The coupling step with the nucleoside is accomplished in a 30 Aliqouts are removed from the reaction mixture at various manner similar to the one described in step B of example 3. time points and the reaction stopped by addition of methanol The following compounds are made in this manner: to 60%. Following centrifugation and filtration, the methan 9-(2-(N-Methyl-2-oxo-6-phenyl-1,3,2-oxazaphosphorin-2- olic aliquots are analyzed for generation of MPO (NHR) oxy)-ethoxy)methyl-guanine; or other metabolites by standard reverse phase, ion-pairing, or 5-(N-Methyl-2-oxo-6-phenyl-1,3,2-oxazaphosphorin-2-yl)- 35 ion exchange HPLC methods. 9-3-D-arabinofuranosyladenine. Results: MPO (NHR) or metabolites thereof are not Examples of use of the method of the invention includes the detected following exposure of the prodrugs to carboxy following. It will be understood that these examples are lesterase, alkaline phosphatase or plasma. exemplary and that the method of the invention is not limited solely to these examples. 40 Example C For the purposes of clarity and brevity, chemical com pounds are referred to as synthetic example numbers in the Activation of Phosphoramidate Prodrugs by Rat biological examples below. Liver Microsomes

BIOLOGICAL, EXAMPLES 45 Methods: The microsomal fraction is prepared from fresh, saline-perfused rat liver. Liver tissue is homogenized in three Example A volumes (w/v) of 0.2 MKHPO buffer pH 7.5, containing 2 mMMgCl2 and 1 mMEGTA. The homogenate is centrifuged Chemical Stability of an Antidiabetic at 10,000 g for 1 hour and the supernatant recovered. The Phosphoramidate Prodrug 50 supernatant fraction is then recentrifuged at 100,000 g to pellet the microsomal fraction. The pellet is resuspended in Methods: Aliquots of a 10 g/mL solution of Compound homogenization buffer and recentrifuged. This process is 1.3 in potassium phosphate buffers at pH 3 and 7 (room repeated twice to ensure complete removal of cytosolic temperature) were sampled hourly for 12 hours. Samples enzyme activities. After the last centrifugation, the microso were analyzed by reverse phase HPLC with use of a Beckman 55 mal pellet is resuspended in homogenization buffer at a final Ultrasphere C8 column (4.6x250 mm). The column was protein concentration of about 14 mg/ml. Reaction mixtures equilibrated and eluted with a gradient from 50 mM sodium (0.5 ml) consist of 0.2 M KHPO pH 7.5, 13 Mm glucose phosphate pH 5.5 to 80% acetonitrile at a flow rate of 1.0 6-phosphate, 2.2 mMNADP+, 1 unit of glucose-6-phosphate mL/min. Detection was at 254 nm. Under these conditions, dehydrogenase, 0-2.5 mg/ml microsomal protein and 100LM Compound 1.3 was separated from 2-Amino-5-isobutyl-4- 60 7.1. Reactions are carried out at 37°C. Aliquots are removed 2-(5-phosphonomonoamide)furanylthiazole, and from from the reaction mixtures at appropriate time points, and 2-Amino-5-isobutyl-4-(2-furanyl)thiazole (retention extracted with 60% methanol. The methanolic extracts are times=17, 15.8, and 15.6 min., respectively). centrifuged at 14,000 rpm, and filtered (0.2 uM) prior to Results: The half-life of Compound 1.3 at pH 3.0 was 3.8 h; analysis by HPLC. Standard HPLC techniques including prodrug decomposed to an unidentified metabolite at this pH. 65 reverse phase, ion-pairing, or ion exchange chromatography Compound 1.3 was fully stable at pH 7.0; there was no evi are used. Eluted peaks are quantified relative to authentic dence of decomposition throughout the 12-hour incubation. standards of known concentration. US 8,080,536 B2 87 88 Alternatively, the activation of prodrugs is monitored by Methods: Reactions (0.5 ml (a 37° C.) consist of 0.2 M the depletion of NADPH, an essential cofactor in the reaction. KHPO, 13 mM glucose-6-phosphate, 2.2 mM NADP+, 1 This assay is performed in reactions mixtures consisting of unit of glucose-6-phosphate dehydrogenase, 0-2.5 mg/ml 0.2M. KHPO, 0.22 mM NADPH, 0-2.5 mg/ml microsomal human microsomal protein (InVitro Technologies, Inc.), 250 protein, and 100 uM 28.4 or 30.1. Reactions are monitored uM prodrug, and 0-100 uM P450 isozyme inhibitor. Reac spectrophotometrically at 340 nm. A decrease in absorbance tions are stopped by addition of methanol to a concentration is indicative of cofactor depletion and thus of the enzymatic of 60%, and filtered (0.2 uM filter). MPO (NHR) is oxidation of prodrug to MPO (NHR). quantified by Standard reverse phase, ion-pairing, or ion Results: Prodrugs are converted to MPO (NHR) in the exchange HPLC methods. Eluted peaks are quantified rela presence, but not in the absence of NADP+ (this cofactor is 10 tive to authentic standards of known concentration. enzymatically reduced to NADPH by the dehydrogenase Results: Ketoconazole inhibits the formation of MPO. present in the reaction mixtures). This result indicates that an (NHR) in a dose-dependent fashion. The other inhibitors, oxidative step is involved in the activation of the prodrug. The furafylline and Sulfaphenazole, show no significant inhibi rate of activation of prodrugs to MPO (NHR) is linearly tion. The results indicate that CYP3A4 is the primary P450 dependent on microsomal protein concentration, confirming 15 isoform responsible for activation phosphoramidate prodrugs that activation occurs by an enzyme-dependent mechanism. in human liver. Example D Example F Activation of Phosphoramidate Prodrugs by Human Activation of Compound 1.3 by Recombinant Microsomes and the Identification and Tissue CYP3A4 Distribution of the Microsomal Enzymes Involved in Activation Activation of 1.3 was evaluated in reactions containing 25 microsomes from baculovirus-infected insect cells co-ex Methods: Human liver microsomes and cloned human pressing human recombinant CYP3A4 and cytochrome p450 P450 enzymes representing all major isoforms are obtained reductase (Panvera Corp., Madison, Wis.). from In Vitro Technologies and Gentest Inc., respectively. Methods: Reaction mixture composition was similar to Reaction mixtures typically contain 100 mM potassium phos that described in Example D. Reactions were terminated by phate buffer, pH 7.4, 2 mM NADPH, 0-2 mg/ml microsomal 30 addition of methanol to a final concentration of 60% and protein or cloned microsomal isozyme, and the prodrugat, for products were analyzed by HPLC as described in Example A. example, a 100 uM concentration. Formation of MPO Results: 2-Amino-5-isobutyl-4-2-(5-phosphonomonoa (NHR) is monitored by standard reverse phase, ion pairing, mide)furanylthiazole was generated from 1.3 at a rate of 4.2 or anion exchange HPLC methods. MPO(NHR) is nmoles/min./nmole CYP3A4. detected by uv absorbance and quantified relative to authentic 35 standards. To determine the tissue distribution of the activat Example G ing enzyme, homogenates are prepared from key tissues including the liver, lung, intestine, kidney, heart, and skeletal Activation of Antiviral Phosphoramidate Prodrugs in muscle. The rate of conversion of prodrug to MPO Isolated Rat Hepatocytes (NHR) in each homogenate is assessed as described for the 40 microsomal reactions above. Alternatively, once the major The activation of 3.1 and 3.2 was evaluated by monitoring catalytic isoform is identified, immunohistological methods the intracellular generation of the corresponding nucleoside using antibodies raised against the specific enzyme (Waziers triphosphates in rathepatocytes. et al 1989, J. Pharm. Exp. Ther. 254:387), or molecular Methods: Hepatocytes were prepared from fed, dexam biological methods using PCR techniques (Sumidaetal 2000, 45 ethasone-induced Sprague-Dawley rats (300 g) according to BBRC 267: 756) are employed to determine the tissue distri the procedure of Berry and Friend (Berry, M.N., Friend, D.S. bution of the activity. J. Cell Biol. 43, 506-520 (1969)) as modified by Groen Results: Prodrugs are transformed to MPO (NHR) in (Groen, A. K. et al. Eur J. Biochem 122, 87-93 (1982)). the human microme-catalyzed reaction at a readily measur Hepatocytes (62 mg wet weight/ml) were incubated in 2 ml able rate. In the screen of cloned microsomal isozymes, the 50 Krebs bicarbonate buffer containing 10 mM glucose, and 1 CYP3A4 isozyme catalyzes activation of the prodrugs at the mg/ml BSA. Incubations were carried out in a 95% oxygen, highestrate although other isozymes also show some activity 5% carbon dioxide atmosphere in closed, 50-ml Falcon tubes towards the prodrugs. The tissue distribution studies indicate submerged in a rapidly shaking water bath (37° C.). 3.1 and a high activity or abundance of the activating enzyme in the 3.2 were dissolved in methanol to yield 25 mM stock solu liver relative to other tissues. 55 tions, and then diluted into the cell Suspension to yield a final concentration of 250 uM. Two hours following administra Example E tion, aliquots of the cell Suspension were removed and spun through a silicon/mineral oil layer into 10% perchloric acid. Identification of the P450 Isozyme Involved in The cell extracts in the acid layers were neutralized by the Phosphoramidate Prodrug Activation with Use of 60 addition of 0.3 volumes of 3M KOH/3M KHCO, and the Specific Enzyme Inhibitors extract was then centrifuged, filtered (0.2 micron) and loaded onto an anion exchange HPLC column equilibrated with 70% Prodrugs are evaluated for human microsome-catalyzed A (10 mM ammonium phosphate pH 3.5, 6% ETOH) and conversion to MPO. (NHR) in the absence and presence 30% B (1 M ammonium phosphate pH 3.5, 6% ETOH). of specific inhibitors of three major P450 isozymes: keto 65 AraATP and acyclovir-triphosphate, the expected metabo conazole (CYP3A4), furafylline (CYPIA2), and sul lites of 3.1 and 3.2, respectively, were eluted from the column faphenazole CYP2C9). with a linear gradient to 80% B in 20 minutes. Detection was US 8,080,536 B2 89 90 by uv absorbance at 254 nm. Peak areas were quantified by with IC50s of 8.5 and 2.5uM, respectively. Since 1.3 is not comparison of peak areas to authentic standards spiked into an inhibitor of FBPase in vitro, inhibition of gluconeogenesis naive hepatocyte extracts. in hepatocytes confirms that prodrug was converted to Results: Both prodrugs generated their respective triphos 2-Amino-5-isobutyl-4-(2-furanyl)thiazole intracellularly. phates in isolated rathepatocytes. Levels formed following The -3-fold lower potency of 1.3 relative to 2-Amino-5- two hours of incubation are summarized below: isobutyl-4-(2-furanyl)thiazole is consistent with time-depen dent activation of prodrug to parent compound in cells. Example J Intracellular concentration 10 Compound nmoles.g Activation of Antidiabetic and Antiviral 3.1 15.7 16 Phosphoramidate Prodrugs in Induced Rat 3.2 13.33 Hepatocytes 15 Compounds 1.7 and 12.1 was tested for activation in hepa Example H tocytes isolated from rats in which CYP3A4 expression was induced by treatment with dexamethasone. This treatment Kinase Bypass results in more extensive intracellular activation of phospho ramidate prodrugs. The generation of acyclovir triphosphate from a phospho Methods: Rats were treated with dexamethasone (50 ramidate prodrug of acyclovir (Compound 3.2) and acyclovir mg/kg, intraperitoneally) for 4 days as described (Brain EG Cetal 1998, Br. J. Cancer 7: 1768). Isolated hepatocytes were itself was compared in rathepatocytes. prepared, and prodrugs tested for activation as described in Methods: 3.2 and acyclovir were evaluated in isolated rat Example G. hepatocytes at 250 uM as described in Example G. 25 Results: 1.7 activated to yield 20.5+3 nmole/g levels of Results: Levels of acyclovir triphosphate formed following 2-Amino-5-methylthio-4-2-(5 phosphono) furanylthiazole two hours of incubation are summarized below: following two hours of incubation. 12.1 generated 110+0.2 mmoles 1 g acyclovir triphosphate.

30 Intracellular concentration Example K Compound nmoles.g Cytotoxicity Testing of Phosphoramidate Prodrugs in acyclovir <2 Non-CYP3A4 Expressing Cell Lines 3.2 13.33 35 Administration of a drug in a plasma-stable prodrug form The results demonstrate that a phosphoramidate prodrug of can reduce systemic toxicities by limiting exposure to free a parent nucleoside Such as acyclovir, that is poorly phospho MPO (NHR) and metabolites. This potential advantage rylated in cells, can bypass the nucleoside phosphorylation of prodrug delivery is demonstrated by comparing the toxic step and thus generate higher intracellular levels of triphos ity profile of the prodrug and the corresponding parent com phate than the free nucleoside. 40 pound in a cell line which does not express the CYP3A4 enzyme required for prodrug activation. Example I Methods: The choice of cell line is dictated by the known toxicity profile of the parent compound. If the toxicity profile Inhibition of Glucose Production in Rat Hepatocytes of a parent compound is unknown, a panel of different cul by Compound 1.3 45 tured cell lines can be tested. Cells are exposed to a range of prodrug and parent compound concentrations for hours to Compound 1.3 and its phosphonic acid parent compound, days. Viability is the measured by Trypan blue exclusion, 2-Amino-5-isobutyl-4-(2-furanyl)thiazole, were tested in enzyme marker leakage, incorporation of labeled thymidine isolated rathepatocytes for inhibition of glucose production. into DNA or other standard method. Methods: Isolated rat hepatocytes were prepared from 50 Results: Phosphoramidate prodrugs are either noncyto fasted Sprague Dawley rats (250-300 g) as described in toxic or cytotoxic at considerably higher concentrations than Example G. Hepatocytes (75 mg wet weight/ml) were incu the corresponding parent compound. The latter profile Sug bated in 1 ml Krebs bicarbonate buffer containing 10 mM gests that even if systemic exposure to the prodrug is high, it glucose, and 1 mg/ml BSA. Incubations were carried out in a is likely to show reduced peripheral toxicity in vivo relative to 95% oxygen, 5% carbon dioxide atmosphere in closed, 50-ml 55 free parent compound. Testing prodrugs in this manner allows Falcon tubes submerged in a rapidly shaking water bath (37° an assessment of the intrinsic toxicity of the prodrug and can C.). After 10 minutes of equilibration, lactate and pyruvate be used to select prodrugs with favorable cellular safety pro were added to 10 mM and 1 mM concentrations, respectively. files. Addition of these gluconeogenic Substrates was immediately followed by the addition of test compound to concentrations 60 Example L ranging from 1-100 uM. After 1 hour, an aliquot (0.25 ml) was removed, and transferred to an Eppendorf tube and centri Cytotoxicity Testing in Cellular Systems that fuged. The resulting Supernatant (50LLA) was then assayed for Catalyze Phosphoramidate Prodrug Activation glucose content using a Glucose Oxidase kit (Sigma Chemi cal Co.) as per the manufacturers instructions. 65 Cytotoxicity testing in Systems in which phosphoramidate Results: 1.3 and 2-Amino-5-isobutyl-4-(2-furanyl)thiaz prodrugs are activated allows an assessment of the safety of ole inhibited glucose production in a dose dependent manner MPO (NHR) and other metabolites of the prodrug or US 8,080,536 B2 91 92 parent compound that are generated. Several cellular systems Results: Acetaminophen exposure (50, 250 and 500 are useful for testing: (a) fresh primary cultures of hepato mg/kg) resulted in a dose-dependent reduction of hepatic cytes of human or other origin in which CYP3A4 expression glutathione. Liver toxicity in the form of liver enzyme eleva is maintained with an appropriate inducer Such as Rifampicin, tions and the appearance of necrotic lesions was evident only (b) cell lines in to which CYP3A4 and the requisite reductase at the highest dose, which depleted hepatic glutathione by are cotransfected, (c) cell lines that are coincubated with >80%. This is in accordance with the literature; glutathione primary hepatocytes that express CYP3A4, (d) cell lines that forms an adduct with the electrophilic acetaminophen are coincubated with CYP3A4 containing micromal fractions metabolite formed, and protects against liver injury until a or a cloned CYP3A4 preparation in addition to a standard threshold level of depletion is reached (Mitchell J R et al NADPH recycling system. Prodrugs are incubated in these 10 1973, J. Pharm. and Exp. Ther 187: 211). Phosphoramidate systems for hours or days. Cytotoxicity is assessed by stan prodrug administration also results in glutathione depletion, dard methods such as the Trypan blue exclusion technique, indicating that the byproduct formed is efficiently detoxified. the leakage of enzyme markers, the incorporation of labeled Liver injury is apparent only at very high doses well above the precursors into cellular DNA, etc. projected therapeutic doses in man. The data also show dose 15 dependent accumulation of MPO (NHR) and/or its Example M metabolites following phosphoramidate prodrug administra tion. Byproduct Toxicity Testing. In Vitro Example O Activation of certain phosphoramidate prodrugs, in addi tion to generating MPO (NHR), also results in the for Enhanced Delivery of Drugs to the Liver in Animals mation of an electrophilic byproduct capable of alkylating Pretreated with CYP3A-Inducing Agents cellular constituents. As the prodrugs in question are acti vated primarily in the liver, primary rathepatocytes, known to Hepatic CYP3A enzymes are induced in rats by treatment contain both the microsomal enzymes required for prodrug 25 with dexamethasone or other suitable agent. Enhanced activation as well as critical detoxification mechanisms, are expression of CYP3A will result in a higher rate of prodrug the cell type of choice for testing the potential toxicity of the activation and thus enhance the distribution of MPO byproduct formed. (NHR) and/or its metabolites to this organ. Methods: Isolated rathepatocytes are prepared and incu Methods: Rats are treated with dexamethasone (50 mg/kg, bated with test compound as described in Example G. 30 intraperitoneally) for 4 days as described (Brain E G C et al Acetaminophen (0-10 mM) is used as a positive control in 1998, Br. J. Cancer 7: 1768). Other CYP-inducing agents these tests since its microsomal metabolism is also known to such as phenobarbital or rifampicin may also be used. The generate an electrophilic metabolite in these cells (Smolarek induced animals are then administered prodrug orally or T A et al 1989, Metabolism and Disposition 18: 659). This Sytemically and serially sacrificed at various time points. metabolite is detoxified by addition to glutathione, a protec 35 Livers are removed and homogenized in perchloric acid tive thiol present in mM concentrations in liver cells. Incuba (10%). Following clarification by centrifugation and neutral tions with acetaminophen and phosphoramidate prodrugs are ization, MPO (NHR) and/or its metabolites in the homo for 4 hours. Cellular viability is assessed by lactate dehydro genates are quantified by standard HPLC methods. A similar genase or other liver enzyme leakage and/or by Trypan blue study is conducted in uninduced animals. exclusion. Intracellular glutathione stores are also quantified 40 Results: The AUC of MPO (NHR) and/or its metabo as described (Baker MAetal 1990, Anal. Biochem. 190:360). lites in livers of induced rats will exceed that of uninduced Results: Acetaminophen depleted intracellular glutatione rats. Enhanced liver delivery of MPO (NHR) and/or its levels by 90% at the highest dose tested (10 mM) but did not metabolites resulting from CYP3A induction is expected to compromise cellular viability as measured by Trypan blue result in increased efficacy. exclusion. The data Suggest that the byproduct generated as a 45 result of acetaminophen metabolism was readily detoxified in Example P the hepatocytes, and that the cellular capacity for detoxicifi cation was not exceeded. Activation of phosphoramidate pro Oral Bioavailability of Phosphoramidate Prodrugs drugs does not cause byproduct-related toxicities in this hepa tocyte system. 50 The oral bioavailability (OBAV) of phosphoramidate pro drugs is estimated by comparison of the area under the curve Example N (AUC) of MPO (NHR) and/or its metabolites generated in liver following oral administration to that generated fol Byproduct Toxicity Testing. In Vivo lowing intravenous administration. Alternatively, OBAV is 55 determined by means of a urinary excretion assay of MPO Toxicity of the electrophile generated following microso (NHR) and/or its metabolites following prodrug adminis mal activation of certain phosphoramidate prodrugs is tested tration. in the mouse. Methods: Rats are dosed orally and systemically with a Methods: Mice are administered various doses of prodrug phosphoramidate prodrug in a suitable vehicle and serially and acetaminophen (a positive control, see Example M) and 60 sacrificed at appropriate time point following drug adminis serially sacrificed at various time points following drug tration. Liver and plasma samples are obtained, processed, administration. Livers are removed and processed for glu and analyzed for MPO (NHR) and/or its metabolites by tathione analysis as described in Example M, and for drug standard reverse phase, ion-pairing, or ion exchange HPLC analysis by standard reverse phase, ion-pairing, or ion methods. Alternatively, animals are placed in metabolic cages exchange HPLC methods. Plasma samples are subjected to a 65 following drug administration, and urine is collected for standard chemistry panel in which markers of liver function 24-48 hours. The quantity of MPO (NHR) and/or its are evaluated. metabolites excreted into urine is then determined by HPLC US 8,080,536 B2 93 94 analysis. Oral bioavailability is calculated either by compar ing as a MPO. (NHR) reservoir and, by virtue of a pro ing the AUC of MPO (NHR) and/or its metabolites in the longed in vivo half-life, providing Sustained release of liver or the quantity in urine following oral and systemic MPO (NHR) and/or its metabolites to the liver. administration. Results: The data indicate that phosphoramidate prodrugs We claim: are suitable for the oral delivery of MPO (NHR) and/or 1. A compound of formula I: its metabolites. Example Q 10 Enhanced Liver Delivery and Reduced Systemic Exposure of MPO (NHR) and/or its Metabolites Following Administration of Phosphoramidate Prodrugs 15 Method: Rats are dosed either orally or systemically with a phosphoramidate prodrug, MPO(NHR), and/or its metabolites. Animals are serially sacrificed at appropriate wherein: time points following drug administration. Liver and plasma V, W, and W are independently selected from the group samples are obtained, processed, and analyzed for MPO. consisting of —H, alkyl, aralkyl, alicyclic, aryl, Substi (NHR) and/or its metabolites by standard reverse phase, tuted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, ion-pairing, or ion exchange HPLC. and 1-alkynyl; or Results: Administration of phosphoramidate prodrugs togetherV and Z are connected via an additional 3-5 atoms results in the generation of higher levels of MPO (NHR) to form a cyclic group containing 5-7 ring atoms, option and/or its metabolites in liver and their reduced concentra 25 ally 1 heteroatom, substituted with hydroxy, acyloxy, tions in plasma than when free MPO (NHR) is adminis alkoxycarbonyloxy, or aryloxycarbonyloxy attached to tered. a carbon atom that is three atoms from both Y groups attached to the phosphorus; or Example R togetherV and Z are connected via an additional 3-5 atoms 30 to form a cyclic group, optionally containing 1 heteroa Reduced Renal Excretion of MPO (NHR) tom, said cyclic group is fused to an aryl group at the beta and/or its Metabolites Following Administration of and gamma position to the Y adjacent to V: Phosphoramidate Prodrugs together V and Ware connected via an additional 3 carbon atoms to form an optionally substituted cyclic group Methods: Phosphoramidate prodrugs and respective 35 containing 6 carbon atoms and Substituted with one Sub MPO (NHR) are administered systemically to rats. Rats stituent selected from the group consisting of hydroxy, are Subsequently caged in metabolic cages and urine col acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and lected over a 24-48 hour period. MPO (NHR) and/or its aryloxycarbonyloxy, attached to one of said additional metabolites are quantified in urine by means of standard carbonatoms that is three atoms from aYattached to the reverse phase, ion-pairing, or ion exchange HPLC methods. 40 phosphorus; By relating the amount of parent compound/metabolites together Zand Ware connected via an additional 3-5 atoms excreted in urine to the dose administered, the '% renal clear to form a cyclic group, optionally containing one het ance is calculated. eroatom, and V must be aryl, substituted aryl, heteroaryl, Results: Renal excretion of MPO (NHR) and/or its or substituted heteroaryl; metabolites following phosphoramidate prodrug administra 45 together W and W are connected via an additional 2-5 tion is lower than when parent compound is administered. atoms to form a cyclic group, optionally containing 0-2 This result indicates that the renal exposure and thus the renal heteroatoms, and V must be aryl, substituted aryl, het toxicity associated with certain parent compounds and/or eroaryl, or substituted heteroaryl; their metabolites may be avoided by administration of the Z is selected from the group consisting of —CHROH, compound in phosphoramidate prodrug form. 50 CHROC(O)R, CHROC(S)R, CHROC(S) OR, CHROC(O)SR, CHROCOR, OR, Example S —SR, —CHRN, -CHaryl, -CH(aryl)OH, -CH (CH=CR)OH, -CH(C=CR)OH, R, NR, Sustained Liver Drug Levels –OCOR, OCOR, SCOR, SCOR, NH 55 COR, NHCO.R, CH-NHaryl, -(CH.) OR', Methods: Rats are dosed orally and systemically with a and -(CH.). SR': phosphoramidate prodrug or the corresponding MPO p is an integer 2 or 3: (NHR) and serially sacrificed at appropriate time point with the provisos that: following drug administration. Liver samples are obtained, a) V, Z, W, Ware not all-H; and processed, and analyzed for MPO (NHR) and/or its 60 b) when Z is R, then at least one of V, W, and W is not metabolites by standard reverse phase, ion-pairing, or ion —H, alkyl, aralkyl, or alicyclic; exchange HPLC. Liver half-lives are determined with the aid R’ is selected from the group consisting of Rand-H; of WinNonLin 1.1 software (Scientific Consulting, Inc.). R is selected from the group consisting of alkyl, aryl, Results: The half-life of MPO (NHR) and/or its alicyclic, and aralkyl; metabolites in liver is longer when administered in phospho 65 R is selected from the group consisting of H, and lower ramidate prodrug form than when free MPO (NHR) is alkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl and lower administered. This result is consistent with the prodrug serv acyl: US 8,080,536 B2 95 96 R" is selected from the group consisting of H, and lower 12. The compounds of claim 1 wherein acyl: V, W, and W are independently selected from the group each Y is independently selected from the group consisting consisting of —H, alkyl, aralkyl, alicyclic, aryl, Substi of O. , NR with the proviso that at least one Y tuted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, is NR : 5 and 1-alkynyl; or Misan agent selected from the group that when attached to together V and Ware connected via an additional 3 carbon H, PO, P.O., P.O." or P(O)(NHR)O is a biologi atoms to form an optionally substituted cyclic group cally active agent, but is not an FBPase inhibitor, and is containing 6 carbon atoms and Substituted with one Sub attached to the phosphorus in formula I via a carbon, stituent selected from the group consisting of hydroxy, oxygen, Sulfur or nitrogen atom of the M group; 10 acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and with the provisos that: aryloxycarbonyloxy, attached to one of said additional 1) M is not NH(lower alkyl). —N(lower alkyl), NH carbonatoms that is three atoms from aYattached to the (lower alkylhalide), N(lower alkylhalide), or phosphorus. —N(lower alkyl)(lower alkylhalide); and 15 13. The compounds of claim 12 wherein V is selected from 2) R' is not lower alkylhalide; the group consisting of aryl, Substituted aryl, heteroaryl, Sub and pharmaceutically acceptable salts thereof. stituted heteroaryl; or 2. The compounds of claim 1 wherein M is an agent together V and Ware connected via an additional 3 carbon selected from M-P(O)(NHR)O, M-PO, M-PO, and atoms to form a cyclic Substituted group containing 6 M-PO, such that M-P(O)(NHR)O, M-PO, carbon atoms and mono-Substituted with a Substituent M-PO, and M-PO, is a biologically active agent Selected from the group consisting of hydroxyl, acyloxy, selected from the group consisting of an antiviral, anticancer, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxy antihyperlipidemic, antifibrotic, antiparasitic agents, metal carbonyloxy attached to one of said additional carbon loprotease inhibitor, and TS inhibitor. 25 atoms that is three atoms from an Y attached to the 3. The compounds of claim 2 wherein M is an agent phosphorus. selected from M-H, such that M-H is a biologically active 14. The compounds of claim 13 wherein V is selected from agent selected from the group consisting of LdC. LdT, araA, the group consisting of aryl, Substituted aryl, heteroaryl, and AZT, d4T, dd, ddA, ddC, L-ddC, L-FddC, L-d4C, L-Fd4C, substituted heteroaryl. 3TC, ribavirin, penciclovir, 5-fluoro-2'-deoxyuridine, FIAU, 30 FIAC, BHCG, 2R,5'S(-)-1-(2-(hydroxymethyl)oxathiolan 15. The compounds of claim 14 wherein Z. W. and Ware 5-ylcytosine, (-)-b-L-2',3'-dideoxycytidine, (-)-b-L-2',3'- H; and R is selected from the group consisting of -H, and dideoxy-5-fluorocytidine, FMAU, Bvara J., E-5-(2-bromovi lower alkyl. nyl)-2'-deoxyuridine, Cobucavir, TFT, 5-propynyl-1- 16. The compounds of claim 14 wherein V is selected from arabinosyluracil, CDG, DAPD, FDOC, d4C, DXG, FEAU, 35 the group consisting of phenyl, and Substituted phenyl. FLG, FLT. FTC, 5-yl-carbocyclic 2'-deoxyguanosine, Cytal 17. The compounds of claim 14 wherein V is selected from lene, Oxetanocin A, Oxetanocin G, Cyclobut A, Cyclobut G, the group consisting of 3,5-dichlorophenyl, 3-bromo-4-fluo fluorodeoxyuridine, dFdC, araC, bromodeoxyuridine, IDU, rophenyl, 3-chlorophenyl, 3-bromophenyl, 3,5-difluorophe CdA, F-ara.A, 5-FdUMP, Coformycin, 2'-deoxycoformycin, nyl, and 4-pyridyl. ACV, GCV, penciclovir, (R)-9-(3,4-dihydroxybutyl)guanine, 40 18. The compounds of claim 13 wherein together V and W and cytallene. are connected via an additional 3 carbon atoms to form an 4. The compounds of claim 1 wherein M is selected from an optionally substituted cyclic group containing 6 carbonatoms agent selected from M-PO, such that M-PO, is a bio and mono-substituted with one substituent selected from the logically active agent selected from the group consisting of group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, PMEA, PMEDAP, HPMPC, HPMPA, FPMPA, and PMPA 45 alkylthiocarbonyloxy, and aryloxycarbonyloxy attached to 5. The compounds of claim 1 wherein MP(O)(NHR)O, one of said additional carbon atoms that is three atoms from MPO. , MPO, or MP.O." is useful for the treatment of an Yattached to the phosphorus. diseases of the liver or metabolic diseases where the liver is 19. The compounds of claim 18 wherein together V and W responsible for the overproduction of a biochemical end prod 50 form a cyclic group selected from the group consisting of uct. —CH-CH(OH)—CH , —CHCH(OCOR)-CH , 6. The compounds of claim 5 wherein said disease of the and —CHCH(OCOR)-CH-. liver or metabolic disease is selected from the group consist 20. The compounds of claim 12 wherein said compound is ing of hepatitis, cancer, fibrosis, malaria, gallstones, chronic selected from a compound of formula VI: cholecystalithiasis, diabetes, atherosclerosis, and obesity. 55 7. The compounds of claim 5 wherein said biochemical end product is selected from the group consisting of glucose, VI cholesterol, fatty acids, and triglycerides. 8. The compounds of claim 7 wherein MPO' or MP(O) (NHR)O is an AMP activated protein kinase activator. 60 9. The compounds of claim 1 wherein Y is —O—located adjacent to the W and W groups. 10. The compounds of claim 1 wherein Y is —O—located adjacent to the V group. 65 wherein 11. The compounds of claim 1 wherein both Y groups are V is selected froth the group consisting of aryl, substituted NR . aryl, heteroaryl, and substituted heteroaryl. US 8,080,536 B2 97 98 21. The compounds of claim 12 wherein said compound is D is H: selected from a compound of formula VII: D is selected from the group consisting of -H, alkyl, -OH, and —OC(O)R. 23. The compounds of claim 12 wherein Wand Z are —H, VII 5 W and V are both the same aryl, substituted aryl, heteroaryl, or substituted heteroaryl, and both Y groups are the same —NR , such that the phosphonate moiety:

10 V wherein O Y Z is selected from the group consisting of: V / -P CHROH, CHROC(O)R’, CHROC(S)R, V CHROCOR, CHROC(O)SR, CHROC(S)OR, Y and —CH2aryl. 15 22. The compounds of claim 12 wherein said compound is W selected from a compound of formula VIII: has a plane of symmetry through the phosphorus-oxygen double bond. VIII 20 24. A method of enhancing oral bioavailability of a parent drug by administering to an animal a compound of claim 1. 25. The method of claim 24 wherein said oral bioavailabil ity is at least 10%. 26. The method of claim 24 wherein said oral bioavailabil 25 ity is enhanced by 50% compared to the parent drug admin wherein istered orally. Z is selected from the group consisting of —OH, - OC (O)R, OCO, R, and OC(O)S R: