AU2004200670A1 - Novel Prodrugs for Phosphorous-containing Compounds - Google Patents

Novel Prodrugs for Phosphorous-containing Compounds Download PDF

Info

Publication number
AU2004200670A1
AU2004200670A1 AU2004200670A AU2004200670A AU2004200670A1 AU 2004200670 A1 AU2004200670 A1 AU 2004200670A1 AU 2004200670 A AU2004200670 A AU 2004200670A AU 2004200670 A AU2004200670 A AU 2004200670A AU 2004200670 A1 AU2004200670 A1 AU 2004200670A1
Authority
AU
Australia
Prior art keywords
group
aryl
alkyl
substituted
alicyclic
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
AU2004200670A
Other versions
AU2004200670B2 (en
Inventor
Mark D. Erion
K. Raja Reddy
Edward D. Robinson
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Metabasis Therapeutics Inc
Original Assignee
Metabasis Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU30699/99A external-priority patent/AU767599B2/en
Application filed by Metabasis Therapeutics Inc filed Critical Metabasis Therapeutics Inc
Priority to AU2004200670A priority Critical patent/AU2004200670B2/en
Publication of AU2004200670A1 publication Critical patent/AU2004200670A1/en
Application granted granted Critical
Publication of AU2004200670B2 publication Critical patent/AU2004200670B2/en
Anticipated expiration legal-status Critical
Expired legal-status Critical Current

Links

Classifications

    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Description

20.FEB-2004 17:28 SPRUSON FERGUSON NO. 7145 P. S&F Ref: 520864D1
AUSTRALIA
PATENTS ACT 1990 COMPLETE
SPECIFICATION
FOR A STANDARD PATENT Name and Address of Applicant: Actual Inventor(s): Address for Service: Invention Title: Metabasis Therapeutics, Inc.
9390 Towne Centre Drive San Diego California 92121 United States of America Mark D Erion K Raja Reddy Edward D Robinson Spruson Ferguson St Martins Tower Level 31 Market Street Sydney NSW 2000 (CCN 3710000177) Novel Prodrugs for Phosphorus-containing Compounds The following statement is a full description of this invention, including the best method of performing it known to me/us:- 5845C [R:\LIBF]0018 d4oo:stl COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:28 SPRUSON FERGUSON NO. 7145 P. 6 NOVEL PRODRUGS FOR PHOSPHORUS-CONTAINING
COMPOUNDS
Field of the Invention The present invention is directed towards novel prodrugs of phosphate, phosphonate, and phosphoraridate compounds which in their active form have a phosphate, phosphonate, or phosphoramidate group, to their preparation, to their synthetic intermediates, and to their uses.
More specifically, the invention relates to the area of substituted cyclic 1,3-propanyl phosphate, phosphonate and phosphoramidatc esters.
Backround of the Invention The following description of the background of the invention is provided to aid in understanding the invention, but is not admitted to be, or to describe, prior art to the invention.
Free phosphorus and phosphonic acids and their salts are highly charged at physiological pH and therefore frequently exhibit poor oral bioavailiability, poor cell penetration and limited tissue distribution CNS). In addition, these acids are also commonly associated with several other properties that hinder their use as drugs, including short plasma half-life due to rapid renal clearance, as well as toxicides renal, gastrointestinal, etc.) Antimicrob Agents Chemother 1998 May; 42(5): 1146-50). phosphates have an additional limitation in that they are not stable in plasma as well as most tissues since they undergo rapid hydrolysis via the action of phosphatases alkaline phosphatasc, nucleoridases). Accordingly, phosphate esters are frequently used as a prodrug strategy, especially for water insoluble compounds, since the phosphate group enables high water solubility and thereby enables delivery of the drug parenterally and is rapidly broken down to the parent drug.
Prodrugs of phosphorus-containing compounds have been sought primarily to improve the limited oral absorption and poor cell penetration. In contrast to carboxylic acid proesters, many phosphonate and phosphate esters fail to hydrolyze in vivo, including simple alkyl esters.
The most commonly used prodrug class is the acyloxyalkyl ester, which was first applied to COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:28 SPRUSON FERGUSON NO. 7145 7- 2 phosphate and phosphonate compounds in 1983 by Farquhar e: al. 1 PhA S 72(3): 324 (1983). The strategy entails cleavage of a carboxylc ester by esterses to generate an unstable hydroxyalkyl intermediate which subsequently breaks down to generate the drug and an aldehyde. In some cases this biproduct formaldehyde), can be toxic. This strategy is uscd to enhance the bioavailability for several drugs. For example, the bis(pivoyloxymthyl) prodnag of the antiviral phosphonate 9 2 -phospbonylmethoxyethyl)adenine (PMEA) has been studied clinically for the treatmentof CMV infection and the bis(pivaloyloxymethyl) prodrug of the squalene synthetase inhibior BMS 187745 is undergoing clinical evaluation for the treatment of hypercholesterolemia and associated cardiovascular diseases. The marketed antihypei-jjv, fosinopril, is a phosphinic acid angiotensin converting enzyme inhibitor that requires the use of an isobutrylaxyethyl group for oral absorption.
Several other esters have been used as prodrugs of phosphorus-containing compounds For example, aryl esters, especially phonyl esters, are another prodrug clus reported to be sful for the delivery of phosphorus-containing compounds. DeLambert et al., Chm- 37 498 (1994). Phenyl esters containing a carboxylic ester ortho to the phosphate have also been described. IKhamnei and Torrence, 1. Md Chcj.; 39:4109-4115 (1996).
Benzyl esters are reported to generate the parent phosphonic acid. In some cases using substituents at the ro_gr,- or pra-position can accelerate the hydrolysis. Benzyl analogs with an acylated phenol or an alkylated phenol can generate the phenolic compound through the action of enzymes, e.g. esterases, oxidases, etc., which in tumrn undergoes cleavage at the benzylic
C-O
bond to generate the phosphonic acid and the potentially toxic quinone methide intermediate.
Examples of this class of prodrugs are described by Mitchell et al., L Chem. oc. Prkd Trans. I 2345 (1992); Brook. et al. WO 91/19721. Still other benaylic prodrugs'have been described containing a carboxylic ester-containing group attached to the benzylic methylene. Glazier et al.
WO 91/19721.
Cyclic phosphonate esters have also been described for phosphorus-containing compounds. In some cases, these compounds have been investigated as potential phosphate or phosphonate prodrugs. Hunston et at., Med. Chem. 27: 440-444(1984). The numbering for these cyclic esters is shown below: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:29 SPRUSON FERGUSON NO. 7145 P. 8 3 p0 2' 3' The cyclic 2,2'-difluoro-l',3'-propane ester is reported to be hydrolytically unstable with rapid generation of the ring-opened monoester. Starrett et al. J. M Ch m 37: 1857-1864 (1994), Cyclic 3',5'-phosphate esters of araA, araC and thioinosine have been synthesized.
Meier et alJ. L~Md.Chm.22: 811-815 (1979). These compounds are ring-opened through the action of phosphodiesterases which usually require one negative charge.
Cyclic 1 '.3'-propanyl phosphonate and phosphate esters are reported containing a fused aryl ring, i.e. the cyclosaligenyl ester, Meier et al., Bioore. Md. Chem. Lett 7: 99-104 (1997).
These prodrugs are reported to generate the phosphate by a "controlled, non-enzymatic mechanism(s] at physiological pH according to the designed tandem-reaction in two coupled steps". The strategy was purportedly used to deliver d4-T monophosphate to CEM cells and CEM cells deficient in thymidine kinase infected with HIV-1 and HIV-2.
Unsubstituted cyclic 1', 3 '-propanyl esters of the monophosphates of (Farquhar et aL, J. Med, Chem 26:; 1153 (1983)) and ara-A (Farquhar et Med.Chem. 28: 1358 (1985)) were prepared but showed no in vivo activity. In addition, cyclic 1 ',3'-propanyl esters substituted with a pivaloyloxy methyloxy group at C-I' was prepared for 5-fluoro-2'deoxy-uridinemonophosphate (5-dUMP: (Freed et al., Biochem. Pharmac. 38: 3193 (1989); and postulated as potentially useful prodrugs by others (Biller et al., US 5,157,027). In cells, the acyl group of these prodrugs underwent cleavage by esterases to generate an unstable hydroxyl intermediate which rapidly broke down to the free phosphate and acrolein following a Jl.
elimination reaction as well as formaldehyde and pivalic acid.
Cyclic phosphoramidates are known to cleave in vivo by an oxidative mechanism. For example, cyclophosphoramide is thought to undergo oxidation at C- to form the hydroxylated intermediate, which like the 1 '-substituted cyclic '-propane esters described above, breaks down to acrolein and the corresponding phosphoramidate. Cyclophosphoramidates were also prepared as potential prodrugs of both 5-FdUMP and araAMP and shown to have modest activity in vivo.
A variety of substituted propanyl cyclic phosphoramidates, wherein 1' represents the carbon alpha to the nitrogen were prepared as cyclophosphamide analogs (Zon, Progress in COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:29 SPRUSON FERGUSON NO. 7145 P. 9 4 Med. Chem. 19, 1205 (1982)). For example, a number of 2' and 3'-substituted proesteres were prepared in order to decrease the propensity of the a,-unsubstituted carbonyl bi-product to undergo to a Michael reaction. 2 '-Substituents included methyl, dimethyl, bromo, trifluoromethyl, chloro, hydroxy, and methoxy whereas a variety of groups were used at the 3'position including phenyl, methyl, trifluoromcthyl, ethyl, propyl, i-propyi, and cyclohexyl.
Analogs with a 3'-aryl group underwent oxidation alpha to the nitrogen and accordingly exhibited anticancer activity in the mouse L1210 assay. A variety of I '-substituted analogs were also prepared. In general these compounds were designed to be "pre-activated" cyclophosphamide analogs that bypass the oxidation step by already existing as a I '-substituted analog capable of producing the final compound, e.g. hydroperoxide and thioether. A series of 1 '-aryl analogs were also prepared in order to enhance the oxidation potential. In contrast to the I '-hydroperoxy analogs, the I '-aryl compounds exhibited either no activity or very poor activity in the standard anticancer in vivo screen asay, Le. the mouse L1210 assay. The lack of activity was postulated to arise from the steric hinderance of the phenyl and therefore the limited oxidation of the prodnig. Supponr for this postulate was the potent activity of the acyclic phenyl keto analog which exhibited activity similar to cyclophosphamide.
Cyclic esters of phosphorus-containing compounds are reported in the chemical literature, however they were not tested as prodrugs in biological systems. These cyclic esters include: di and tri esters of phosphoric acids as reported in Nifantyev et al., Phosghorus. Sulfui Si.licon and Related Eelements, 113: 1 (1996); Wijnberg et al., EP-180276
AI;
t2] phosphorus (III) acid esters. Kryuchkov et al., I. Akad. Nauk SSSR. Ser Khim. 6: 1244 (1987). Some of the compounds were claimed to be useful for the asymmnetric synthesis of L-Dopa precursors. Sylvain et atl., DE35I27B1 AI; phosphoramidates. Shih et al., Bull. Inst. Chem. Acad. Sin 41: 9 (1 9 9 4);Edmundson et al., J. Chem, Res. Svno. 5: 122 (1989); and phosphonatces. Neidlein et al., He1rocycles 35: 1185 (1993).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:29 SPRUSON FERGUSON NO. 7145 P. Numerous phosphous-containing compounds are known to exhibit pharmacological activity but remain far from optimal due to one or more of the above-described limitations.
Some of the activities described include phosphonic acids that are useful as antihypertensives and therapy for heart failure via inhibition of NEP 24.11, phosphonic acids that are useful for treating a variety of CNS conditions (stroke, epilepsy, brain and spinal cord trauma, etc.) via binding to excitory amino acid receptors NMDA receptor), bisphosphonic acids that are useful for treating osteoporosis, phosphonic acids that are useful as lipid lowering agents (e.g.
squalene synthase inhibitors), phosphonates that are useful in treating inflammation (e.g.
collagenase inhibitors), phosphonates and phosphates that are useful in treating diabetes, cancer and parasitic and viral infections.
Phosphates and phosphonates that are known to be particularly useful in glucose lowering activity and therefore are anticipated to be useful in treating diabetes are compounds that bind to the AMP site of fructose 1,6-bisphosphatase (FBPase) as described by Gruber US 5,658,889.
Other examples of phosphorus-coniaining drugs include squalene synthetase inhibitors (e.g.
BMS 188494).
A large class of drugs known to be active against hepatitis are generally nucleoside or nucleotide analogs that are phosphorylated inside cells to produce the biologically active triphospbace. Examples include Lamivudine (3TC) and Vidarabine (araA). In each case, the drug interferes with viral replication via the triphosphate form through either inhibition of DNA polymerases or DNA chain termination. Some specificity for virus-infected cells is gained by both preferential phosphorylation of the drug by virally-encoded kinases as well as by specific inhibition of viral DNA polymerases. Nevertheless, all of the nueleoside-based drugs are associated with significant non-hepatic toxicity. For example, araA frequently produces neurological toxicity with many patients showing myalgia or a sensory neuropathy with distressing pain and abnormalities in nerve conduction and a few showing tremor, dysarthria, conflision or even coma. Lok et al., J. Animicrob. Chemother n. 14: 93-99 (1984), Phosphonic acids also show antiviral activity. In some cases the compounds are anivirals themselves phosphonofonnic acid), whereas in other cases they require phosphorylation to the disphosphate, e.g. 9 -(2-phosphonylmethoxyethyl)adenine
(PMEA,
Adefovir). Frequently, these compounds are reported to exhibit enhanced activity due to either poor substrate activity of the corresponding nuclcoside with viral kinases or because the viral nucleoside kinase which is required to convert the nucleoside to the monophosphate is down COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:30 SPRUSON FERGUSON N0. 7145 P. 11 6 regulated viral resistance. Monophosphates and phosphonic acids, however, are difficult to deliver to virally-infccted cells after oral administration due to their high charge and in the case of the monophosphate instability in plasma, In addition, these compounds often have short half.
lives PMEA, Adefovir) due in most cases to high renal clearance. In some cases, the high renal clearance can lead to nephrotoxicities or be a major limitation in diseases such as diabetes where renal &tnction is often compromised.
Liver cancer is poorly treated with current therapies. In general, liver tumors are resistant to radiotherapy, respond poorly to chemotherapy and are characterized by a high degree of ceU heterogeneity. Similar compounds as those described for hepatitis are also compounds that are useful for cancer 2-Fluoroarabinosyladenosine (F-ara-A, Fludarabine), 2'2'difluorodeoxycytidine (dFdC, Gemcitabine) and 5-fluorouracil or 5-fluoro-2'-deoxy uridme.
Hepatitis and liver cancer remain poorly treated with current therapies due to doselimiting extrahepatic side effects or inadequate delivery of chemotherapeutic agents to the target tissue. Efforts to deliver drugs to the liver with relatively high organ specificity have primarily focused on strategies involving receptor mediated endocytosis (RME). RME transport systems are common to normal macrophages, hepatocytes, fibroblasts and reticulocytes.
Macromolecules internalized via RME include asialoglycoproteins, LDL, transferrin and insulin.
Another strategy for drug delivery to the liver uses colloids or liposomes both of which are subject to phagocytosis by the macrophage (Kupffer cells in liver) and localization in tissues of the reticuloendothclial system liver, spleen and bone). Of these possible approaches, most of the attention has focused on the use of glycoprotein and oligosaccharide drug conjugates as a method for organ specific delivery. Natural desialylated glycoproteins, e.g. asialoorosomucold and asialofemin, neoglycoproteins, e.g. mannosylated and lactosylared albumin, and polysacharrides such as arabinogalactan have been used to successfully deliver drugs to the liver.
Conjugates of several drug classes have been reported, including the antiviral drug araAMP. For example, araAMP conjugated to lactosaminated serum libumin was effective in treating chronic type B hepatitis without signs of neurotoxicity. Fiume et al., The Lancet 13 (1988). Because conjugation of drugs to plasma proteins may have several limitations, including uptake by scavenger receptors on non-hepatocytes, immunogenicity and instability of the protein to conjugation conditions, and in vivo metabolism, efforts have focused on the use of ollgosaccharide conjugates. One such approach uses arabinogalactan conjugate. The araAMP COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:30 SPRUSON FERGUSON NO. 7145- P. 12 7 conjugate is reported to have good activity in woodchucks carrying the hepatitis virus. Enriquez et al., Diani..Chem. 6: 195-202 (1995).
Limitations in approaches described above include dnig loading capacity, complexity of the manufacture and characterization of the conjugate, and receptor down regulation. Thus, there is still a need for prodrugs of phosphorus containing drugs.
Brief Description of the Drawinga Figure IA depicts the amount of araATP in nmoles per gram of liver found over time after i.v. administration of compound 30.1 at 10 mg/kg and 3 mg/kg and after i.v. administration of 10 ng/kg araAMP. Significantly higher liver levels of the biologically active araATP was found after administration of prodrug 30.1.
Figure IB depicts the amount of araH found in the plasma over time after i.v.
administration ofprodrug 30.1 at 10 mg/kg and 3 mg/kg and after i.v. administration of mg/kg araAMP. In contrast to an AMP, virtually no araH (atoxic metabolite) was detected in the blood after administration of prodrug 30.1.
Summary of the Inention The present invention is directed towards novel prodrugs of phosphate phosphonate, and phosphoramidate compounds, their preparation, their synthetic intermediates, and their uses. In one aspect, the invention is directed towards the use of the prodrugs to enhance oral drug delivery. Another aspect of the invention is the use of the prodrugs to treat diseases that benefit from enhanced drug distribution to the liver and like tissues and cells, including hepatitis, cancer, liver fibrosis, malaria, other viral and parasitic infections, and metabolic diseases where the liver is responsible for the overproduction of the biochemical end product, e.g. glucose (diabetes); cholesterol. fatty acids and triglycerides (hyperlipidemia) (atherosclerosis) (obesity). In another aspect, the prodrugs are used to prolong phannacodynamic half-life of the drug. In addition, the prodrug methodology of the current invention is used to achieve.sustained delivery of the parent drug. In another aspect, the prodrugs are used to increase the therapeutic index of the drug. In another aspect of the invention, a method of making these prodrugs is described. A further aspect is the novel intermediates to these prodrugs. In another aspect, the prodrngs are also useful in the delivery of diagnostic imaging agents to the liver.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:30 SPRUSON FERGUSON NO. 7145 P, 13 8 One aspect of the present invention concerns compounds that are converted in vitro or in vivo to the corresponding.phosphonic acid or phosphate monocster and are of formula I
V
W H M-P z 0 w w
I
wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z ae connected via 3-5 atoms to form a cyclic group, optionally containing I heteroarom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W arc connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, l-alkenyl, 1-alkynyl, and Z is selected from the group consisting of-CHR3OH -CHR 2 OC(O)R -CHROC(S)R', -CHR'OC(S)OR-, -CHR'OC()SWR,
-CHR'OCO
2
-OR
2
-SR
2
CHR'N
3
-CH
2 aryl, -CH(aryl)OH, -CH(CH=CR2)OH, -CH(CzCR)OH; -R 2
-NR
2 -OCOR, -OCOzR', -SCOR', -SCO z
-NHCOR
2 -NHCOR', -CHzNHaiyl, -(CHz2)-OR 2 and -(CHi),-SR; R is an R' or .H; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:31 SPRUSON FERGUSON NO. 7145- P. 14 9 RJ is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R' is selected from the group consisting of alky], aralkyl, and alicycUc; p is an integer from 2 to 3; with the provisos that: a) V, Z, W and W' are not all and b) when Z is -R 2 then at least one of V and W is not or and M is solected from the group that attached to PCO p0- or P30,' is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
The present invention provides several novel methods of making the prodrugs of the VH
V
M'-CH L-P NH L=-NR' 2 halogen present invention. One method relies on the reaction of the following novel P(IU) reagent: The resulting phosphize is then oxidized to the cyclic phosphate ester.
A second method relies on the reaction of a novel P(V) reagent: N L'-NR2, ayl, halogen Ni L'=-NR' 2 -0-aryl, halogen COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:31 SPRUSON FERGUSON NO. 7145 P. A third method relies on reacting another novel P(V) compound with a diol: -G OH
OH
0 Ldiol
I
NH, fH L" halogen Since chase compounds have asymmetric centers, the present invention is directed not only to racemic and diastereomeric mixtures of these compounds, but also to individual stereoisomers. The present invention also includes pharmaceutically acceptable and/or useful salts of the compounds of formula I, including acid addition salts. The present inventions also encompass prodrugs of compounds of formula I.
Definito M In accordance with the present invention and as used herein, the following Ctrms are defined with the following meanings, unless explicitly stated otherwise.
The term "aryl" refers to aromatic groups which have 5-14 ring atoms and at least one ring having a conjugated pi electron system and includes carbocyclic aryl, heterocyclic aryl and biaryl groups, all of which may be optionally substituted. Suitable aryl groups include phenyl and Carbocyclic aryl groups are groups wherein the ring atoms on the aromatic ring are carbon atoms. Carbocyclic aryl groups include monocycUc carbocyclic aryl groups and polycyclic or fused compounds such as optionally substituted naphthyl groups.
Heterocyclic aryl groups are groups having from 1 to 4 hetoroatoms as ring atoms in the aromatic ring and the remainder of the ring atomr being carbon atoms. Suitable heteroatoms include oxygen, sulfur, and nitrogen. Suitable beteroaryl groups include furanyl, thienyl, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:31 SPRUSON FERGUSON NO, 7145 P. 16 11 pyridyl, pyrrolyl, N-lower alkyl pyrrolyl, pyridyl-N-oxide, pyrimidyl, pyrazinyl, imidazolyl, and the like, all optionally substituted.
The term "biaryl" represents aryl groups containing more than one aromatic ring including both fused ring systems and aryl groups substituted with other aryl groups. Such groups may be optionally substituted. Suitable biaryl groups include naphthyl and biphenyl.
The term "alicyclic" means compounds which combine the properties of aliphatic and cyclic compounds and include but are not limited to aromatic, cycloalkyl and bridged cycloalkyl compounds. The cyclic compound Includes heterocycles. Cyclohexenylethyl and cyclobexylethyl are suitable alicyclic groups. Such groups may be optionally substituted.
The term "optionally substituted" or "substituted" includes groups substituted by one to four substituents, independently selected from lower alkyl, lower aryl, lower aralkyl, lower alicyclic, hydroxy, lower alkoxy, lower aryloxy, pcrhaloalkoxy, aralkoxy, hoeeroaryl, heteroaryloxy, heceroarylalkyl, hcteroaralkoxy, azido. amino, guanidino. halo, lower alkylthio, oxo, acylalkyl, carboxy esters, carboxyl, carboxamido, nitro, acyloxy, aminoalkyl, alkylamrinoaryl, alkylaryl, alkylaminoalkyl, alkoxyaryl, arylamino, aralkylamino, phosphono, sulfonvi. carboxamidoalkylaryl. carboxamidoaryl. hydroxyalkyl. haloalkyl.
alkylaminoalkylcarboxy, aminocarboxamidoalkyl, cyano, lower alkoxyalkyl, lower perhaloailcyl, and arylalkyloxyalkyl.
The term "aralkyl" refers to an alkyl group substituted with an aryl group. Suitable aralkyl groups include benzyl, picolyl, and the like, and may be optionally substituted. The term "-aralkyl-" refers to a divalent group -aryl-alkylene-.
The term "-alkylaryl." refers to the group -alk-aryl- where "alk" is an alkylene group.
"Lower -alkylaryl-" refers to such groups where alkylene is lower alkylene.
The term "lower" referred to herein in connection with organic radicals or compounds respectively defines such as with up to and including 10, preferably up to and including 6, and advantageously one to four carbon atoms. Such groups may be straight chain, branchcd, or cyclic.
The terms "arylamino" and "aralkylamino" respectively, refer to the group -NRR' wherein respectively, R is a yl and R' is hydrogen, alkyl, aralkyl or aryl, and R is aralkyl and R' is hydrogen or aralkyl, aryl, alkyl.
The term "acyl" refers to -C(O)R where R is alkyt and aryl.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:32 SPRUSON FERGUSON NO. 7145 P. 17 12 The term "carboxy esters" refers to -C(O)OR where R is alkyl, ayl, aralkyl, and alicyclic, all optionally substituted.
The term "carboxyl" refers to -C(O)OH.
The term "oxo" refers to -0 in an alkyl group.
The tenrm "amino" refers to -NRR' where R and R' are independently selected from hydrogen, alkyl, aryl, aralkyl and alicyclic, all except H are optionally substituted; and R and R' can form a cyclic ring system.
The term "carbonylamino" and "-carbonylamiffo-" refers to RCONR. and -CONR., respectively, where each R is independently hydrogen or alkyl.
The tenri "halogen" or "halo" refers to -CI, -Br and -I.
The term "-oxyalkylamino-" refers to -0-alk-NR-, where "slk" is an alkylene group and R is H or alkyl.
The term "-alkylaminoalkylcarboxy-" refers to the group -alk-NR-alk-C(0)-O- where "ali" is an alkylene group, and R is a H or lower alkyl.
The term "-alkylaminocarbonyl-" refers to the group -alk-.NR-C(O)- where "alk" is an alkylene group, and R is a H or lower alkyl.
The tenrm "-oxyalkyl-" refers to the group -0-alk- where "alk" is an alkylene group.
The term "-alkylcarboxyalkyl." refers to the group -alk-C(O)-O-alk- where each alk is independently an alkylene group.
The term "alkyl" refers to saturated aliphatic groups including straight-chain, branched chain and cyclic groups. Alkyl groups may be optionally substituted. Suitable alkyl groups include methyl, isopropyl, and cyclopropyl.
The term "cyclic alkyl" or "cycloalkyl" refers to alkyl groups that are cyclic. Suitable cyclic groups include norbornyl and cyclopropyl. Such groups may be substituted.
The term "heterocyclic" and "heterocyclic alkyl" refer to cyclic groups containing at least one heteroatom. Suitable heteroatoms include oxygen, sulfutr, and nitrogen. Heterocyclic groups may be attached through a nitrogen or through a carbon atom in the ring. Suitable heterocyclic groups include pyrrolidinyl, morpholino, morpholinoethyl, and pyridyl.
The term "phosphono" refers to -PO3Rz, where R is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic.
The term "sulphonyl" or "sulfonyl" refers to -SO 3 R, where R is H, alkyl, aryl, aralkyl, and alicyclic.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:32 SPRUSON FERGUSON NO, 7145 P. 18 13 The term "alkenyl" refers to unsaturatcd groups which contain at least one carbon-carbon double bond and includes straight-chain, branched-chain and cyclic groups. Alkenyl groups matbe optionally substituted. Suitable alkenyl groups include allyl.
The term "alkynyl" refers to unsaturated groups which contain at. least one carbon-carbon triple bond and includes straight-chain, branched-chain and cyclic groups, Alkynyl groups may be optionally substituted. Suitable alkynyl groups include ethynyl.
The term "alkylene" refers to a divalent straight chain, branched chain or cyclic saturated aliphatic group.
The term "acyloxy" refers to the ester group where R is H, alkyl, alkenyl, alkynyl, aryl, aralkyl, or alicyclic.
The term "aminoalkyl-" refers to the group NR 2 -alk- wherein "alk" is an alkylene group and R is selected from H, alkyl, aryl, aralkyl, and alicyclic.
The term "-alkyl(hydroxy)-" refers to an -OH offthe alkyl chain. When this term is an X group, the -OH is at the position a to the phosphorus atom.
The termnn "alkylaminoalkyl-" refers to the group alkyl-NR-alk- wherein each "alk" is an independently selected alkylene, and R is H or lower alkyl. "Lower alkylaminoalkyl-" refers to groups where each alkylene group is lower alkylene.
The term "arylaminoalkyl-" refers to the group aryl.NR-alk- wherein "alk" is an alkylene group and R is H, alkyl, aryl, aralkyl, and alicyclic. In "lower arylaminoalkyl-", the alkylene group is lower alkylene.
The term "alkylaminoaryl-" refers to the group alkyl-NR.aryl- wherein "aryl" is a divalent group and R is H, alkyl, aralkyl, and alicyclic. In "lower alkylaminoaryl.", the alkylene group is lower alkyl.
The term "alkyloxyaryl-" refers to an aryl group substituted with an alkyloxy group. In "lower alkyloxyaryl-", the alkyl group is lower alkyl.
The term "aryloxyalkyl-" refers to an alkyl group substituted with an aryloxy group.
The term "aralkyloxyalkyl[-" refers to the group aryl-alk-O-alk- wherein "alk" is an alkylene group. "Lower aralkyloxyalkyl-" refers to such groups where the alkylene groups are lower alkylene.
The term "-alkoxy." or "-alkyloxy." refers to the group -alk-O- wherein "alk" is an alkylene group.
The term "alkoxy-" refers to the group alkyl-O-.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:32 SPRUSON FERGUSON NO. 7145 P. 19 14 The term "-alkoxyalkyl." or "-alkyloxyalkylj" refer to the group -alk-O-alk. wherein each "alk" is an independently selected alkylene group. In "lower -alkoxyalkyl-", each alkylend is lower alkylene.
The terms "alkylthio-" and "-alkylthio-" refer to the groups alkyl.S-, and -alk-S., respectively, wherein "alk" is alkylene group.
The term "-alkylthioalkyl." refers to the group -alk-S-alk- wherein each "alk" is an independently selected alkylene group. In "lower -alkylthioalkyl-" each alkylene is lower alkylene.
The terms "amido" or "carboxamido" refer to NR 2 and RC(0)-NR-, where R and R4 include H, alkyl, aryl, aralkyl, and alicyclic. The term does not include urea, -NR-C(O).NR- The term "-alkylcarboxamido-" or "-alkylcarbonylamino-" refers to the group -alkwherein "alk" is an alkylene group and R is H or lower alkyl.
The term "-alkylaminocarbonyl-" refers to the group ,alk-NR.C(O)- wherein "alk" is an alkylene group and R is H or lower alkyl.
The term "aminocarboxamidoalkyl-" refers to the group NR2-C(0)-N(R).alk. wherein R is an alkyl group or H and "alk" is an alkylcne group. "Lower aminocarboxamidoalkyl." refers to such groups wherein "alk" is lower alkylene.
The term "heteroarylalkyl"r' refers to an alkyl group substituted with a heteroaryl group.
The term -dihaloalkyl-" refers to an X group where the I position and therefore halogens are ct to the phosphorus atom.
The term "perhalo" refers to groups wherein every C-H bond has been replaced with a C-halo bond on an aliphatic or aryl group. Suitable perhaloalkyl groups include -CF3 and -CFC1 2 The term "guanidino" refers to both -NR-C(NR)-NR 2 as well as -N-C(NRz) 2 where each R group is independently selected from the group of alkyl, alkenyl, alkynyl, aryl, and alicyclic, all except -H are optionally substituted.
The term "amidino" refers to -C(NR)-NR 2 where each R group is independently selected from the group of-H, alkyl, alkenyl, alkynyl, aryl, and alicyclic, all except -H are optionally substituted.
The term "pharmaceutically acceptable salt" includes salts of compounds of formula I and its prodrugs derived from the combination of a compound of this invention and an organic or inorganic acid or base.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:33 SPRUSON FERGUSON NO. 7145 P. The term "prodrug" as used herein refers to any compound that,when administered to a biological system Senerates a biologically active phosph(on)ate compound may be further phosphoylated to produce a biologically active compound either as a result of spontaneous chemical reaction(s) or by enzyme catalyzed or metabolic reaction(s). Standard prodrugs are formed using groups attached to functionality, e.g. HO-, HS-, HOOC-,
R
2 associated with the drug, that cleave in vivo. Standard prodrugs include but are not limited to carboxylate esters where the group is alkyl, aryl, aralkyl, acyloxyalkyl, alkoxycarbonyloxyalkyl as well as esters of hydroxyl, thiol and amineswhere the group attached is an acyl group, an alkoxycarbonyl, aminocarbonyl, phosphate or sulfate. The groups illustrated 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 are not biologically active themselves, but rather must undergo a chemical transformation to produce the compound that is biologically activite or is a precursor of the biologically active compound. The biologically active compounds include, for example, anticancer agents, antiviral agents, and diagnostic imaging agents.
The sructure
V
w has a plane of symmety running through the phosphorus-oxygen double bond when V-sW, and V and W are either both pointing up or both pointing down.
The term "bidentare" refers to an alkyl group that is attached by its terminal ends to the same atom to form a cyclic group. For example, propylene imine contains a bidentate propylene group.
The term "cyclic 1',3'-propane ester", "cyclic 1,3-propane ester", "cyclic l',3'-propanyl ester", and "cyclic 1,3-propanyl ester" refers to the following: 3' COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 17:33 SPRUSON FERGUSON N0. 7145 P. 21 16 The phrase "together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing one heteroatom, that is fused to an aryl group attached at the beta and gamma position to the oxygen attached to the phosphorus" includes the following: w* The term "phosph(oramid)ite" refers to phosphoramidites and phosphites which are compounds attached via 0 or N to including cyclic forms.
The term "phosph(on)ate" refer to compounds attached via C, 0, or N to PO3'.
The term "phosphonate" refers to -C-PO 3 2 and its acids.
The term "phosphate" refers to and its acids.
The term "phosphoramidate" refers to -N-PO1', and its acids.
X group nomenclature as used herein in formulae II-V describes the group attached to the phosphonate and ends with the group attached to the heteroaromatic ring. For example, when X is alkylamino, the following structure is intended: (heteroaromatic ring)-NR-alk-P(0)(OR) 2 Likewise. Y, A, B, C, and D groups and other substituents of the hctcroaromatic ring are described in such a way that the term ends with the group attached to the heteroaromatic ring.
Generally, substituents are named such that the term ends with the group at the point of attachment.
The term "nucleoside" refers to a purine or pyrimidine base, including analogs thereof connected to a sugar, including heterocyclic and carbocyclic analogs thereof.
The term "liver" refers to liver and to like tissues and cells that conmain the CYP3A4 isozyme or any other P450 isozyme found to oxidize the phosphonate esters of the invention.
Based on Example F, we have found that prodrugs of formula VI and VIII are selectively COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:33 SPRUSON FERGUSON NO. 7145 P. 22 17 oxidized by the cytochrome P450 isoenzyme CYP3A4. According to DeWaziers Ct al Pharm.
Exp. Ther., 253, 387-394 (1990)), CYP3A4 is located in humans in the following tissues (determined by immunoblotting and enzyme measurements): TiMeS of liver activity Liver 100 Duodenum jejunum ileum 4 colon <5 (only P450 isoenzyme found) stomach esophagus kidney not detectable Thus, "liver" more preferably refers to the liver, duodenum, jejunum, ileum, colon, stomach, and esophagus. Most preferably, liver refers to the liver organ.
The term "enhancing" refers to increasing or improving a specific property.
The term "liver specificity" refers the ratio: fdrug or a drug metabolite in liver tissue [drug or a drug metabolite in blood or another tissue] as measured in animals treated with the drug or a prodrug. The drug metabolite measured in the liver may or may not be the same metabolite measured in the other tissue. For example, in studies comparing araA and prodrugs of araA. the ratio was determined by measuring the liver metabolite araATP and blood metabolite aran. The ratio can be determined by measuring tissue levels at a specific time or may represent an AUC based on values measured at three or more time points.
The term "increased or enhanced liver specificity" refers to an increase in the liver specificity ratio in animals treated with therprodrug relative to animals treated with the parent drug.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:33 SPRUSON FERGUSON NO. 7145 P. 23 18 The term "enhanced oral bioavailability" refers to an increase of at least 50% of the absorption of the dose of the parent drug or prodrug(not of this invention) from the gastrointestihal tract. More preferably it is at least 100%. Measurement of oral bioavailability usually refers to measurements of the prodrug, drug. or drug metabolite in blood, tissues, or urine following oral administration compared to measurements following systemic administration.
The term "parent drug" refers to any compound which delivers the same biologically active compound. This would include standard prodrugs, such as esters. This would also include drugs such as AZT which can be thought of as a parent drug in the form of MH. in the body AZT is first phosphorylated to AZT-PO 2 and then further phosphorylated to form AZTtriphosphate, which is the biologically active form. The parent drug form MH only applies when M is attached via N or O. Preferably, the parent drug is M-PO, 2 or M-H. More preferred is M-POj 2 The term "drug metabolite" refers to any compound produced in vive or in vitro from the parent drug, which can include the biologically active drug.
The term "pharmacodynamic half-life" refers to the time after administration of the drug or prodrug to observe a diminution of one half of the measured pharmacological response.
Pharmacodynamic half-life is enhanced when the half-life is increased by preferably at least The term "pharmacokinetic half-life" refers to the time after administration of the drug or prodrug to observe a dimunition of one half of the drug concentration in plasma or tissues.
The term "therapeutic index" refers to the ratio of the dose of a drug or prodrug that produces theraputically beneficial response relative to the dose that produces an undesired COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:34 SPRUSON FERGUSON NO. 7145 P. 24 19 response such as death, an elevation of markers that are indicative of toxicity, and pharmacological side effects.
The term "sustained delivery" refers to an increase in the period in which there is adequate blood levels of the biologically active drug to have a therapeutic effect.
The term "bypassing drug resistance" refers to the loss or partial loss of therapeutic effectiveness of a drug (drug resistance) due to changes in the biochemical pathways and cellular
L
activities important for producing and maintaining the biologically active form of the drug at the desired site in the body and to the ability of an agent to bypass this resistance through the use of alternative pathways and cellular activities.
The term "biologically active drug or agent" refers to the chemical entity that produce a biological effect. Thus, active drugs or agents include compounds which as the free phosphonate or phosphate are biologically active, or which must undergo further phosphorylation to be biologically active. It does not include MH.
The term "therapeutically effective amount" refers to an amount that has any beneficial effect in treating a disease or condition.
The following well known drugs are referred to in the specification and the claims.
Abbreviations and common names are also provided.
araA; 9-b-D-arabinofuranosyladcnine (Vidarabine) AZT; 3'-azido-2',3'-dideoxythymdine (Zidovudine) d4T; 2',3'-didehydro-3 'deoxythymidine (Stavudine) ddl; 2',3'-dideoxyinosine (Didanosine) ddA; 2',3'-dideoxyadenosine ddC; 2 3 '-dideoxycytidine (Zalcitabine) L-ddC; L-2',3'-dideoxycytidine L-FddC; L-2*,3'-dideoxy-5-fluorocytidine L-d4C; L-3'-deoxy-2',3'-didehydrocytidinc L-Fd4C; L-3'-deoxy-2',3'-didehydro-5-fluorocytidine 3TC; (-)-2',3'-dideoxy-3'.thiacytidine (Lamivudine) l-b-D-ribofiranosyl-l,2,4-triazole-3-carboxamide (Ribavirin) FIAU; 1-(2-deoxy-2-fluoro-b-D-arabinofuranosyl)-5-iodouridine FIAC; 1-( 2 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:34 20. EB. 004 7:34 SPRUSON FERGUSON N.74 .2 NO, 7145 P. BHCG; a,2b 1 3a)9 ,-i~hdoyohl~ylbtl guanine 2'RS t 1 4 2 -(hYdroxyrchy)oxawolal..s.yloytosie~ -didcoxycyfldinc (Zalcitabiuc) FMAU. 2 BvanU; I-b-D-arbinofuifnosyI.ES5(2-bmmovinyI)waci1 (Sodivudine) E-$-(2-broniovinyl).2' -deoxyuxidine Cobucavir ThT, Trifluorothyrnidine (Trifluorothyznidinc) 5-propynyl- 1-arabinosyluracjl (Zonavir) CDG; curbocycli4,2'-dcoxyguanosjpe DAPD; (-)-B-D-2,6-dianduopuririe dioxolane FDOC: 5-fluoro- i-[2-Qtydroxymty>-1 3 -dioxoisnolcytosine d4C; 3 -deoxy-2'4 '-didehydrocydidine DXG; dioxolane guanosine FEA(J; 2'-dcoxy-2 '-fluoro-1 -b-fl-arahinofinnosyl-5cjjhylura 0 j Ff43; 2 -dideoxy-3 -fluoroguanosizic FLT; 3 '-deoxy-3'-fluorotbymidine FTC; .S-fluoro- 1-[2-(hydroxymetbyl)-l 1 3 -oxathiolan-s-yijcytosine 5-yI-carbocyclic 2'-deoxyguanosine (BMS200,475) [1-(4'-hyciroxy- 1',2'-butadienyi)cyrosineI (Cytallene) Oxetanocin A; 9-2doy2hdoyehlbw-rtr-xtmy~dnn Oxetmnocin 0; 9-2doy2hdoyehlbtaDcyr-oeaoy~unn Cyclobut A; beta,2 alpha,3 bd)23bshdoyehl) ccouy~dnn Cyclobut 0; bcta.,2 alpha3 bea-,-i~ymynty)iccauy~un~L (Lobucavir) 5'-fluoro-T2-deoxywiaine d~dC;, 2 2 '.difluorodeoxycytidine (Qomcitabine) =mC; arabinosyicytosine (Cytarabine) brorodcoxyuridlne IDU; 5-iodo-2 '-deoxyuridinc (Idoxuridiuc) CdA; 2 -cblorodeoxyudenosine (Cladxibinc) P-ara-A; fluoroamabinosyladcnosiuc (Pludarabine) ACV; 9 2 -hydroxyethoxyhnerhyj)guanjne (Acyclovir) GCV; 9-(1I, 3 -dhydroxy-2-propoxymethyz)gii*,ine (gangcyciovir) 9 <(4.hydroxy.3-hytroxymcthybut.. -yl)gumine (Penciclovir) (R)-9-(3,4-dibydroxybutyI)gSuaninc (Buviciovir) phosphonofornilic acid (Foscaruct) PA; pbosphonoacedc acid PMBA; 9 2 -pbosphonylmezhoxyezhyl) adeninc (Adctovir) PMEDAP; 9-2popoyntoyty)26dai~rn HPMPC;- (S)-9-(3-hydroxy-2phosphony~nmtoxypwpy) cytosine (Cidotovir) 1{PMPA; 9 -(3-hydroxy2-phosponymexpmpyj) adenine FPM.PA; 9 -03-fluoro-2-phosphonynetloxypyopyl) adenine FMPA; (R) 9 -(2,phosphonyznczhoxypropyl) adenine COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:.45 Date 2004-02-20 FEB, 2004 17:34 SPRUSON FERGUSON NO. 7145 P. 26 21 Detafiled Desription of tbe_ ventio The invention is directed to the use of new cyclic phosph(on)ate ester methodology which allows compounds, to be efficiently converted to phospb(on)ate containing compounds by p450 enzymes found in large amounts in the liver and other tissues containing these specific enzymes. This methodology can be applied to various drugs and to diagnostic imaging agents.
More specifically, the invention is directed to the use of prodrug-esters of highly cbarged phosphate, phosphoramidate, and phosphonate containing drugs that undergo non-esterasemediated hydrolysis reactions to produce the phosphate, phosphoramidate, and phosph(on)ate containing compounds. Because highly charged phosph(on)ate containing compounds are not readily absorbed in the gasrointestinal tract, this prodrug methodology can be used to enhance absorption of the active compound after oral administration.
In another aspect of the invention, this prodrug methodology can also be used to prolong the pharmaoodynamic half-life of phosph(on)at-.containing drungs because the cyclic phosph(on)ates of the invention can prevent the action ofenzymes which degrade the parent drug.
In another aspect of the invention, this prodrug methodology can be used to achieve sustained delivery of the parent drug because various novel prodrugs are slowly oxidized in the liver at different rates, The novel cyclic phosphonate methodology of the present invention may also be used to increase the distribution of a particular drug or imaging agent to the liver which contains abundant amounts of the p 4 50 isozymes responsible for oxidizing the cylic phosphonate of the present invention so that the free phosphonate or phosphate is ultimately produced. Accordingly, this prodrug technology should prove useful in the treatment of liver diseases or diseases where the liver is responsible for the overproduction of the biochemical end product such a glucose, cholesterol, fatty acids and triglycerides. Such diseases include viral and parasitic infections, liver cancer, liver fibrosis, diabetes, hyperlipidemia. and obesity. In addition the liver specificity of the prodrugs should also prove useful in the delivery of diagnostic agents to the liver, These specific p450 enzymes are also found in other specific Tissues and cells, and thus this methodology may also be used to increase the delivery of these agents to those dtissues.
In another aspect of the invention, the characteristic that most of the cyclic phosph(on)ates of the present invention are metabolized in the liver to produce the phosph(on)ate drug can enable the use of the prodrug methodology of the present invention to increase the COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:35 SPRUSON FERGUSON NO, 7145 P. 27 22 therapeutic index of various drugs which tend to have side effects related to the amount of the drug or its metabolizes which are distributed in extrahepatic tissues.
In yet another aspect of the invention, the cyclic phosph(on)ate esters of the present invention can bypass drug resistance caused by decreased transport into the target cells, increased drug export by transporters, increased drug metabolism, or decreased precursor metabolism to the active drug.
In another aspect of the invention, novel phosphire and phosphonate intermediates are described. C In another aspect, methods of preparing the cyclic phosph(on)ate prodrugs arc described.
Theses aspects are described in greater detail below.
Enhancing Oral Bioavailablty The invention pertains to certain cyclic 1 3 '-propanyl esters of phosph(on)ates and the use of these esters to deliver, most preferably via oral administration, a therapeutically effective amount of the corresponding phosph(on)ate compounds, preferably to an animal in need thereof The active drug may be M-POt". Alternatively, M-POz- may instead undergo further phosphorylation by kinases to form M-P 2 06 3 and/or M-PO 4 as the active drug substance.
Compounds containing a free phosphonic acid or a phosphoric acid group generally exhibit poor oral bioavailability since these groups are highly charged at physiological pH. Charged groups on compounds with molecular weights greater than 250 Daltons impede passive diffusion across cell membranes as well as absorption across the gut epithelial cell layer.
Neutral prodrugs of these compounds have therefore been studied since these compounds would be more lipophilic and therefore more likely to exhibit improved intestinal permeability Although many prodrug classes have been reported, few have been found that exhibit properties suitable for drug development.
The most common prodrug class, and the class almost exclusively used for clinical candidates, is the acyloxyalkyl esters. These prodrugs, however, often exhibit only a modest improvement in oral bioavailability due to poor aqueous stability, poor stability to acidic/basic pH and rapid degradation by esterases in the gastrointestinal tract (Shaw Cundy, Pharm. Ra.
10, (Suppl), S294 (1993). Another class of prodrugs are the bis-aryl prodrugs DeLombert et al. J. Mad Chem. 37, 498 (1994)) which have shown in a few isolated cases to provide good to modest improvements in oral bioavailability. The major limitation with this class of compounds COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:35 SPRUSON FERGUSON NO. 7145 P, 28 23 is that the prodrug ester often is degraded to the monoacid rapidly in-xjyv but conversion to the parent drug occurs only slowly (sometimes over days) if at all.
The prodrugs of the invention exhibit improved properties that lead to enhanced oralbioavailability relative to the parent drug. Several characteristics of the present cyclic phosph(on)ate prodrugs may contibute to their ability to enhance oral bioavailabihy. First, the prodrugs exhibit good stability in aqueous solutions across a wide range of pHs. In Example A, 30.1 was found to be stable for at least seven days in 100 mM potassium phosphate buffer solutions at pH 3, 7, and 9. This pH stability prevents immediate hydrolysis in the mouth and GI tract prior to absorption. The pH stability can also be. beneficial during formulation of the product Second, the prodrugs are resistant to esterases and phosphatases which are abundant in the gastrointestinal tract. The resistance to esterases and phosphatases can be assayed according to Example B. In addition, Example C demonstrated that 30.1, 1.1, and 1,2 were not degraded by esterases found in fresh rat plasma. Because much of the administered dose remains intact in the G.I. tract, the compound remains less highly charged than a free phosphonate which means more of the drug can be absorbed by passive diffusion and enter the blood stream, Last, the prodrug can limit metabolism at other sites on the molecule. For example, the prodrugs of the invention eliminate metabolism of the purine base of araA by adenosine deaminase which is also abundant in the GI tract. In Example C, the cyclic 1 '-(4-pyridyl)-3' propanyl phosphate ester prodrug of araA was not susceptible to deamination by adenosine deaminase found in rat plasma. The amine of araA which is normally deaminated by the enzyme is protected by the cyclic phosphate moiety. Reduced metabolism at other sites of the molecule enables more of the drug to circulate in the blood stream. Although not all of these properties will be applicable to every prodrug of every drug, each of these properties can enable more drug to survive the GI tract and be available for absorpdioi.
The novel prodrug strategy of the invention will be useful for the oral delivery of drugs that act in the liver as well as certain drugs that act on targets located in the vascular system or extrahepatic tissues. Because the highest concentration of CYP3A4 (the enzyme responsible for activating the novel prodrugs is in the liver, the biologically active drug has a high concentration in the liver, relative to other tissues, In one aspect, parent drugs which act in the liver are preferred.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB-2004 17:35 SPRUSON FERGUSON NO, 7145 P. 29 24 However, some of the phosph(on)ates are exported by organic anion transporters in the liver and enter the blood stream. Many phosph(on)ates in the blood stream are cleared quickly by the kidneys. Examples of such compounds are the FBPase inhibitors described herein and PMEA. Such compounds probably will not reach therapeutic levels in extrahepatic issues.
However, there are some phosph(on)ates and phosphates that are able to remain in circulation because they are not rapidly cleared by the kidneys NEP inhibitors). Such compounds are able to achieve therapeutically effective levels in blood and extrahepatic tissues. Thus, in another aspect, oral delivery to extrahepatic tissues of phosph(on)ates which are not cleared by the kidneys is preferred. Thus, such parent drugs that act at sites accessible to the free phosph(on)ic acid such as targets within the vasculature system, or enzyme or receptor targets that are located on cell membranes which are exposed to the blood or fluid in the intrastitial space are preferred. Targets suitable for this aspect of the invention would be targets in which the phosphonic acid administered parenterally via i.v. injection) produces a pharmacological or biochemical response expected to be useful for treating a disease condition.
For example, phosph(on)ic acids that inhibit neutral endopeptidase 24.11 ("NEP inhibitors") are known to inhibit the degradation of atrial natriuretic factor in vive and to produce an associated antihypertensive and diuretic effect (DeLambert et al., J. Med. Chem. 37, 498 (1994)) which may be useful for the treatment of hypertension and congestive heart failure.
Since the inhibitors exhibit poor oral bioavailability prodrugs of the type described in this invention could enhance the oral bioavailability and produce the phosphonic acid following prodrug cleavage in the liver. Suitable circulating drug levels are expected after prodrug cleavage in the liver, since the liver is known to excrete phsphonic acids into the circulation.
For example, phosphonic acids that inhibit FSPase are exported out of hepacocytes intvin presumably by an organic anion transporter.
Oral bioavailability can also be calculated by comparing the area under the curve of prodrug, drug. and/or metabolite concentration over time in plasma, liver, or other tissue or fluid of interest following oral and i.v. administration. In Example M. prodrug 30.1 demonstrated an oral bioavailability of 17.4% by analysis of hepatic levels of the phosphorylated parent compound, ara-ATP, following oral and i.v. administration.
Oral bioavailability can also be measured by comparing the amount of the parent compound exreted in the urine, for example, after oral and i.v. administration of the prodrug. A lower limit of oral bioavailability can be estimated by comparison with the amount of parent COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:36 SPRUSON FERGUSON NO. 7145 P. drug excreted in the urine after administration of the i.v. parent drug. Analysis of prodmugs of FBPase inhibitors in Example M shows that these compounds exhibit improved oral bioavailability across a wide specunum of prodrugs, with many showing a 2.5-25-fold increase in oral bioavailability.
Preferably, oral bioavailability is enhanced by at least 50% compared to the parent drug.
More preferably, oral bioavailability is enhanced by 100%.
Sustained Delivery Drugs that undergo rapid elimination iLiv often require multiple administrations of the drug to achieve therapeutically-effective blood levels over a significant period of time. Other methods are also available including sustained release formulations and devices. Prodrugs that breakdown over time can also provide a method for achieving sustained drug levels. In general, this property has been not been possible with the known phosph(on)ate prodrugs since either they undergo rapid hydrolysis inLyi acyloxyalkyl esters) or very slow conversion diayl prodrugs).
The cyclic phosph(on)ates of the invention are capable of providing sustained drug release by providing a steady release of the drug over time. For example, most phosphates undergo dephosphorylation inj dv within minutes after systemic administration via the action of phosphatases present in the blood. Similarly, acyloxyalkyl esters of these phosphates undergo rapid esterase-mediazed hydrolysis to the phosphate which then is rapidly dephosphorylated.
Some prodrugs of the current invention may enable prolonged drug delivery since many of the present prodrugs are oxidized slowly over time to the phosph(on)ate in the livers.
Sustained delivery of the drugs is achievable by selecting the prodrugs of formula I that are hydrolyzed iliuva at a raLe capable of achieving therapeutically effective drug levels over a period of time. The cleavage rate of the drug may depend on a variety of factors, including the rate of the p450 oxidation, which is dependent on both the substituents on the prodrug moiety, the stereochemistry of these substituents and the drug itself. Moreover, sustained drug production will depend on the rate of elimination of the intermediate generated after oxidation and the rate and availability of the prodrug to the liver, which is the major site of oxidation.
Identification of the prodrug with the desired properties is readily achieved by screening the prodrugs in an assay that monitors the rate of drug production in the presence of the major p450 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20
FEB.
r^AAAl 4 iU 1I: o SPKUUN LFEKUSON NO. 7145 P. 31 26 enzyme involved in the metabolism, in the presence of liver microsomes or in the presence of hepatocytes. These assays are illustrated in Examples G, D, and E, and I, respectively.
It is contemplated that prodrugs of the present invention could be combined to include, for example, one prodrug which produces the active agent rapidly to achieve a therapeutic level quickly, and another prodrug which would release the active agent more slowly over time, Examples of drugs with different rates of cleavage are shown in Example S. As indicated in this example, the rate of a drug release depends on the prodrug stercochemistry.
L
Improved Pharmacodvnamic Half-Life The pannrmacodynamic half-life of a drug can be extended by the novel prodrug methodology as a result of both its ability to produce drug over a sustained period and in some cases the longer phannacokinetic half-life of the prodrug. Both properties can individually enable therapeutic drug levels to be maintained over an extended period resulting in an improvement in the pharmacodynamic half.life. The pharmacodynamic half-life can be extended by impeding the metabolism or elimination pathways followed by the parent drug. For some drugs, the prodrugs of the present invention are able to impede the metabolism or elimination pathways followed by the parent drug and thereby exist for extended periods in an animal.
An example of the ability of the prodrug class to impede metabolic pathways associated with the parent drug is shown by the araAMP prodrug In comparison to araAMP, 30.1 shows no ara-hypoxanthine ("araH") which is the known metabolic byproduct of araA produced in e.g. plasma and the gastrointestinal tract after oral or Lv/administration (Example 0).
AraAMP on the other hand is rapidly and nearly completely converted to araH, which is produced by first dephosphorylation to araA via phosphatases followed by deamination of the base via adenosine deaminase. The prodrug moiety prevents both dephosphorylation and deamination from occurring, as shown in Examples B and C.
A common route of elimination of phosph(on)ate drugs is via the kidneys and a transporter that recognizes anionic compounds. Complete elimination of phosphonate and phosphate containing drugs from the circulation often occurs only minutes after drug administration PMEA). The prodrugs of this Invention slow the elimination of the drug by removing the negative charge until after oxidation and hydrolysis in liver and like tissues.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:37 SPRUSON FERGUSON NO, 7145 P. 32 27 The prodrug of PMEA 28.4 results in high PMEA disphosphate levels in the liver.
Moreover, minor amounts of the parent drug is climinated via the kidneys (Examples 0 and In contrast, PMEA the bis POM prodrug of PMEA result in high levels of PNEA in the urine.
Thus, prodrugs of the invention can improve the pharmacodynamic half-life by reducing the amount eliminated by the kidneys.
Enhanced Selective Delivery of Agents to the Liver and Like Tissue Delivery of a drug to the liver with high selectivity is desirable in order to treat liver diseases or diseases associated with the abnormal liver properties diabetes, hyperlipidemia) with minimal side effects. Efforts to deliver drugs to the liver with relatively high organ specificity have primarily focused on strategies involving receptor mediated endocytosis
(RME).
RME transport systems are common to normal macrophages, hepatocytes, fibroblasts and reiculocytes [Wileman et al., Biochem. J. 232, 1-14 (1985)]. Macromolccules internalized via RME include asialoglycoproteins, LDL, transferrin and insulin. Another strategy for drug delivery to the liver uses colloids or liposomes both of which are subject to phagocytosis by the macrophage (Kupffer cells in liver) and localized in tissues of the reticuloendothelial system liver, spleen and bone), Of these possible approaches, most of the atention has focused on the use ofglycoprotein and oligosaccharide drug conjugates as a method for organ specific delivery [Meijer, D.KJ.. and van der Sluijs, P. Pharm. Res., 6 105-118 (1989)]. Natural desialylated glycoproteins, e.g. asialoorosomucoid and asialofetuin, and neoglycoproteins, e.g.
mannosylated and lactosylated albumin, and polysacharrides such as arabinogalactan have been used to successfully deliver drugs to the liver.
Conjugates of several drug classes have been reported, including the antiviral drug araAMP. For example, araA-MP conjugated to lactosaminated serum albumin was effective in creating chronic type B hepatitis without signs of neurotoxicity [Fiume et al., The Lancer 13 (19S8)J. Because conjugation of drugs to plasma proteins may have several limitations, including uptake by scavenger receptors on non-hepatocytes, immunogenicity, instability of the protein to conjugation conditions, and in vive metabolism, efforts have focused on the use of oligosaccharide conjugates. One promising approach uses arabinogalactan conjugates. The araAMP conjugate is reported to have good activity in woodchucks carrying the hepatitis virus Enriquez, Jung, Josephson, L. Bioeonj. Chem. 6, 195-202 (1995)]. Limitations in COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:37 SPRUSON FERGUSON NO. 7145 P. 33 28 approaches described above include drug loading capacity, complexity of the manufacture and characterization of the conjugate, receptor downregulation, etc.
The prodrugs of the current invention circumvent these limitations since they represent simple, low molecular weight modifications of the drug which enable liver-selective drug delivery on the basis of the their sensitivity to liver-abundant enzymes. The prodrug cleavage mechanism was identified through studies shown in Example L. As shown in Example
A,
prodrugs are stable to aqueous solution across a broad pH range and therefore do not undergo a chemical cleavage processto produce the parent drug. In addition the prodrugs are stable to esterases and blood proteins (Examples B and In contrast to the parent drug, the prodrugs are rapidly cleaved in the presence of liver microsomes from rats (Example D) and humans (Example The drug Is also produced in freshly isolated rat hepatocytes where it is detected as the parent drug (Example I) or as a further metabolite generated by phosphoryation of the drug (Example Moreover, when the parent drug is an FBPase inhibitor, the production of the drug is supported by the ability of the prodrug to result in potent gluconeogenesis inhibition (Examples J and W).
Possible specific enzymes involved in the cleavage process were evaluated through the use of known cytochrome p450 inhibitor (Example The studies indicate that the isoonzyme cytochrome CYP3A4 is responsible based on ketoconozole ihibition of drug fonnation.
Moreover, the recombinant form of CYP3A4 was shown to catalyze prodrug cleavage (Example 0).
Analysis of the tissue distribution of CYP3A4 indicates that it is largely expressed in the liver (DcWaziers et al., J Pharnn. Ep. her. 23: 387 (1990)). Moreover, analysis of tissue.
homogenates in the presence of prodnrugs indicates that-only the liver homogenate cleaves the prodrug. Kidney, brain, heart, stomach, spleen, muscle,lung, and testes showed no appreciable cleavage of the prodrug.
Evidence of the liver specificity was also shown in vivo after both oral and i.v.
administration of the prodrugs. Administration of the prodrug of araAM (30.1) iv. gave liver levels of the bioactive drug araATP 2-5-fold greater than achieved by an equivalent dose either araA or araAMP (Example In contrast, the prodrug failed to produce detectable amounts of the araA bi-product araH, which, as reported in the literature, was readily detected after both araA and araAMP administration (Example Similarly, the prodrug 30.1 achieved high liver levels without production of the metabolite araH after oral administration. Since the prodrugs COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:37 SPRUSON FERGUSON NO, 7145 P. 34 29 PCT/US99/04908 are cleaved by liver abundant enzymes, oral administration may enable even higher liver specificity via a firn pass effect. Example P demonstrates the liver specificity for the prodrug 28.4 of PMEA compared to PMEA and the bisPOM of PMEA (283) (Example Q).
Administration of these compounds i.v. led to detection of the active metabolite PMEA diphosphate in the liver. In contrast to both PMEA and bisPOM PMEA, prodrug 28.4, showed no detectable PMEA in either the blood or urine supporting its high liver specificity (Example
Q).
Drug was also detected in the liver following administration of drugs of formulae VI- VII, shown below:: Prodrugs of the following formulas arc particularly preferred.
H
vu M o_ M P The mechanism of cleavage could proceed by the following mechanisms. Further evidence for these mechanisms is indicated by analysis of the bi-products of cleavage. Prodrugs of formula VI generate phenyl vinyl ketone whereas prodrugs of formula VIII were shown to generate phenol (Example L).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:37 SPRUSON FERGUSON NO. 7145 P. o 0 OO
H
0PX OH 0 (0H+ 00 H OP(OH) Although the esters in the invention are not limited by the above mechanisms, in general, each ester contains a group or atom susceptible to microsomal oxidation alchohol, benzylic methine proton), which in turn generates an intermediate that breaks down to the parent compound in aqueous solution via B-elimination of the phosph(on)atc diacid.
Increased Therapeutic Index The prodrugs of this invention can significantly inCrease the therapeutic Index of certain drugs. In many cases, the increased TI is a result of the high liver specificity For example, arA and raAMP are known to produce significant systemic side effects and that these side effecs are associated with blood levels otaraA byproduct.araH. Presumably the side effects are a result of toxicitics of araH or araA in rahvepatic tissues nerves) which produce e.g.
the neuropathies associated with the drug in man of patients receiving araA).
Administration of araAMP conjugated to lactosaminated serum albumin led to potent antihopatitis activity in man without the peripheral side effects. Studies of the conjugates in rats showed that the liver/blood level ratio for araA metabolites in animals treated with araA verses the conjugate had shifted approximately 3-fold, which, if a similar shift was found in man, was apparently enough to eliminate the side effect As indicated in Example 0, prodmrg 30.1 showed a substantial shift in the ratio in comparison with araAMP.
In some Cases, prodrugs of phosphoramidates that undergo cleavage by liver microsomes have been described. These drugs, however, are not used for liver diseases and are thought to COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:38 SPRUSON FERGUSON NO, 7145 P. 36 31 diffuse out of the liver after the initial oxidation and then undergo a slow base catalyzd elimination in other tissues and cells to generate the biologically active agent. The prodrugs described in this invention can be tailored such that the oxidation, but especially the elimination step, are fast and therefore occur primarily in the liver. For example, cyclophosphamide after oxidation in the liver and before the 3-elimination reaction exists as a mixture of the hydroxylated compound and the ring-opened aldehyde. Only the latter compound is converted to the phosphonic acid and acrolein. The conversion is slow due to the high propensity of the aldehyde to hydrate, undergo recyclization or undergo further oxidation. In fact, the aldehyde exists only as a minor component in solution Prodrugs of formula VI-VII do not readily recyclize, since the carbonyl product is a ketone except when Z-CH2OR in formula VII.
Ketones do not hydrate to a great extent nor do they undergo the same metabolism associated with the aldehyde.
Renal toxicity is a common toxicity associated with phosphonic acids. The toxicity results from transport, e.g. via the organic anion transporters, of the negatively charged drug into e.g. tubular cells which then accumulate the drug to high concentrations unless there is an equally efficient transport of the drug out of the cell via transporters on the basolateral side.
Many examples have been reported in the literature of nephrotoxic phosphonic acids, e.g. PMEA and HPMPA. The novel prodrug of PMEA showed only small amounts of PMEA in the urine relative to either PMEA or bisPOM PMEA at doses that achieved similar liver drug levels (Examples 0 and Q).
Another common toxicity associated with phosphonic acid drugs is gastrointestinal toxicity via in some cases GI erosions. Prodrugs of the current invention can decrease GI toxicities. especially toxicities produced by direct action of the drug on the GI tract after oral administration, since the largest proportion of the phosphonate is not revealed until after absorption and cleavage in the liver.
Severe toxicities are also associated with nearly all anticancer agents. In an effort to decrease these toxiciies during treatment of primary or secondary liver cancers, drugs are sometimes administered directly into the portal artery 5-FU and 5-FdUMP). The high liver specificity of the prodrugs in the current invention suggest that systemic side effects will be minimized by the novel prodrug approach.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB-2004 17:38 SPRUSON FERGUSON NO, 7145 P. 37 Prodrugs of the invention are generated by a postulated mechanism involving an initial oxidation followed by a 0-elininadion reaction. In some cases, e.g. cenain prodrugs of formula' VI and formula VII, the biproduct of the reaction is an a $-unsaruats carbonyl compound, e.g.
vinyl phenyl ketone for prodrugs where V- Ph, Z, W and W' H. Compounds that react with nucleophiles via a Michael addition can lead to certain toxicitics, acrolein produces bladder toxicities) and mutagenic activity. The degree to which these activities limit the use of compounds of Formula V] is dependent on the severity of the toxicity and the indicated disease.
Prodrugs that produce non-toxic and non-mutagenic biproducts arc especially preferred for the treatment of chronic diseases diabetes), Frequently, it is difficult to predict the mutagenic properties of a compound. For example, a number of acrylates have been shown to produce positive mutagenic responses as indicated by increased chromosome aberrations and micronucleus frequencies in cultured L5179Y mouse lymphoma cells (Dearlield et al., Mutagenesis 4,381.393 (1989)). Other acrylates, however, are negative in this test Tox.
Envir. Health, 34, 279-296 (1991)) as well as in the Ames test and the CHO assay which measures newly induced mutations at the hypoxanthine-guanine phosphoribosyltransferase (hgprt) locus (Mutagenesis 6, 77-85 (1991)). Phenyl vinyl ketone lacks teratogenic activity in rat embryos in culture suggesting that it may not be mutagenic nor highly toxic (Teratology 39, 31-37 (1989)).
Since mutagenicity and toxicity are not highly predictable properties. non-mutagenic prodrugs of formula I and their associated bi-products can be readily identified by conducting well known in vitro and in vivo assays. For example, compounds can be tested in nonmammalian cell assays such as the Ames test, a fluctuation test in Kl. pneumaniae, a forward mutation assay with S. typhimurium. a chromosome loss assay in Saccharomyces ccrevisiae ,or a D3 recombinogenicity assay in Saccharomyce: cerevisiae. Compounds can also be tested in mammalian cell assays such as the mouse lymphoma cells assay heterozygotes of L5178Y mouse lymphoma cells), assays in Chineese hamster overy cells CHO/HGPRT assay), and an assay in rat liver cell lines RL1 or RL4), Each of these assays can be conducted in the presence of activators liver microsomes) which may be ofparticular importance to these prodrugs. By conducting these assays in the presence of the liver microsomes, for example, the prodrug produces products, such as phenol or vinyl ketone. The mutagencity of the by-product is measured either directly or as a prodrug where the rsults are compared to the parent drug alone. Assays In liver cell lines are a preferred aspect of the COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:39 SPRUSON FERGUSON NO. 7145 P. 38 33 invention since 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 compounds. For example, the liver contains reductases that with some bi-products might result in reduction of the carbonyl.
A variety of end points are monitored including cell growth, colony size, gene mutations, micronuclei formation, mitotic chromosome loss, unscheduled DNA synthesis, DNA elongation, DNA breaks, morphological transformations, and relative mitotic activity.
In vivo assays are also known that assess the mutagenicity and carcinogenicicty of compounds. For example, a non-mammalian in vivo assay is the Drosophila sex-linked recessive lethal assay. Examples of mammalian in vivo assays include the rat bone marrow cytogenetic assay, a rat embryo assay, as well as animal teratology and carcinogenicity assays.
Resistancea vvass Drug resistance following prolonged treatment is a common finding for anticancer drugs and antiviral drugs used to treat hepatitis. The mechanisms for the drug resistance have been identified in many cases and involve both decreased drug transport into cancer cells, increased drug export, increased drug metabolism and decreased precursor conversion to the active drug.
Many of the drugs used to treat these diseases are drugs that are converted to the corresponding triphosphate, which in turn acts as a DNA chain terminator, inhibitor of DNA polymerase or inhibitor of reverse transcriptase. In some cases, drug resistance results from a decrease in activity of the enzymes responsible for synthesis of a nucleoside mono-phosphate kinases such as thymidylate kinase or enzymes in the biosynthesis pathway of 5-fluoro-2'-deoxy UMP).
Administration of the prodrug generates the monophosphate by a different pathway avoiding the pathways that cause the resistance to the parent drug, Thus, the prodrugs of the present invention can achieve a therapeutic effect in cells resistant to the parent drug.
Tynes of Parent Drugs Various kinds of parents drugs can benefit from the prodrug methodology of the present invention. Parent drugs of the form MH, which are phosphorylated to become the biologically active drug are well suited for use in the prodtug methodology of the present invention. There are many well known parent drugs of the form MM which become biologically active via phosphorylation. For example, it is well known that antitumor and antiviral nucleosides are COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB-2004 17:39 20. EB. 004 7:39SPRUSON FERGUSON NO 7145 P. 39 34 activiate through PhOsPhOrylation3 -These Compounds include aA, AZT, d4T, ddl, ddA, ddC, L-ddC, L-FddC, L-d4c, L-Fd4c, ZTC, ribavirin. penciclovir, S-fluoro-2'-dcOXYtUidinc,
FIAU,
FTAC, BHCG ,p
I-(
2 -(hydroxyrnehyl)oxahioIans-y,]c,.± 08 In, .2 dideoxycyridine, ',3'-dideoxy-S-fluorocyrjdj, FMjAU, BvarU, E-$*(2_bromovinyz).
2 '-dcoxyuridnc, Cohucavir, TFT. S-PrOPynyl-I -lbi syluracil, CDO, DAPD, FDOC, d4c, DXG, FEFAT., FiG, FLT, FTC, S-yl-cabocyclik 2 '-dcxyguanosine Cytallene, Owxeanocin
A,
Oxctanocin G, Cyclobut A, Cyclobut 0, fluorodcoxynridine, dFdC, araLC, bromodeoxyudidin 8 IDU, CdA., F-araA, 5-FdUMvP, Cofonnydin, 2 1 Acoxycotoycin, PMEA, PMEDAP, I{PMc, ilMPA, FPMPA. and PMPA.
Preferd antiviral drugs include: aaA; 9 -b-D-arabinofbnanosyladeine (Vidarabnc); AZT; 3 -Azido-2'Y3-dideoxythymdine (Zidovudine); d4T; 2 .$3'-didehydro-3'-deoxyJhymidinc (Stavudine); ddA; 2 3 -diderntyadenosinc; ddC; 2 7 ,3'-didcoxycytidine (Zakcitabine); 3TC; 2 ',3'-dideoxy-3'-tlacytiji 0 (Lamivudine); I -b-D-riboftzraniosy.. 2 ,l-tri2ale-3.varboxamide (Ribavirin); PMEA; 9-(2-rhospboaymcthoxyethyl) adenine (Addbovir); HFMPA; 9 -(3-hydroxy2.phosponylmeho,)?rpyj) adunine; ACV; 9 2 -hycdroxyerhoxyimacjhyj)ggu,.jn. (Acyclovir); 9 ,1( 4 -hYdroY-3hydroy t I t--yl)guaninc (Penciclovix'); S-yl-earbocycic 2'-deoxyguanosjnc (BMSZO,41s); and phosphonobyrnic acid (Foscaniet).
More preferred antiviral drugs include: araA; 9-b-D-arabinofuranosyiadernen (Vidaratine); AZT; 3 3 zido2'Y-dldeaxyuiymdlne (Zidovudine); d4T; 2
'X
3 .didehydro..3'-deoxythymnidixe (Stavudine); 3TC; 2 'Y3-dideoxy-3'-thiacyicin. (Larnivudinc); I -b-D-ribofuraosy1-i 2 3 4 triazol-3-caboxide (Ribavirin); PME-A- 9 2 -phosphonylmcthoxyethyI) adenine (Adefovir); ACV; 9 2 -bydroxyerhoxylrnctnyl)gusrine (Acyciovir); 9 4 -hydroxy.3-hydoxymethybut- 1 -yi)guariine (Penciclovir); and -yl-carbocyclic 2 -deoxcyguanosinc (BMS200,475).
Preferred anticancer drugs include:, d~'dC; 2 2 '-difluorodoxycyiin. (gemaitabine); uriC ;arabinosylcytoainc (cytarabine); F-ara-A Z-Oluoroanabiuosyladenosinc (fludarabine); and 2 -chloradeoxyadenasine (ci adribine).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:39 SPRUSON FERGUSON 20.FEW206 1739SPRSON& FRGUONNO. 7145 P. Drugs containing a phosphoriic acid (C-P0 3 2 Moiety are also suitable parent rugs advantageously used in the present invention. These dru s are biologically active either in the form of(MPO, 2 or MP,0 9 t Phosphonic acids that are also suitable for this prodrug delivery strategy include protons inhibitors that amc useful for example as antihypertensives, anticancer or anciinflaxnmtnaozy agents. The novel prodrug methodology can be applied to NEP inhibitors, (DeLambert et al. Med. Chem 37:498 (1994)), ACE inhibitoars, endothelin conventing enzyme inhibitors, purine nucleoside phosphatase inhibitors of mnetalloproreases involved in tumor mezasclsis, and inhibitors of collagenase (Bird et al.,LI Mod. Chem. 37, 158- 169 (1994). Moreover, phosphonic acids usefull as NMDA antagonists which are usefUl for treating a variety of conditions, including stroke, head tratuma, pain, and epilepsy. Other phosphonic acids that could benefit from the prodrug tegies are phosphonic acids reported by Squibb chat inhibit squalene synthase by Hoechst to be iznmunomodulators, by Merck to be antidepressants, by Ciba-Geigy and Marion Moetc Dow to be irnmunasuppresaanls via inhlibition of purine nucleoside phosphorylase, antiviral HIV) by Bristol-Myers Squibb, IS Gilead. Certain antibiotics might be suitable, especially antibiotics such as D-alanine nacrnase inhibitors and fosfomycin and associated analogs.
The following compounds and their analogs can be used in the prodnig methodology of the resent invention: NEP Inhibitors [(PhosPhonomethy)aminol-3.4biphonyly)pwpionyl]nmino~propionjc acid by lDeLombaert et al in J Med Chem. 1994 Feb 18;37(4):498-511I Collagen ase Inhibitors I -phosphorwpropyl(4(S)-leucyl]-(S)..phenytaljgjne N-methyl amide by Bird St ak~in J Mfed Chemn 1994 Jan 7;37(t):158-69 Angloceusin Caverting Enzyme Inhibitors l-(N-(N-acetyl-L-isoleucyl)-L-tyosyl)amnino-2-(4-hydroxyphenyl)ethy I- phosphonic acid by Hirayemia cc al. in lar J Pepe Protein Res 1991 Jul;3 3(1l):20-4.
Endotbdlin Inhibitor CGS 26303 by Detonbaert et al. Biochem Slop hys Rej Commun 1994 Oct 14;204(1).-407-12 S)-3-Cyclohexyl-2-[[5-(2, 4 -diflucrophcnyl)-2.[(phosphonomeliyl)aixiopte.
ynoyljaminoJ propionic acid by Wallace et al. J~ded Chain 1998 Apr 23;41 (9):1513-23 S)2[5(-loohnl--(hshnoehla~n~ct4yol=nl4 metbylpeawnoic acid COMS ID No: SMBI-0062962-7 Received by IP Australia: Time (F1:m) 18:45 Date 2004-02-20 20-FEB-2004 17:40 20. EB. 004 7:40 SPRUSON FERGUSON N.74 .4 NO. 7145 P. 41 36 mcthylpcnanoic acid NMDA/AMPA Antagonists dsrbdi Bioorg Med Chem Lam. 1999 Ja 1 8;9(2):249,.54 3-2croyieai--l--rpnllpopoi avid by Bespalov et al. in EurJ Pharmacat 1998 Jun 26,351(3):299-30S 2 8 7 9 -dioxo-2,6-diazabicyclo[S-2o0noa..i( 7 )-en-2-yI)-ethyllphosphonic acid D4L-(E)-2-amino.4-(aa]..propyls.phosphono3ptenoi acid &7-dichloro-2(1H)-axoqiinoine.3..pnosphoic acid by Des ot al in I1 Med Chew. 1996 Jan 5.39(l):197-206.
cis-4-(pbosphonrnrby)pipedidizc.2..carijoxylic acid (CGS 19755) Purius Nucleoside Phosphorylase Inhilbitors [7-(2-awino-1 3 6-dilydro-6-chloro.911.purin..9.yl) 1, l-diflorcheptyllphosphonjc acid and E4"5-unino-6,7-dihydro-7-ozo-3x- lP 2 t 3 I-triazolo(4 1 5.d-pyhimida-..y)buw1JlphosphoInic adid.
[[tS-(2-umino-1 7 6-dihydro-6-oxo-9H-pwin-9. yipny~hshmcomty~hpo acid by Kelly at al. indJMed Chum 1995 Mar 17 -38(6):1005-14 amio-i IS-dihydro-6oxo-9Fi-puri- 9 .yI)methyll-phcnyljcthenyl) -pbosphnnic acid by Weibel at al in Blochemn Phannacol. 1994 Jul! 19;48(2):245-52.
9 -(3,3-Dimethyl-5phospbonopenty)gjanjn by Guida et al. in IMud Chem 1994 Apr 15;37(g):1109-14.- Alanina Racewuse Jnhibitors DL-(1-Amino-Z-propenyl)phosphonic acid by Vo-Quang at al. in JMd Churn 1956 Ap3r.29(4):579-8I Squailene Syutbuse Inhibitors I -fYdroxy.3-(mcthylpontylai1g)-propyiidenc- 1,4 -bisphosphonic acid by Amin a 4 in Arnzcumutelforschung. 1996 Aug;46(8):-759.42, BMS 188494 is POM prodrug of BMS 187745 by Dickson et al. in J Mad Chew. 1996 Feb 2;39(3):661 -4.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:40 SPRUSON FERGUSON NO. 7145 P. 42 37 Treatment of Canceri The prodrug strategy in the current invention encompasses several features that are advantageously used in cancer therapies. Many of the known anticancer drugs are nucleosides that undergo phoaphorylation to the monophosphate and in many cases to the triphosphate. The prodrug strategy can be effective in the treatment of liver cancer because the drug is cleaved by liver-abundant enzymes which suggests that a greater TI will result since much less drug is present in the blood and therefore available to produce side effects via distribution to other tissues. With these drugs, the monophosphate generated after prodrug cleavage is rapidly converted to the triphosphate which in turn causes DNA chain termination, DNA polymerase inhibition, etc. The prodrug strategy also enables bypass of some well-know resistance mechanisms including mechanisms involved in the production, export and metabolism of the monophosphate. Examples of preferred drug candidates that are specifically amenable to the strategy include, e.g. dFdC, araC, F-araA, and CdA.
Some prodrugs may result in some accumulation of the monophosphae in cells. Certain monophosphates are useful for the treaunent of cancers, e.g. monophosphates that are potent inhibitors of thymidylate synthase. Some TS inhibitors are reported to be moderately effective in treating liver cancers. For example, 5-FU and 5-FdUMP are effective. These drugs, however, are plagued by drug resistance and severe side effects. To avoid the latter, 5-FU analogs are often delivered via the portal artery to achieve the highest possible liver levels. Drug resistance is also very common. Accordingly, 5-FdUJMP and associated analogs are suitable targets for the prodrug strategy.
Treatment of Viral Infections: Drugs useful for treaing viruses that infect the liver and cause liver damage, e.g. hepatitis virus strains, exhibit similar properties to the anticancer nucleoside drugs in terms of efficacy, side effects and resistance. Prodrugs of drugs such as araA, AZT, d4T, ddl, ddA, ddC, L-ddC, L-FddC, L-d4C, L-Fd4C, 3TC, ribavirin, penciclovir, 5-fluoro-2'-deoxyuridine, FIAU, FIAC, BHCG, 2'R,5'S(-)-l-[2-(hydroxymethyl)oxathiolan-5-yl]cytosine, (-)-b-L-2',3'-dideoxycytidine, (-)-b-L-2',3'-dideoxy-5-fluorocytidine, FMAU, BvaraU, E-5-(2-bromovinyl)-2'-deoxyuridine, Cobucavir, TFT, 5-propynyl-l-arabinosyluracil, CDG, DAPD, FDOC, d4C, DXG. FEAU, FLO, FLT, FTC, 5-yl.carbocyclic 2'-deoxyguanosine. Cytallene. Oxetanocin A, Oxetanocin G, Cyclobut A, Cyclobut 0, fluorodeoxyuridine, dFdC, araC, bromodeoxyuridine, IDU, CdA, F- COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:40 20. EB. 004 7:40 SPRUSON FERGUSON N.74 .4 NO, 7145 P. 43 38 MrA, 5-EdUMP, Cofonnyrip, Z'-deoxycofonnyein, PMEA, PMEDAP, HPM.PC, RPMPA, FPMI'A. and PMFA would therefore be usefuli in treating hepatitis. In some Cases, the drugs axe already targeted for hepatitis (tg. mrA, 3TC, FIAU, EMS 200,975). The prodrugs ofthene copounds could enhance the efficacy, increase the therapeutic index, improve the phannacodynamic half-life and/or bypass drug resistance. Prodrugs of other agents used to treat viral ifections other than hepatitis miay also be made useful by adininistra~jn of the prodrugs of this inven~tion since the resistance will be delivery of monophospan HSV). Ofte they require phosphoiylatinn to. the monophospbate by a viral kinase, which is not present in all viruses nor in'mammaliani cells. The monophosphate is convented to the biologically active triphosphate by mamnmalian kinases. Accordingly, delivery of the monophosphace using this clan of prodrugs enables treatment of hepatitis by drugs normally used to treat other viral infections.
The following antiviral drugs are preferred: araA4 9-b-D-arabinofizranolayladcpine (Vidarabine); AZT; 3 '-azido-2',3 dideoxythymdine (Zidovudine); d4T, 2'.3'-didehydro-3 'Aeoxythyrnidine (Stavudine); ddA; 2'3 '-dideoxyadenosine: ddC; 2'3'-dideoxycytidine (Zalcitatine); 3TC; '-dideoxy-3'-thiacyujdjne (Laznivudips); 1 -b-D-ribofiaranosyl- lZ24-triazole-3-carboxnmid 0 (Ribavirin); PMEA4 9-(2-phosphonylmecthoxyeihyl) adenine (Adefovir); fIPMPA; 9 3 .hydroxy-2-phsphonyrmthoyprpyl) adenine; ACV; 9 -(2-hydroxyehoxylmthyl)guaine(Acyclj 0 i) 9 4 -hydroxy-3-hydroxymnetylbut- 1 -yl)guanine(Pencjclovjr); 5-yl-carbovyclic 2-deoxyguanosine (BMS200A47s): and phosphonoforrnic (Foscmet).
More prefered are the following mntiviral drugs: amA; 9-b-D-arabinofumaosyladenine (Vidarabirie); AZT; 2 '-azido-2',3 '-dideoxythyngline (Zidovudine); d4T; 2',3 '-didehydro.2 '-deoxyrbymidine (Stavuadine); 3TC; (-)-2',3'-dideoxy-3 '-thiacytidine (Lamnivudine); COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:41 SPRUSON FERGUSON NO. 7145 P. 44 39 1-b-D-ribofuranosyl-1 ,2,4-triazole-3-carboxanmide (Ribavirin); PMEA; 9 2 -phosphonylmethoxyethyl)adenine (Adefovir); ACV; 9 2 -hydroxyerhoxylmcthyl)guanine (Acyclovir); 9 4 -hydroxy-3-hydroxymethylbut-. -yl)guanine (PenCiclovir); and 1 5-yl-carbocyclic 2'-dcoxyguanosine (BMS200,475).
Treatment of Diaberes: A variety of phosphonic acids have been described that are useful in inhibiting the enzyme fructose 1.6-bisphosphatase (FBPase) and flux through the pathway that uses FBPase activity, namely gluconeogenesis. Inhibition ofgluconeogenesis results in significant blood glucose lowering in diabetic animals. As with other phosphonic acids, these compounds are poorly orally bioavailable and exhibit short plasma half-lives. Prodrugs of compounds from the following structural classes are cleaved by cytochrome p450 CYP3A4, rat and human liver microsomes, rat hepatocytes. The prodrugs exhibit enhanced oral bioavailability and good liver drug levels.
The phosphate and phosphonate compounds may be inhibitors of FBPase activity, preferably with ICS0s of about 10 ;M on the human liver enzyme, or are other compounds with a biological activity such that they are useful in preventing or treating diseases or conditions, viral infections, cancer, hyperlipidemia, liver fibrosis, and parasitic infections such as malaria. The esters increase oral bioavailability of the parent compound and preferably achieve an oral bioavailability of greater than Compounds that exhibit glucose lowering activity in vivo and bind to the AMP site of FBPase as the corresponding 5'-mono-phosphate are compounds represented by formula A where Y is hydroxy, acyloxy or alkoxycarbonyloxy; E is selected from group consisting of hydrogen, alkyl, amino or halogen; J L
A
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:41 SPRUSON FERGUSON NO. 7145 P. L and J are independently selected from the group consisting of hydrogen, hydroxy, acyloxy or when taken together form a lower cyclic ring containing at least one oxygen; and A is selected from the group consisting of amino and lower alkyl amino; and pharmnaccutically acceptable salts thereoC Phosphonates containing a purine, benzimidazole, indole or imidazopyridine also bind to the AMP site of FBPase and lower glucose in diabetic animal models. WO 98/39344, WO 98/39343, and WO 98/39342. These compounds are represented by formulae B D. Prodrugs of these compounds are ednsidered of potential use in oral delivery.
N N -P
Y
B
A
I Y OH
C
A
E~ 0 qOH S Y Esters disclosed in the invention are converted to the parent phosph(on)ate in cells and tissues, especially hepatocytes'and liver, as indicated by measurement of the intrcellular drug metabolites in hepatocytes using the procedure described in Example I and by the inhibition of glucose production by rat hepatocytes when M-P03, is an FBPase inhbitor (Example J).
Treatment of Hygrlinidemia: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:41 SPRUSON FERGUSON NO. 7145 P. 46 Phosphonic acids arc known to produce an antihyperlipidemic effect in animals' The amihyperlipidemic activity is associated with inhibition of squalene synthase. The drugs exhibil poor oral bioavailability, For example BMS 188494 exhibited oral bioavailability in rodents. The bisPOM diester provided modest improvement. l-Hydroxy-3- (methylpentylamino)-propyliden- 1,1 -bisphosphonic acid and BMS 187745 are preferred squalene synthetase inhibitors for use in the present invention.
Treatment ofLiver Fibrosis: A variety of compounds are reported to be useful in treating liver fibrosis that are also compounds suitable for the prodrug strategy described in this invention. For example, N,[N- (CR)-1-phosphonopropyl(1-(S)-lencyl]-(S)-phenylalanine N-methyl amide was described that inhibit collagenase (Bird et al. J. Med. Chem. 37:158-169 (1994)).
Preferred Compounds The compounds of the invention are substituted 6-membered cyclic 1,3-propane diester prodrugs of certain phosphates, phosphonates and phosphoramidates (M-PO3') as represented by Formula 1:
V
"H
M-P 2 W W wherein; V is selected fromrn the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, I-alkenyl, 1-alkynyl, and -KR; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atorms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:41 SPRUSON FERGUSON NO. 7145- P. 47- 42 together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6.carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alykthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl, aralkyl, alicyclic. aryl, substituted aryl, heteroaryl, substituted heteroaryl, I-alkenyl, I -alkynyl, and Z is selected from the group consisting of-CHR 2 OH, -CHR 2
OC(O)R
-CHR
2 OC(S)R', -CHRzOC(S)OR', -CHROC(O)SR,
-CHR
2 OC,, -OR 2
-SR
-CHRN3,
-CH
2 aryl, -CH(aryl)OH, -CH(CH-CR2)OH, -CH(C=CR)OH, -NR -OCOR', -OCOR', -SCOR', -SCORW, -NHCOR -NHCOaRj, -CHiNHyl,
-(CH
2 )p-OR 2 and -(CHz)p-SR2; R is an R or -H; SR is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and
R
9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3: with the provisos that: a) V, Z. W, and W' are not all and b) when Z is then at least one of V and W is not or and M is selected from the group that attached to PO," P20 6 3 or P,0 9 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
In the composition, method of use, and prodrug plus effect claims, the following compounds are preferred.
In general, preferred substituents, V, Z, W, and W' of formula I are chosen such that they exhibit one or more of the following properties: enhance the oxidation reaction since this reaction is likely to be the rate determining step and therefore must compete with drug elimination processes.
the prodrug is stable in aqueous solution and in the presence of other non-p450 enzymes; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:42 SPRUSON FERGUSON N0. 7145 P. 48 S~ 43 the prodrug is not charged or of high molecular weight since both properties can limit oral bioavailability as well as cell penetration; promote the P-elimination reaction following the initial oxidation via one or more of the following properties;: a) fail to recyclize after ring opening; b) undergo limited covalent hydration; c) promote P-eliminaion by assisting in the proton abstraction; d) impede addition reactions that form stable adducts, e.g. thiols to the hydroxylated product or nucleophilic addition to the carbonyl generated after ring opening; and e) limit metabolism of reaction intermediates ringopened ketone); lead to a non-toxic by-product with one or more of the following characteristics: a) non mutagenic; b) poor Michael acceptor c) electron donating groups that decrease double bond polarization; d) W groups that sterically block nucleophilic addition to P-carbon; e) Z groups that eliminate the double bond after the elimination reaction either through retautomerization (enol->keto) or hydrolysis enamine);.
f) Z groups that form a stable ring via Michael addition to double bond; and g) groups that enhance detoxification of the byproduct by one or more of the following characteristics: confine to liver; and (ii) make susceptible to detoxification reactions ketone reduction); and capable of generating a pharmacologically active product.
Suitable alkyl groups include groups having from I to about 20 carbon atoms. Suitable aryl groups include groups having from 1 to about 20 carbon atoms. Suitable aralkyl groups include groups having from 2 to about 21 carbon atoms, Suitable acyloxy groups include groups having from I to about 20 carbon atoms. Suitable alkylene groups include groups having from 1 to about 20 carbon atoms. Suitable alicyclic groups include groups having 3 to about 20 carbon atoms. Suitable heteroaryl groups include groups having from 1 to about 20 carbon atoms and from 1 to 5 heteroatoms, preferably independently selected from nitrogen, oxygen, phosphorous, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 17:42 SPRUSON FERGUSON NO. 7145 P. 49 44 and sulfur. Suitable heteroalicyclic groups include groups having from 2 to about twenty carbon atoms and from I to 5 heteroaoms, preferably independently selected from nitrogen, oxygen, phosphorous, and sulfur.
In one preferred aspect, M is attached to the phosphorus in formula I via an oxygen atom.
Preferably, M is a nucleoside. Preferably, M is anached via an oxygen that is in a primary hydroxyl group on a ribofuranosyl or an arabinofuranosyl group. In another aspect, it is preferred when M is attached via an oxygen in a hydroxyl on an acyclic sugar it is preferred when such MH is ACV, GCV, 9-(4-hydroxy-3-hydroxymethylbutI -yl)guanine, or dihydroxyburyl)guanine.
In general, it is preferred that when M is attached via an oxygen, said oxygen is in a primary hydroxy group. In such an instance, it is preferred that MH is araA, AZT, d4T, ddl.
ddA, ddC, L-ddC, L-FddC, L-d4C, L-Fd4C, 3TC, ribavirin, penciclovir, 5-fluoro-2'deoxyuridinc, FIAU, FIAC. BHCG, 2 2 -(hydroxymethyl)oxatholaan-5-ylJcytosine, )-b-L-2',3'-dideoxycytidine, (-)-b-L-2',3'-dideoxy-$-fluorocytidine, FMAU, BvaraU. E-5-(2bromovinyl)-V-deoxyuridine, Cobucavir, TFT, 5-propynyl-l-arabinosyluracil. CDG, DAPD, FDOC, d4C, DXG, FEAU, FLG, FLT, FTC, 5 -yl-carbocyclic 2'-deoxyguanosine, Cytallenc, Oxetanocin A, Oxetanocin G, Cyclobut A, Cyclobut 0, fluorodeoxyuridine, dFdC, araC, bromodeoxyuridine. IDU, CdA, F-araA, 5-FdUMP, Coformycin. or 2'-deoxycoformycin.
Another preferred group of compounds with M attached via oxygen are M as a compound of formula II:
,A
J L
II
wherein E is selected from the group consisting of alkyl, amino or halogen; L and I are independently selected from the group consisting of hydrogen, hydroxy, acyloxy, alkoxycarbonyioxy, or when taken together form a lower cyclic ring containing at cast one oxygen; and COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:42 SPRUSON FERGUSON NO. 7145 P. A is selected from the group consisting of amino and lower alkylamino; and pharmaceutically acceptable salts thereof.
In another aspect, compounds of formula I wherein M is attached to the phosphorus in formula I via a carbon atom are preferred. In such compounds, preferably M-PO, 2 is phospohonoformic acid, or phosphonoacetic acid.
For compounds where M is attached via a carbon atom, it is also preferred when M is a compound of formula lI:
A
NN
E1
X-
Y
wherein A is selected from the group consisting of -NR82, NHSOzR -OR 5 -SR, halogen, lower alkyl, -CON(R guanidine, amidine. and perhaloalkyl; E is selected from the group consisting of-H, halogen, lower alkylthio, lower perihaloalkyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, -CN, and -NR72; X is selected from the group consisting of alkylamino, alkyl, alkcnyl, alkynyl, alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonate), alkyl(sulfonate), aryl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthio, alicyclic, 1,1-dihaloalkyl, carbonylalkyl, alkylaminocarbonyl, alkylcarbonylamino, aralkyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including cyclic alkyl, heterocyclic, and aryl; Y is selected from the group consisting of-H, alkyl, alkcnyl, alkynyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl, -C(0)R 3 -S(0)2
R
.C(0)-ORI, -CONHR -NR22, and -ORJ, all except H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic;
R
4 is independently selected from the group consisting of H. lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:26 SPRUSON FERGUSON NO. 7146 P. 2/51 46
R
3 is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic;
R
6 is independently selected from the group consisting of H, and lower alkyl;
R
7 is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, lower aryl, and -C(O)R 1 0; R' is independently selected from the group consisting of-H, lower alkyl, lower aralcyl, lower aryl, lower alicyclic,
-C(O)R
i or together they form a bidentate alkyl; R1 0 is selected from the group consisting of--H, lower alkyl, -NH2, lower aryl, and lower perhaloalkyl;
R
11 is selected from the group consisting of alkyl, aryl, -OH, -NH2 and and pharmaceutically acceptable prodrugs and salts thereof.
For compounds where M is attached via a carbon atom, it is also preferred when M is a compound of formula IV:
A
N
A)-
wherein: A. E, and L are selected from the group consisting of-NR 8 2 -NO2,
-OR
7
-SR
7 -C(O)NR42, halo, -COR 1 S2R.
3 guanidinc. amidine, -NHSO 2 R, SO2NR 4 2
-CN,
sulfoxide, perhaloacyl, perhaloalkyl. perhaloalkoxy, Cl-C5 alkyl, C2-CS alkenyl, C2-CS alkynyl. and lower alicyclic, or together A and L form a cyclic group, or together L and E form a cyclic group, or together E and J form a cyclic group including aryl, cyclic alkyl, and heterocyclic; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:26 SPRUSON FERGUSON NO 7 14 6 P. 3/ 5 1 47- J is selected from the group consisting of -NR 8 2, -N02,
-OR
7
-SR
7
.C(O)NR
4 2, halo, -C(O)R I 1 -CN, sulfonyl, sulfoxide, perhaloalkyl, hydroxyaLkyl, perbaloalkoxy, alkyl, haloalkyl, aminoalkyl, alkcnyl, alkynyl, alicyclic, aryl, and aralkyi, or togethcr with y forms a cyclic group including aryl, cyclic alkyl and heterocyclic alkyl; X is selected from the group consisting of alkylamino, alkyl(hydroxy), alkyl(carboxyl), alkyl(phosphonatc), alkyl, alkenyl, allcynyt, alkyl(sulfonate), aryl, carbonylalkyl, 1.1dihaloalkyl, akylaminoalkyl, alkoxyalkyi. alkylthioalkyl, alkylthio, alkylaminocarbonyl, alkylcarbonylamino, alicyclic, aralicyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including aryl. cyclic alkyl, and hcterocyclic; Y is selected from the group consisting of-H, alkyl, alkenyl, alkynyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl,
-C(O)R
3 -S(0)2R, -CONH.R,
.NR
2 2 and
-OR
3 all except -H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic;
R
4 is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, and lower aryl;
R
5 is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic;
R
6 is independently selected from the group consisting of-H, and lower alkyl; R is independently selected from the group consisting of-H, lower alkyl, lower alicycliq, lower aralkyl, lower aryl, and -C(O)RI0O
R
8 is independently selected from the group consisting of-H, lower alkyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R 10 or together they form a bidentate alkyl; R 1 0 is selected from the group consisting of-H, lower alkyl, -NH 2 lower aryl, and lower perhaloalkyl; Ril is selected from the group consisting of alkyl, aryl, -OH, -NH2 and
-OR
3 and pharmaceutically acceptable prodrugs and salts thereof; with the provisos thata) when X is alkyl or alkene, then A is -NR2; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:26 SPRUSON FERGUSON NO, 7146 P. 4/51 48 b) X is not alkylamine and alkylaminoalkyl when an alkyl moiety is substituted with phosphonic esters and acids; and c) A, L, E, J, Y, and X together may only form 0-2 cyclic groups.
Preferred A, L, and E groups include .NR'2, -NO 2 hydroxy, alkylaminocarbonyl, halogen. -OR 7
-SR
7 lower perhaloalkyl, and CI-C5 alkyl, or together E and J form a cyclic group. Such a cyclic group may be aromatic, cyclic alkyl, or.heterocyclic alkyl, and may be optionally substituted. Suitable aromatic groups include thiazolidine. Particularly preferred A, L and E groups are -NR 2, hydroxy, halogen, lower alkoxy, lower perhaloalkyl, and lower alkyL Preferred A groups include, halogen, lower perhaloalkyl, and lower alkyl.
Preferred L and E groups include lower alkoxy, lower alyl, and halogen.
Preferred J groups include halogen, lower alkyl, lower hydroxylalkyl,
-NR*
2 lower R'zN-alkyl, lower haloalkyl, lower perhaloalkyl, lower alkenyl, lower alkynyl, lower aryl, heterocyclic, and alicyclic, or together with Y forms a cyclic group. Such a cyclic group may be aromatic, cyclic alkyl, or heterocyclic, and may be optionally substituted.
Particularly preferred J groups include halogen, and lower alkyl, lower hydroxyalkyl, -NRz, lower R'aN-alkyl, lower haloalkyl, lower alkenyl, alicyclic, and aryl. Especially preferred are alicyclic and lower alkyl.
Preferred X groups include alkyl, alkynyl, aryl, alkoxyalkyl, alkylthio, alkylaminocarbonyl, alkylcarbonylamino, 1,1-dihaloalkyl, carbonylalkyl, alkyl(OH), and alkyl(sulfonate). Particularly preferred is hereroaryl, alkylaminocarbonyl, 1,1 -dihaloalkyl, alkyl(sulfonate), and alkoxyalkyl. Also particularly preferred are heteroaryl, alkylaminocarbonyl, and alkoxyalkyl. Especially preferred are methylaminocarbonyl, methoxymethyl, and ftranyL In one preferred aspect X is not substituted with a phosphonic acid or ester. In another preferred aspect, when X is substituted with a phosphonic acid or ester, then A is and Y is not In another preferred aspect, when X is aryl or alkylaryl, these groups are not linked 1,4 through a 6-membered aromatic ring.
Preferred Y groups include alkyl, aralkyl. aryl, and alicyclic, all except -H may be optionally substituted. Particularly preferred are lower alkyl, and alicyclic.
Preferred R and R 7 groups include and lower alkyJ.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:27 SPRUSON FERGUSON NO. 7146 P. 5/51 49 In one preferred aspect A, L, and E are independently lower alkyl, hydroxy, halogen, lower alkoxy, lower pcrhaloalkyl, and -NR 2 X is aryl, alkoxyalkyl, alkyl, alkylhio, I,1-dihaloalkyl, carbonylalkyl, alkyl(hydroxy), alkyl(sulfonate), alkylaminocarbonyl, and alkylcarbonylamino; and each R. and R 7 is independently and lower alkyl. Particularly preferred are such compounds where A, L, and E are independently
-H,
lower alkyl, halogen, and -NR'2; J is halogen, haloalkyl, hydroxyalkyl, R'z2Nalkyl, lower alkyl, lower aryl, heterocyclic, and alicyclic, or together with Y forms a cyclic group; and X is heteroaryl, alkylaminocarbonyl, 1,1-dihaloalkyl, and alkoxyalkyl. Especially preferred are such compounds where A is -NHI, and -CH3, L is -F, -OCH3, -CI, and -CH3, E is -H and -Cl, J is halo, Cl-C5 hydroxyalkyl, CI-C5 haloalkyl, Cl- R'zN-alkyl, Cl-C5 alicyclic, and Cl-CS alkyl, X is -CHZOCHz-, and 2,5-furanyl, and Y is lower alkyl. Most preferred are the following such compounds and their salts, and prodrug and their salts: 1) A is -NHz, L is E is J is Y is isobutyl, and X is 2) A, L, and J are E is -Cl, Y is isobutyl, and X is 3) A is -NH2, L is E and J are Y is cyclopropylmethyl, and X is 4) A is -NH 2 L is E is J is ethyl, Y is isobutyl, and X is A is -CH 3 L is -Cl, E and J are Y is isobutyl, and X is 6) A is -NH2, L is E is J is -Cl. Y is isoburyl, and X is 7) A is -NH 2 L is E is J i -Br, Y is isobutyl, and X is -CHzOCHz; and 8) A, L, E, and I arc -CH3, Y is cyclopropylmethyl, and X is Also especially preferred are compounds where A is -NH 2 L is E is J is bromopropyl, broaobutyl, chlorobutyl, eyclopropyl, hydroxypropyl, or N,Ndirnethylaminopropyl, and X is Indole and 9-Azaindole compounds of formula V are another preferred aspect:
A
L 0 E- U X 1 -P-oR E OR' J Y wherein; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:27 SPRUSON FERGUSON NO. 7146 P. 6/51' is selected from the group consisting of -Na and 1 1 D is selected from the group consisting-of- -Ca and Q is selected from the group consisting of and with the proviso that when B is -NH- then Q is and D is when B is -CH= then Q is and D is when S is then D is IlJ and Q is A. E. and L are selected from the group consisting of -NR8 2
-NO
2 -SR 7 -C(O)NR halo, -COR", -50 2 guanidine, amidine;
-NHSO
2
-SO
2 NR2, -CN, sulfaxide, perhaloacyl, perhaloalkyl, perhaloalkoxy, CI -CS alkyl, C2-CS alkcnyl, C2-CS alkynyl, and lower alicyclic, or together A and L form a cyclic group, or together L and E fom a cyclic group, or together E and J form a cyclic group including aryl, cyclic alkyl, and heterocyclic: J is selected from the group consisting of -NR, -NO 2 -SR7, -C(O)NR' 2 halo, -CN, sulfonyl, sulfoxide, perhaloalkyl, hydroxyalkyl, perhalcalkoxy, alkyl, haloalkyl, aminoalkyl, alkenyl, alkynyl, alicyclic, aryl, and aralkyl, or together with Y forms a cyclic group including aryl, cyclic alkyl and heterocyclic alkyl; X is selected from the group consisting of alkylamino, alkyl(hydroxy), alkyl(carboxyl), alkyl(phosphonate), alkyl, alkenyl, alkynyl, alkyl(sulfionate), aryl, carbonylalkyl, 1,1dibaloalkyl, alkylaminoalkyl, alkoxyalkyl, alkylthicalkyl, alkyithio, alkylaminavarbonyl, alkylcarbonylarnino, alicyclic, aralkyl, arid alkylaryl, all optionally substituted; or together with Y forms a cydlic group including aryl 1 cyclic alkyl,- and heterocyclic: COMS ID No: SMBI-00629627 Received by IP Australia; Time 18:45 Date 2004-02-20 FEB. 2004 18:27 SPRUSON FERGUSON NO. 7146-P. 51 Y is selected from the group consisting of alkyi, alkenyl, alkynyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl,
-C(O)-OR,
-CONHR -NR 2 and all except H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; Preferred A, L, and E groups include
-NO
2 hydroxy, halogen, -OR alkylaminocarbonyl,
-SR
7 lower perhaloalkyl, and C1-C5 alklyl, or together E and I form a cyclic group. Such a cyclic group may be aromatic or cyclic alkyl, and may be optionally substituted. Suitable aromatic groups include thiazole. Particularly preferred A, L and E groups are -NR 2 hydroxy, blogen, lower alkoxy, lower perhaloalkyl, and lower alkyl.
Preferred A groups include -NR 2 lower alkyl, halogen, and lower perhaloalkyl.
Preferred L and E groups include lower alkoxy, lower alkyl, and halogen.
Preferred J groups include halogen, lower alkyl,-lower bydroxyalkyl,
-NR
2 z, lower R'2N-alkyl, lower haloalkyl, lower perhaloalkyl, lower alkenyl, lower alkynyl, lower aryl, hoterocyclic, and alicyclic or together with Y forms a cyclic group. Such a cyclic group.may be aromatic or cyclic alkyl. and may be optionally substituted. Particularly preferred J groups -H, halogen, lower alkyl, lower hydroxyalkyl, -NRZ, lower R'zN-alkyl, lower haloalkyl, lower alkcnyl, alicyclic, and aryl.
Preferred X groups include alkyl, alkynyl, alkoxyalkyl, alkylthio, aryl, alkylaminocarbonyl, alkylcarbonylamino, 1,1-dihaloalkyl, carbonylalkyl, alkyl(OH), and alkyl(sulfonite). Particularly preferred is l,.1-dihaloalkyl, alkyl(sulfonate), alkylaminocarbonyl, alkoxyalkyl, and heteroaryl. Such compounds that are especially preferred are heteroaryl, alkylaminocarbonyl, and alkoxyalkyl. Most preferred is methylaminocarbonyl, methoxymethyl, and furanyl.
In one preferred aspect, X is not (C2-C3 alkyl)aminocarbonyl.
In one preferred aspect, when X is aIlcyl and alkene substituted with a phosphonic acid or ester, then A is and Y is not In another preferred aspect, X is not substituted with a phosphonic acid or ester.
Preferred Y groups include alkyl, aryl, aralkyl, and alicyclic, all except -H may be optionally substituted. Particularly preferred Y groups include lower alkyl, and alicyclic.
Preferred R 4 and R 7 groups include and lower alkyl.
In one preferred aspect, B Is NH, D is and Q is In another preferred aspect, B is D is and Q is COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:27 SPRUSON FERGUSON NO. 7146 P. 8/51-- 52 In another preferred aspect, A, L, and E are independendy
-NR'
2 lower alkyl, lower perhaloalkyl, lower alkoxy, halogen, -OH, or X is aryl, alkoxyalkyl, alkyl, alkylthio, 1,1dihaloalkyl, carbonylalkyl, alkyl(hydroxy), alkyl(sulfonate), alkylaminocarbonyl, and alkylcarbonylamino, and each R 4 and R 7 is independently or lower alkyl. Particularly preferred are such compounds where A, L, and E are independently lower alkyl, halogen, and -NR 3; J is halogen, haloalkyl, hydroxyalkyl,
-RY
2 N-alkyl, lower alkyl, lower aryl, heterocyclic, and alicyclic, or together with Y forms a cyclic group; and X is heteroaryl, alkylaminocarbonyl, 1,1-dihaloalkyl, and alkoxyalkyl. Especially preferred are such compounds where A is -NH 2 or -CH 3 L is -OCH 3 or E is or -Cl, J is halo, Cl- C5 hydroxyalkyl, CI-C5 haloalkyl, Cl-C5 R',N-alkyl, CI-CS alicyclic or CI-C5 alkyl, X is
-CH
z OCH2-, or 2 ,5-furanyl; and Y is lower alky. Preferred are such compounds where B is N, D is ,and Q is or where B is is and Q is Most preferred are compounds where: 1) A is -NHi, L is E is I is Y is isobutyl, and X is 2) A is -NH 2 L is E is J is -Cl, Y is isobutyl, and X is 3) A is L is E is -Cl, J is B is -NH, D is Q is and Y is isobutyl; and 4) A is -CHj, L is E is J is -HL B is D is Q is and Y is isobutyl.
Particularly preferred are such compounds where R' is -CHO0C(O)-C(CH,)3.
Another especially preferred aspect are such compounds where A, L, and E are lower alkyl. halogen, or -NR' 2 J is halogen, lower alkyl, lower ayl, heterocyclic, or alicyclic, or together with Y forms a cyclic group, and X is heteroaryl, alkylaminocarbonyl, or alkoxyalkyL For compounds where M is attached via a carbon atom it is also preferred when MH is selected from the group consisting of PMEA, PMEDAP, HPMPC, HPMPA. FPMPA, and
PMPA.
In another preferred aspect, MPO3 MP 2 O63-, or MPO3 9 is useful for the t.armnent of diseases of the liver or metabolic diseases where the liver is responsible for the overproduction of a biochemical end product. Preferably, such disease of the liver is selected from the group consisting of hepatitis, cancer, fibrosis, malaria, gallstones, and chronic cholecystalithiasis. It is more preferred when treating such diseases that MH. MPOa', MP20s 3 or MP 3 09' is an antiviral or anticancer agent.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:28 SPRUSON FERGUSON NO. 7146 P. 9/51 53 Preferably, the metabolic disease that MPO 2 MPa20 or MP 3 0 4 are useful for diabetes, atherosclerosis, and obesity.
In another aspect, it is preferred when the biochemical end product is selected from the group consisting of glucose, cholesterol, fatty acids, and triglycerides. More preferred is when MH or MPO, 2 is an AMP activated protein kinase activator.
In another aspect, it is preferred when M .PO 3 2 is a compound that inhibits human liver FBPase. It is more preferred when such FBPase inhibitor inhibits human liver FBPase with an ICjo of less than I 0M. More preferred are such FBPase inhibitors wherein M is a group T-X wherein T is selected from the group consisting ofbeczimidazole, indole, purine, and 9-azaindole, all of which contain at least one substituent; X is attached at the 2, 2, 8, and 2 positions of said T groups, respectively; and X is selected from the group consisting of alkylamino, alkyl, alkenyl, alkynyl, alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonate), alkyl(sulfonate), aryl, alkylaminoalkyl.
alkoxyalkyl, alkylthioalkyl, alkylthio, alicyclic, 1,1-dihaloalkyl, carbonylalkyl, alkylaminocarbonyl, alkylcarbonylamino, aralkyl, and alkylaryl, all optionally substituted- In compounds of formula I, preferably V is selected from the group consisting of aryl, substitued aryl, heteroaryl, substituted heteroaryl; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing one hetcroatom, that is fused to an aryl group attached at the beta and gamma position to the oxygen attached to the phosphorus or together V and W are connected via 3 carbon atoms to form a cyclic group containing 6 carbon atoms mono-substituted with a substituent selected from the group consisting of hydroxyl, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy and aryloxycarbonyloxy three atoms from an oxygen attached to the phosphorus. More preferred is when V is selected from the group consisting of aryl, substitued aryl, heteroaryl, and substituted heteroaryL Especially preferred is when V is phenyl, or substituted phenyl. V is 3-bromophenyl is a particularly preferred substituted phenyl.
It is also especially preferred when V is selected from the group consisting of heteroaryl and substituted heteroaryl.
Most preferred is when such heteroaryl is 4-pyridyl.
It is also preferred when together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:28 SPRUSON FERGUSON NO. 7146 P. 10/51 54 positions to the oxygen attached to phosphorus. In such compounds preferably said aryl group is an optionally substituted monocyclic aryl group and the connection between Z and the gamma position of the aryl group is selected from the group consisting of O, CH2,CHzCH 2
OCH
2 or
CH
2 0.
In another aspect, it is preferred when together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and monosubstituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkosycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy three atoms from an oxygen attached to the phosphorus. In such compounds, it is more preferred when together V and W form a cyclic group selected from the group consisting of-CHz-CH(OH)-CH 2 CHiCH(OCOR)-CHa-, and -CH2CH(OCOz)R)-CH-.
Another preferred V group is 1-alkene. Oxidation by p450 enzymes is known to occur at benzylic and allylic carbons.
In one aspect, preferred groups include -CHR 2 OH, -CHOCOR', and -CHOCO 2
R'.
In another aspect, preferred Z groups include-OR 2
-SR
2
-CHR
2
N
3
-R
2
NR
2
-OCOR
2 -OCOIzR, -SCOR', -SCO2R, -NHCOR 2
-NHCO
2
R',
-CH
2 NHaryl, -(CH2),-OR 2 and SR'. More preferred Z groups include-OR 2
-R
2 -OCOR', -OCO2R, -NHCOR', -NHCOR', -(CH2),-OR 2 and, .(CH 2
)-SR
2 Most preferred Z groups include -OR 2 -H -OCOR 2
-OCO
2 and -NHCOR 2 Preferred W and W' groups include H, R 3 aryl, substituted aryl, heteroaryl, and substituted aryl. Preferably, W and W' are the same group. More preferred is when W and W' are H.
In one aspect, prodrugs of formula VI are preferred: 0wherein V is selected from the group consisting of aryl, substituted aryl, heteroaryl, and substituted aryl, 1-alkenyl, and 1-alkynyl. More preferred V gropus of fornula VI are aryl, substituted, heteroaryl, and substituted het-oaryl. Particularly preferred aryl and substituted aryl COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:29 SPRUSON FERGUSON.
NO. 7146 P. 11/51 groups include phenyl and substituted phenyl. Particularly preferred heteroaryl groups include monocyclic substiutted andunsubstituted heteroaryl groups. Especially preferred arc pyridyl and 3-bromopyridyl.
In another aspect, prodrugs of formula VII are preferrd: Z ooZ
H
wherein Z is selected from the group consisting of:
-CHR
2 OH, -CHR'OCOR', -CHR'OC(S)R',
-CHRZOCO
2 R, -CHRzOC(O)SR', -CHR'OC(S)OR',
-SR
z and -CH 2 aryl. More preferred groups include -CHR 2
OH,
-CHR'OC(O)R', and -CHR OCO0R'.
Also preferred are compounds of formula VII when M is attached to the phosphorus via a carbon or oxygen atom.
In another aspect, prodrugs of formula VIII are preferred: 1 5 v m I wherein Z' is selected from the group consisting of-OH, -OCO, R 3 and -OC(O)S R'; D' and D" are independently selected from the group consisting of alkyl, OR 2
-OH,
and -OC(O)R 3 with the proviso that at least one of D' and D" are -H.
In one preferred embodiment, W' and Z are W and V are both the same aryl, substituted aryl, hetcroaryl, or substitutmced heteroaryl such that the phosphonate prodrug moietyr COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:29 SPRUSON FERGUSON NO. 7146 P. 12/51 56
V
W
has a plane of symmetry.
In another preferred embodiment, W and W' are H, V is selected from the group consisting of aryl, substituted aryl, heteroaryl, substituted heteroaryl, and Z is selected from the group consisting of OR 2 and -NHCOR 2 More preferred are such compounds where Z is -H.
Preferably, such compound have M attached via oxygen. Most preferred are such compounds where oxygen is in a primary hydroxyl group.
Also more preferred, are those compounds where V is phenyl or substituted phenyl.
Most preferred are such compounds where said oxygen is in a primary hydroxyl group.
Preferably, such compounds have M attached via oxygen.
Also more preferred, are those compounds where V is an optionally substituted monocyclic heteroaryl containing at least one nitrogen atom. Preferably such compounds have M attached via oxygen. Most preferred are such compounds where said oxygen is in a primary hydroxyl group. Especially preferred are such compounds where V is 4-pyridyl, In these compounds it is also preferred when MH is selected from the group consisting of araA, AZT, d4T, ddl, ddA. ddC, L-ddC, L-FddC, L-d4C. L-Fd4C, 3TC, ribavirin, penciclovir, fluoro-2'-deoxyuridine, FIAU, FIAC, BHCG, 2'R,$'S(-)-l-(2-(hydroxymethyl)oxathiolan-.
yl]cycosine, (-)-b-L-2',3'-dideoxycytidine, (-)-b-L-2',3'-dideoxy-5-fluorocytidine,
FMAU,
BvaraU, E-5-(2-bromovinyl)-2'-deoxyuridine, Cobucavir, TFT, CDG, DAPD, FDOC, d4C, DXG, FEAU, FLG, FLT, FTC, 5-yl-carbocyclic 2'-deoxyguanosine, Cytallene, Oxetanocin A. Oxetanocin 0, Cyclobut A, Cyclobut G. fluorodeoxyuridine, dFdC, araC, bromodeoxyuridine, IDU, CdA, F-ara-A, 5-FdUMP, coformycin, and 2'-deoxycoformycin.
Particularly preferred are such compounds where V is selected from the group consisting of phenyl and 4-pyridyl and MH is selected from the group consisting of ribavirin, AZT,, penciclovir, araA, 5-fluoro-2'-deoxyuridine, ddl, ddA, ddC, and F-araA.
Also preferred is when MH is selected from the group consisting of ACV, GCV, 9-(4hydroxy-3-hydroxymethylbut-1-yl)guanine, and (R)-9-(3,4-dihydroxybutyl)guanine.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:29 SPRUSON FERGUSON N0.7146 P. 13/51 57 When W' and W are H, V is aryl, substituted aryl, heteroaryl, or substituted heteroaryl, and Z is H, OR, or -NHCOR 2 it is also preferred when M is attached to the phosphorus via a carbon atom. Preferred are such compounds wherein MPOj2 is selected from the group consisting of phosphonoformic acid, and phosphonoacetic acid. Also preferred are MH is selected from the group consisting of PMEA, PMEDAP, HPMPC, HPMPA, FPMPA, and
PMPA.
In these compounds it is also preferred when M is selected from the group consisiting of:
A
J L
II
wherein E is selected from the group consisting of alkyl, anmino or halogen; L and J are independently selected from the group consisting of hydrogen, hydroxy, acyloxy, alkoxycarbonyloxy, or when taken together form a lower cyclic ring containing at least one oxygen; and A is selected from the group consisting of amino and lower alkylamino; and pharmaceutically acceptable prodrugs and salts thereof.
In another preferred aspect, these compounds where M is selected from the group consisiting of:
Y
III
wherein A is selected from the group consisting of -NR 2, NHSO z
R',
-SR' halogen, lower alkyl, -CON(Rt2, guanidine, amidine, and perhaloalkyl; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:29 SPRUSON FERGUSON NO. 7146 P. 14/51 58 E is selected from the group consisting of-H, halogen, lower alkylthio, lower perhaloalkyl, lower alkyl, lower alkcenyl. lower alkynyl, lower alkoxy, -CN, and -NR 7 2; X is selected from the group consisting of alkylamino. alkyl, alkenyl, alkyny!, alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonae), alkyl(sulfonate), aryl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthio, alicydlic, 1,1 -dihaloalkyl, carbonylalkyl, aminocarbonylamino, alkylaminocarbonyl, alkylcarbonylamino, aralkyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including cyclic alkyl, heterocyclic, and aryl; Y is selected from the group consisting of-H, alkyl, alkcnyl, alkynyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl, -C(O)R 3
-C(O)-ORI,
-CONHR -NR22, and -OR 3 all except H arc optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic;
R
4 is independently seclected from the group consisting of-H. lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; R' is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic;
R
6 is independently selected from the group consisting of-H, and lower alkyl; R is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, lower aryl. and -C(0)RI0; R' is independently selected from the group consisting of lower alkyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R' 0 or together they form a bidenrate alkyl; R 0 is selected from the group consisting of lower alkyl, -NH2, lower aryl, and lower perhaloalkyl; RI is selected from the group consisting of alkyl, aryl, -OH, -NH2 and
-OR
3 and phanraceutically acceptable prodrugs and salts thereof.
In another preferred aspect, those compounds where M is a compound of formula IV: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:30 SPRUSON FERGUSON NO. 7146 P. 15/51 59 j Y
IV
wherein: A. and L are selected from the group consisting of
-NR
8 2, -NO2, -OR 7
-SR
7 -C(O)NR42, halo, -COR I 1 -SO2R, guanidinc, amidine, -NH50 2
R
5 -SO2NR42, -CN, sulfoxide, perhaloacyl, perhaloalkyl, perhaloalkoxy, Cl-C5 alkyl, alkenyl, C2-C5 alkynyl, and lower alicyclic, or together A and L form a cyclic group, or together L and E form a cyclic group, or together E and J form a cyclic group including aryl, cyclic alkyl, and heterocyclic; J is selected from the group consisting of -NR 8 2, -NO2,
-OR
7
-SR
7
-C(O)NR
4 2, halo, -C(O)R 1 1 -CN, sulfonyl, sulfoxide, perhaloalkyl, hydroxyalkyl, perhaloalkoxy. alkyl, haloalkyl, aminoalkyl, alkenyl, alkynyl, alicyclic, aryl, and aralkyl, or together with Y forms a cyclic group including aryl, cyclic alkyl and hetcroyclic alkyl; X is selected from the group consisting of alkylamino, alkyl(hydroxy), alkyl(carboxyl), alkyl(phosphonate), alkyl, alkenyl, alkynyl, alkyl(sulfonalte) aryl, carbonylalkyl, 1,1dihaloalkyl, aminocarbonylamino, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthio, alkylarninocarbonyl, alkylcarbonylamino, alicyclic, aralkyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; Y is selected from the group consisting of-H, alkyl, alkenyl, alkynyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkaxyalkyl, -C(O)R 3 -S(0) 2 R, -CONHR', -NR 2 2, and -OR, all except -H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic;
R
4 is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:30 SPRUSON FERGUSON NO. 7146 P. 16/51 R' is sriected from the grou 2 consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic;
R
6 is independently selected from the group consisting of-H, and lower alkyl;
R
7 is independently selected from the group consisting of H, lower alkyl, lower alicyclic, lower aralkyl, lower aryl, and -C(0)R10; R' is independently selected from the group consisting of-H, lower alkyl, lower aralkyl, lower aryl, lower alicyclic.,-C(O)R'°, or together they form a bidencate alkyl; R is selected from the group consisting of-H, lower alkyl, -N-H2, lower aryl, and lower perhaloalkyl; R is selected from the group consisting of alkyl, aryl, -OH, -NH 2 and and pharmaccutically acceptable prodrugs and salts thereof; with the provisos that: a) when X is alkyl or alkene, then A is -NR 2; b) X is not alkylamine and alkylaminoalkyl when an alkyl moiety is substituted with phosphonic esters and acids; and c) A, L, E, J. Y, and X together may only form 0-2 cyclic groups.
Preferably, oral bioavailability is at least More preferably, oral bioavailability is at leasr Preferred A groups include -NR 8 2, lower alkyl, lower perhaloalkyl, lower alkoxy, and halogen. Particularly preferred are -NR 8 2, and halogen. Especially preferred is -NR 8 2. Most preferred is -NH2.
Preferred E groups include halogen, lower perhaloilkyl, -CN, lower alkyl, lower alkoxy, and lower alkylthio. Particularly preferred E groups include -SMe, -Et, and -Cl. Especially preferred is -H and -SCH3, Preferred X groups include alkylamino, alkyl, alkynyl, alkoxyalkyl, alkylthio, aryl, 1,1dihaloalkyl, carbonylalkyl, heteroaryl, alkylcarbonylamino, and alkylaminocarbonyl, Particularly preferred is alkyl substituted with I to 3 substituents selected from halogen, phosphonate, -CO2H, -SO3H, and -OH. Particularly preferred are alkylaminocarbonyl, alkoxyalkyl, and heteroaryl. Preferred alkoxyalkyl groups include methoxymcthyl. Preferred COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:30 SPRUSON FERGUSON 26.FEB 204 8:0 SRUON FRGUONNO. 7146 P. 17/5 F 61 hereroaryl groups include filranyl, optionally substituted.
Preferred Y groups include aralicyl, alicyc tic, alkyl; and aryl, ali optionally substituted.
Particulofy preferd is lower alkyl. Particularly preferred Y groups include (2naphthyl)mncthyl, cyclohcxylezhyl, pheuylethyl, nonyl, cyclohexylpropyl, ethyl, cyclopropylmethyj, cyclobutyirnethylphcnyl, 2 -methyl)propyl, neopentyl, cyclopropyl, cyclopcruyl, (1 -imidozolyl)propyt, 2-ethoxybenzyl, I -bydroxy-2,2.dimethylpropyL I -chloro.-2,2.
dimethyipropyl, 2,2-dimethylbutyl, 2-(spiro-3 ,3 -dirnethykcyclobex.4-enyl)propyl, and Imethylneopenryl. Especially preferred is neopentyl and isobuzyl.
Preferred R 4 and R 7 groups are and lower alkyl. Particularly preferred are and methyl.
in another preferred aspect, A is -NR 8 2 or halogen, E is halogen, -CN, lower alkyl, tower perhaloalkyl, lower alkoxy, or lower ulkylthio, X is alkylamino, alkyl, alkoxyalkyl, alkynyl, 1,1 -dihaloalkyl, carbonylakyl, alkyl(OH), alkyl(sutfonate), allcylcarbonyiarnino, alklandnocarbonyl, alkyithia, uzyl, or hereroaiyl, and R 4 and R7 is -H or lower. alkyi.
Particularly preferred ame such compounds where Y is aralkyl, aryl, alicyclic, or alkyl.
In another preferrd aspect A is-R2 E is Ck. or methyithia. anid X is optionally substituted fbrmnyl, or ulkoxyalkyL. Particularly preferred am such coumunds where A is -14HZ.
X is 2,5-furanyl, or methoxymethyl, and Y is lower alkyl. Most preferred are such compounds where E is H, X is 2,5-ftirnyl, and Y is neopentyl; those where E is -SCH3, X is and Y is isabulyl; and those where E is X is 2,5-furmnyl, and Y is l-(3-chiorb-2,2-dimcrbyl).
prapyl.
In one aspect, the compounds of formula VI preferably have a group Z which is H, alkyl, alicyclic, hydroxy, alkoxy, 0 0 11 11 OCR, OCOP, amino, or NHCOR. Preferred are such groups in which Z decreases the propensity of the byproduct, vinyl aryl ketone io undergo Michael reactions. Preferred Z groups are groups that donate electrons to the vinyl group which is a known strategy for decreasing the propensity of a.-unsaturated carbontyl compounds to undergo a Michael addition. For eamiple, a methyl group in a similar position on aczylamide results in no mutagenic activity whereas the unsubsituted vinyl analog is highly mutagenic. Other groups could serve a similar fulnction, e.g.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB-2004 18:31 SPRUSON FERGUSON NO. 7146 P. 18/51 62 Z!OR, NHAc, ecc. Other groups may also prevent the Michael addition especially groups that result in removal of the double bond altogether such as Z OH, OR', NH2, which will rapidly undergo retautomcrizacion after the elimination reaction. Certain W and W' groups are also advantageous in this role since the group(s) impede the addition reaction to the A-carbon or destabilize the product. Another preferred Z group is one that contains a nucleophilic group capable of adding to the a,1-unsaturated double bond after the elimination reaction i.e. (CH 2 SH or (CH2).OH where p is 2 or 3.
p450 oxidation can be sensitive to stereochemistry which might either be at phosphorus or at the carbon bearing the aromatic group. The prodrugs of the present invention have two isomeric forms around the phosphorus. Preferred is the stereochemistry that enables both oxidation and the elimination reactiorn Preferred is the cis stereochemistry. In contrast, the reaction is relatively insensitive to the group M since cleavage occurred with a variety of phosphonatc, phosphate and phosphoramidates. Accordingly, the group M represents a group that as part of a compound of formula 1 enables generation of a biologically active compound in vivo via conversion to the corresponding M-P0 3 The atom in M attached to phosphorus may be 0, C or N. The active drug may be M-PO3" or a metabolite of M-POj" such as the triphosphate useful for treatment of diseases in which the liver is a target organ, including diabetes, hepatitis, liver cancer, liver fibrosis, malaria and metabolic diseases where the liver is responsible for the overproduction of a biochemical end products such as glucose (diabetes), cholesterol, fatty acids and triglycerides (atherosclerosis). Moreover,
M-PO
3 may be useful in treating diseases where the target is outside the liver but accessible to a phosph(on)ate.
Preferred M groups are groups in which M is a nucleoside and the phosphate is attached to a hydroxyl, preferably a primary hydroxyl on a sugar or sugar-analog. Especially preferred M groups include araA, AZT, d4T, ddl, ddA, ddC, L-ddC, L-FddC, L-d4C, L-Fd4C, 3TC, ribavirin, penciclovir, 5-fluoro-2'-dcoxyuridine, FIAU, FIAC, BHCG, (-)-b-L-2',3'-dideoxycytidine, (-)-b-L-2',3'-dideoxy- FMAU, BvaraU, E-5-( 2 -bromovinyl)-2'-deoxyuridine, Cobucavir, TFT, propynyl-1-arabinosyluracil, CDG, DAPD, FDOC. d4C, DXG, FEAU, FLO, FLT, FTC, carbocyclic 2'-deoxyguanosine, Cytallene, Oxetanocin A, Oxetanocin G, Cyclobut A, Cyclobut G, fluorodeoxyuridine, dFdC, araC, bromodeoxyuridine, IDU, CdA, F-aaA, Coformycin, 2'-dcoxycoformycin, PMEA, PMEDAP, HPMPC, HPMPA, FPMPA, and PMPA.
Other preferred M groups include phosphonic acids useful in treating diabetes, viral infections, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:31 SPRUSON FERGUSON NO.7146 P. 19/51 63 liver fibrosis, e.g. collagenase inhibitors such as reported in Bird ct aL, J. Med. Chem. 37, 158- 169 (1994), parasitic infections, diseases responsive to metalloprotease inhibition (e.g.
hypertension, liver, cancer), and hyperlipidemia.
The preferred compounds of formula Vm utilize a Z' group that is capable of undergoing an oxidative reaction that yields an unstable intermediate which via elimination reactions breaksdown to the corresponding
M-PO
3 Especially preferred Z' groups is OH. Groups D and D' are preferably hydrogen, alkyl, hydroxy, and -OR 2
-CCOR
3 but at least one of D or D' must be H.
The following prodrug moieties are preferred: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB-2004 18:31 SPRUSON FERGUSON NO. 7146 P. 20/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:31 SPRUSON FERGUSON NO, 7146 P. 21/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20,FEB-2004 16:32 SPRUSON FERGUSON 20.FE. 204181 SRUSN FEGUONNO. 7146' P. 22/5P- COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB-2004 18:32 SPRUSON FERGUSON -NO. 7146-P, 23/51- COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:32 SPRUSON FERGUSON NO. 7146 P. 24/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:32 SPRUSON FERGUSON NO.7146 P. 25/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB.2004 18:32 SPRUSON FERGUSON -NO, 7146' P. 26/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:33 SPRUSON FERGUSON S-NO. 7146 27/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:33 SPRUSON FERGUSON NO. 7146 P. 28/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:33 SPRUSON FERGUSON NO. 7146- P. 29/51 73 SYNT)j sy nOheis QBonahp opts fl SYNTHESIS OF PROnDRUG; The following rocedures n the preparation of prodrugs illustrate the general procedures used ho prepare the prontadrgs of the nv on which apply to all phosphate, phosphonate. and posphornate. ozain drugs. Prodrngs can be introduced at differen stages of synthesis of a drug. Most often they are made at a later stage, because of the general sensitivity of these groups to various reaction conditions, Optically pure prodrugs h onn g single isomer at Phosphorus Centre can be made ei the p ure prodrugs containing single isomer It phophoru c re can be made either by eparation of the diastermers by a combination of column chromatography ad/or crytallyzation, or by enautioseleczivo synthesis of chira activated phosph(on)ue intermediates.
The preparation of prodru s is further orgaize into 1) synthesis via activated
P(V)
intermediates:, 2) synthesis via activated P(fl) incerediates, 3) synthesis via phosph(on)ate diacid, and 4) miscellaneous methods.
V V H V i.0 H O_0.H0
V
Z tz H L
HH
W 0 1.1 Synthesis via activated P(V) intermediate:
W
I. l.a. Synthesis of activated P(V) intermediates: In general, synthesis of phosph(on)ate esters is achieved by coupling the amino or alcohol MH wit the corresponding activated phosphonate precursor for example, Chlorophosphonate (L'-chloro) addition on to 5'-hydroxy of nucleoside is a well known method for preparation of nucleoside phosphate monoeste. e activated precu r c be a prepared by several well known methods. Chlorophosphotates useful for synthesis of the prodrugs are prepared from the COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:33 SPRUSON FERGUSON NO, 7146 P. 30/51 74 substituted. 1,-propanediol (Wissner, et J. Aed Cheu., 1992, 35, 650). Chlorophosphonates are made by oidaion o te corresponding chlorophpoholane (Anderson, t aj, J 0r. Chem:, 1984, 49, 1304) which are obtained by reaction of the substituted diol with phosphorus trichloride. Alternativey, the chlorophosphonate agent is made by treating substituted-l.,3diols Swith phosphorusoxycoride (Patois, et If, J Chem Perin Trn. I. 1990, 1577). Chlorophosphonate species may also be generated in situ from corresponding cyclic phosphites (Silverburg, et al., Tetrahedron lett. 1996, 37, 771), which in turn an be either made from chlorophospholane or phosphoramidate intermediate. Phosphoroflouridae inrmediate prepared either from pYrophosphate or phosphoric acid may also act as preursaor in preparation of cyclic prodrugs (Watanabe at al., Tetrahedron lett., 1988, 29, 5763).
Phosphoramidates are also well known intermediates for the synthesis of phosphate ester. Monoalkyl or dialkyphosphora ndate (Watanabe, ct al, Chem Ptharm Biul 1990, 38, 562), triazolophosphora nmidat e akage, r aL, Tetrahedron ,1989, 45, 5459) and PYrrolidihopho horamidate (Nakayama, eC al2, Am. Chm. Soco, 1990, 112, 6936) are some of the known intermediates used for the preparation of phosphate esters. Another effective phosphorylating procedure is a metal catalyzed addition of cyclic chlorophosphonate adduct of 2 -oxazolone, This intermediate attains high selectivity in phosphorylarion of primary hydroxy 237).oup in presence of secondary hydroyl group (Nagamatsu, at al, Terrahedron Lett., 1987, 28, 2375). These agents arc obtained by reaction of a chlorophosphonate with the amine or alternatively by formation of the corresponding phosphoramidite followed by oxidation.
I1..b. Synthesis of chiral activated phosph(on)ate: Phosphorylation of an enandmiomerically pure substituted diol with tfor example, a commercially available phosphorodichloridate R-OP(O)C1 2 whereRO is a leaving group, preferably aryl substituted with electron withdrawing groups, such as a nitro or a chloro, produces two diasterconeric intermediates that can be separated by a combination otcolmn chromatography and/or crystallization. Such a method may also be utilized in preparing chiral chloro phosphonates. Chiral Phosporamidat intermediates can be obtained by utilization of optically pure amine as the chin auxiliary. This type of intermediate ate known to undergo stereospecifi substitution (Nakayama, t t. Am. Chem. Soc.,199, 112, 6936). The relative confgu on of the phosphorus atom is easily determined by comparison of the 3 1 P NMR spectra. The chemical shift of the equatorial phosphoryloxy moiety (trans-isomer) is always COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:34 SPRUSON FERGUSON NO. 7146-P. 31/51 more upfield than the one of the axial isomer (cis-isom) (Vcade, et al, Or. Chem. 1977, 42, 1549), (Verbde et al Org. Chem.. 1977, aIz, A Cis Trans 1. Synthesis of Prodrugs Using Activated phosphates.
Coupling of activated phosphonates with alcohols or anes is accomplished in the presence of an organic base. For example, Chlorophosphonates synhesie as described in the earlier section react with an alcohol in the Presence of a base such as pyidincs or Nmethylinid.ole. In some cases phosphavlaion is enhanced by in situ generation of iodophosphonate from chloro (Stomberg, et al, Nucleosides Nuclcotide. 1987, 5: Phosphoroflouridate intermediates have also been used in phosphoryladtion reactions in the presence of a base such as CsF.or n-BuLi to generate cyclic prodrugs (Watanabe et al, Tetrahedron lea., 1988, 29, 5763). Phosphortamnidate intermediates are shown to couple in the presence of weak acids t'trazole) and an oxidising agent (eg.s, m-chloroperbenzoic acid or Ibutylhydropcroxide (Watanabe, et al, Chem Pharm Bull., 1990, 38, 562) or by transition metal catalysis (Nagamnacsu, et al, Terrahedreon Lett., 1987, 28, 2375).
The phosphonate prodnrug esters where spacer group X in formula II-IV is an aryl group, can be prepared by lithiation of aromatic ring using methods well described in literature (Gschwend, Org. React. 1979, 26, 1; Durn, Cbmprehenive Carbanion Chemistry Vol. Elsevier, New York, 1984) followed by addition of chlorophosphonat 0 cyclic .1',3'-propanyl ester.
Reaction of the optically pure diastereomer of phosphoramidate intermediate with the hydroxyl of drug in the presence of an acid produces the optically pure phosphate prodrug by direct SN2(P) reaction (Nakayama, et al. J Am. Chem. Soc.,1990, 112 6936). Alternatively, reaction of the optically pure phosphate precursor with a fluoride source, preferably cesium fluoride or teuaburylammorium fluoride, produces the more reactive phosphorofluoridate which reacts with the hydroxyl of the drug to give the optically pure prodrug by overall retention of configuration at the phosphorus atom (Ogilvie, et a, J Am. Chem. Soc.,1977, 99, 1277). Chial phosphonate prodrugs can be synthesized by either resolution of phosphonates (Poganic, et al., COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:34 SPRUSON FERGUSON NO. 7146 P. 32/51 76 Terrahedro Len., 1997, M, 3495) or by chbirality induction (Taapken, et. al.. Tetrahedron Lett., 1995, 36, 6659, J Org. Chem., 1998, 63, 8284).
1.2 Synthesis via phosphite intermcdiate: v V
V
z L'-P u X z WW W 2. [O ww 1.2.a. Synthesis of activated P(III) intermediates: Phosphorylation of hydroxy- and amino groups is achieved using cyclic l',3'-propanyl esters of phosphorylating agents where the agent is at the P(Ill) oxidation state. One preferred phosphorylating agent is a chloro phospholane (L'=chloro). Cyclic chlorophospholanes are prepared under mild conditions by reaction of phosphorus trichloride with substituted 1,3-diols (Wissner, et al, J. Med. Chem., 1992, 35, 1650). Alternatively phosphoramidites can be used as the phosphorylating agent (Beaucage, et al., Tetrahedron, 1993, 49, 6123). Appropriately substituted phoshoramidites can be prepared by reacting cyclic chlorophospholanes with NNdialkylamine (Pcrich, ec al., Aust. J. Chem.. 1990, 43, 1623. Perich, et at, Synthesis, 1988, 2, 142) or by reaction of commercially available dialkylaminophosphorochloridate with substituted propyl-1,3-diols.
1.2.b. Synthesis of chiral activated P(M) intermediate: In the cases where unsymmetrical diols are used, the cyclic phosphite is expected to form a mixture of chiral isomers. When an optically active pure diol is used a chromatographically separable mixture of two stable diastereomers with the leaving group (NRR') axial and equatorial on the phosphorous atom is expected. Pure diasteromers can usually be obtained by chromatographic seperation.
I.2.c. Synthesis of prodrugs Using Activated Phosphites: Chlorophospholanes are used to phosphorylate alcohols on nucleosides in the presence of an organic base (e.g.,tricthylamine, pyridine). Alternatively, the phosphite can be obtained by coupling the nucleoside with a phosphoramidate in the presence of a coupling promoter such as tetrazole or benzimidazolium triflate (Hayakawa at J. Org. Chem., 1996, 61, 7996).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:34 SPRUSON FERGUSON NO. 7146 P. 33/51 77 Phosphite diastereomcrs may be isolated by column chromatography or crystallization (Wang, et al, Tetrahedron Lert, 1997, 38, 3797; Bcntridge et al., J Am. Chem. Soc., 1989, 111, 3981).
Since condensation of alcohols with chlorophospholanes or phosphoramidites is an SN2(P) reaction, the product is expected to have an inverted configuration. This allows for the sicrcoselective synthesis of cyclic phosphites.
The resulting phosphites are subsequently oxidized to the corresponding phosphate prodrugs using an oxidant such as molecular oxygen or t-buylhydroperoxide (Meier et al., Bioorg, Med. Chem. Lett., 1997, 7, 1577). Oxidation of optically pure phosphites is expected to stercoselectively provide optically active prodrugs (Mikolajczyk, ae al., J. Org. Chem., 1978, 43, 2132. Cullis, P. M. J. Chem. Soc.. Chem Commun., 1984, 1510, Verfurth, et al., Chem. Ber., 1991, 129, 1627).
1.3 *Synthesis of Phosphonate Prodrugs via Phosphonic Acids:
V
ww 9 H 0 L W W 0 M "OH M 'L M
WW"
Prodrugs of fbnnula I are synthesized by reaction of the corresponding phosphodichloridatc and an alcohol (Kharnmel, et J. Med. Chem., 1996, 39 4109). For example, the reaction of a phosphodichloridatle with substituted 1,3-diols in the presence of base (such as pyridine, triethylamine, etc) yields compounds of formula I.
Such reactive dichloridats intermediates, can be prepared from the corresponding acids and the chlorinating agents e.g. thionyl chloride (Starrett, et al, J. Med. Chem., 1994, 1857), oxalyl chloride (Stowell, et al, Tetrahedron Lett., 1990, 31; 3261), and phosphorus pentachloride (Quast, et a, Synthesis, 1974, 490). Alternatively, these dichlorophosphonates can also be generated from disilyl esters (Bhongle, et al, Synth. Commun., 1987, 17: 1071) and dialkyl esters (Still, et at, Tetrahedron Let., 1983, 24: 4405; Patois, et al, Bull Soc. Chim: Fr., 1993, 130: 485).
1.4, Miscellaneous Methods: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:35 SPRUSON FERGUSON NO. 7146 P, 34/51-' 78 Phosph(on)ace prodrugs are also prepared from the free acid by Mitsunobu reactions (Mitsunobu, Synthesis, 1981, 1; Campbell, J.0rg. Chem., 1992, 52: 6331), and other acid coupling reagents including, but not limited to, carbodiimids (Alexander, et al, Collect. Czech.
Chem. Commun., 1994, 59: 1853; Casara, et a, Bioorg. Med. Chem. Letr, 1992, 2: 145; Ohashi, er al, Tetrahedron 1988, 29: 1189), and benzotriazolyloxytris- (dimethylamino)phosphonium salts (Campagne, oe al, Tetrahedron Lett., 1993, 34: 6743).
Phosphorylation of an alcohol or an amine is also achieved under Mitsunobu reaction conditidns using the cytlic 1'3'-propanyl ester of phosphoric acid in the presence of triphenylphosphine and diethylazodicarboxylate (Kimura et al., Bull. Chem. Soc. Jpn-, 1979, 52, 1191). The procedure can be extended to prepare chiral phosphates from enantiomerically pure phosphoric acids.
Phosph(on)ate prodrugs can be prepared by an alkylation reaction between the phosphonate corresponding tetrabutylamnmonium salts and substituted. 1 ,3-diiodo propanes made from 1,3-diols (Farquhar, et al, Tetrahedron Lett.. 1995 36. 655). Furthermore, phosphate prodrugs can be made by conversion of nucleoside to the dichloridate intermediate with phosphoryl chloride in presence of triethylphosphite and quenching' with substituted-1,3propane diols(Farquhar ct al., J. Org. Chem., 1983, 26, 1153).
Phosphorylation can also be achieved by making the mixed anhydride of the cyclic diester of phosphoric acid and a sulfonyl chloride, preferably 5-quinolinesulfonyl chloride, and reacting the hydroxyl of the drug in the presence of a base, preferably mcthylimidazole (Takaku.
et al, J. Org. Chem., 1982, 47, 4937). In addition, starting from a chiral cyclic diester of a phosphoric acid, obtained by chiral resolution (Wynberg, el al., J Org. Chem., 1985. 50, 4508), one can obtain optically pure phosphates.
Aryl halides undergo Ni 2 catalyzed reaction with phosphite derivatives to give aryl phosphonate containing compounds (Balthazar, aet al. 1 Org. Chem.. 1980, 45: 5425).
Phosphonates are also prepared from the chlorophosphonate in the presence of a palladium catalyst using aromatic triflates (Petrakis, et al, J. Am. Chaem. Soc., 1987, 109: 2831; Lu, et al.
Synthesis, 1987, 726). In another method, aryl phosphonatc esters are prepared from aryl phosphates under anionic rearrangement conditions (Melvin, Tetrahedron Lett, 1981, 22: 3375; Casteel, et al. Syntheiis, 1991, 691). N-Alkoxy aryl salts with alkali metal derivatives of cyclic alkyl phosphonate provide general synthesis for hetcroaryl-2-phosphonate linkers (Redmore, J.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:35 SPRUSON FERGUSON NO, 7146 P. 35/51 79 Org. Chem., 1970, 35: 4114). These above mentioned methods can also be extended to compounds where the X group is Eeteroaryl e.g. pyridine, furan, chiophene etc.
Compounds of formulae 1 IV, where X is alkyl. substituted alkyl or heteroalkyl are synthesized using well known reactions. For example, when X is substituted with a leaving group halogen) an Arbuzov reaction with a phosphite containing cyclic 1 ',3'-propanyl ester is useful (Chem. Rev. 1984, 84: 577). Cyclic alkyl phosphites also attack the lactones at the 6carbon atom, causing the alkyl-oxygen cleavage of the lactone ring, to yield alkyl phosphonate esters. This can be appliqd to many types of lactones such as -lactones, y-lactones etc. as reported by McConnell et al, J. Am. Chem. Soc., 1956, 78, 4453. Alternatively. compounds wherein X is alkyl heteroatom can be prepared by alkylation of htcro atom with an appropriate cyclic phosphonate electrophile [L(CH2)nPO3R] where L is a leaving group, preferably iodide (Walsh et al, Am. Chem. Soc., 1956, 78, 4455). These above mentioned methods can be extended to the heteroalkyl linkers e.g. -CH2ZCH 2 where Z ,S etc.
IL)1 SYNTHESIS OF 1.3-DIOLS: A variety of synthetic methods are known to prepare the following types of 1,3-dials: a) 1-substituted; b) 2-substituted; and c) 1,2- or 1, 3 -annulated in their recemic or chiral form.
Substitution of V, W, Z groups of formula I, can be introduced or modified either during synthesis of diols or after the synthesis of prodrugs.
-Substituted 1.3-Diols.
1,3-Dihydroxy compounds can be synthesized by several well known methods in literature. Aryl Grignard additions to 1-hydroxy propan-3.al give 1-aryl-substituted propan-1,3diols (path This method will enable conversion of various substituted aryl halides to 1arylsubstituted-1,3-propane diols (Coppi, et. al., J Org. Chem., 1988, 53. 911). Aryl halides can also be used to synthesize 1-substituted propanedlols by Heck coupling of 1, 3 -diox-4-ene followed by reduction and hydrolysis (Sakamoto, et. al., Tetrmhedron Lett., 1992, 33, 6845).
Substituted 1,3-diols can be generated enanadoselective reduction of vinyl ketone and hydoboration or by kinetic resolution of allylic alcohol (path Variety of aromatic aldehydes can be converted to l-substituted-1,3-diols by vinyl Grignard addition followed by hydroboration (path Substituted aromatic aldehydes are also utilized by lithium-t-butylacetate addition followed by ester reduction (path e) (Turner., J. Org. Chem., 1990, 55 4744). In another method, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:35 SPRUSON FERGUSON NO 7146 P. 36/51so commercially available cinnarnyl alcohols can be conven~ed to epoxy alcohols under calytic asymumetric epoxidazion conditions. These epoxy alcohols are reduced by Red-Al to result iii enanriorncrically pure 1,3-dials (path c) (Gag, et. al., ji Org Chem., 1980, 53, 4031).
Alternatively, euantiornerically pure 1,3-diols can be obtained by chiral borny reduction of hydroxycihyl aryl ketone derivatives (Ramachandrmi, et, al., Tetrahedron Lett., 1997, 38 761).
Pyridyl, quinoline, isoquinoline propan-3-ol derivatives can be oxygenated to 1-substicuted-1,3diol by N-oxide formation followed by rearrangemient in acetic anhydride conditions (;azh d) (Yamamnoto, et. al., Tetrahedron 1951, 37, 1371). Aldol condensation is another well described method for synthesis of the 1 3 -vxygenated fimcdonality (Mukaiyama, Org. React., 1982, 28, 203). Chual substituted diols can also be made by enanriaselective reduction of carbonyl compounds, by chiral aIdol condensation or by enzyme promoted kinetic resolution, VMgX H0 z b foMqX W /W YurCHRor N ILfL 2-Substituted 1.3-Dials: Various 2-substiwuted?1,3-diols can be made from commnercially available 2- (hydroxymethyl)-1,3-propmne diol. Pentacryibritol can be converted to trio! via decarboxylazion of diacid followed by reduction (path a) (Wade, at al., Liebigs. Ann. Chem., 1986,79 44) or dig!moncarboxylic acid dczivadves can also be obtained by decarboxylation under known conditions (Iwata, et, al., Tetrahedron let. 1957, 28, 313 Nitrotriol is also known to give triol by reductive elimination (path b) (Latour, et. al., Syunthesis, 1937, 8, 742). The triol can be COMS ID No: SMBI100629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:36 SPRUSON FERGUSON NO. 7146 P. 37/51 81 derivatised by mono acecylation or carbonate formation by treatment with alkanoyl dhloride, or alkylchloroformace(path d) (Greene and Wuts, Protective groups in organi synthesis John Wiley, New York. 1990). Aryl substitution can be Iffected by oxidation to aldehyde and aryl Grignard additions (path c) Aldehydes can also be convened to substituted amines by reductive S amination reaction(path e).
HO- H R- V NR, R
VV
R O OH d R M Ro' M -OCOR w v v W
W
1.3.c) Cyclic-t3- diols Compounds of formula 1 where V Z or V W are fused by four carbons are made from Cyclohexano diol derivatives. Commercially available cis, cis-1,3,S-cyclohexanc triol can be used as is or modified as described in case of 2-substituted propan-l,3-diols to give various analogues. These modifications can either be made before or after ester formation. Various 1,3cyclohexanc diols can be made by Diels-Alder methodology using pyrone as diene (Posner, et 16 al., Tetrahedron Lett., 1991, 32. 5295). Cyclohexyl diol derivatives are also made by nitrile oxide-olefin additions (Curran, et. al., J. Am. Chem. Soc.. 1985, 107, 6023). Alternatively, cyclohexyl precursors are also made from commercially available quinic acid (Rao, et. al., Tetrahedron Lett.. 1991, 32. 547.) COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:36 SPRUSON FERGUSON NO. 7146 P. 38/51 82 II. SYNTHESIS OF PBPase INHITrtraos: Synthesis of FBPase inhibitors is outlined in four sections: AICA riboside based inhibitors, purine based inhibitors, benzimidazole based inhibitors, Indole and 9azaindole based inhibitors.
III.1) AICA RIBOSIDE BASED INHIBITORS: Compounds of AICA riboside may be prepared by a variety of known methods. In general, these compoundA are synthesized by the method ofPrem C. Srivastava, et al, J, Med.
Chem., 1976, 19, 1020-1026 using methodology outlined below. Other methodology is described by Steven G. Wood, et al., J. Med. Chem. 1985, 28, 1198-1203, by G. Sagi, et al., J, Med. Chem. 1992, 35, 4549-4556, by R. Paul, J. Med. Chem. 1985, 28, 1704-1716 and by L. C.
Cohen, J. Amer. Chem. Soc. 1973, 95, 4619-4624.
AICA riboside, a commercially readily available starting material, is acetylated, for example, with acetic anhydride and a suitable base such as pyridine or triethylamine, and then optionally dehydrated by treatment, for example with tosyl chloride and pyridine. If esters other than acetate are desired in the final product, other anhydrides or acid chlorides may be employed in the acetylation step, for example use of isobutyryl anhydride gives the appropriate tri-0isobutyrate. The primary amine function is diazotized, for example with sodium nitrite, and created with a source of the appropriate nucleophile, for example copper(II) bromide to form corresponding bromide. The product is oxidized with, for example 30% hydrogen peroxide to desired compound.
Alternatively, the optional step of dehydration may be omitted to produce more directly compounds in which the alcohols are acylated. These agents may be deacylated, if desired to give the corresponding alcohols.
Alternatively, the imidazole base may be modified separately and then coupled to the appropriate sugar, using well-known glycoside bond forming reactions.
Further, compounds of the present invention might be prepared from compounds whose synthesis is described above. For example, the 5-thiomethyl analog may be prepared by a displacement reaction using sodium thiomethoxide and the 5-chloro compound.
3d Those skilled in the art will recognize that different reagents may be used in place of those listed above to give similar results.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:36 SPRUSON FERGUSON NO. 7146 P. 39/51 83 II111.2) PURTNE BASED INHIBITORS: Synthesis of intermediates of punne based inhibitors typically include some or all of the following general steps: deproteccion of phosphonate ester modification of CS-substirucd purine intennediates: modification ofpurine at positions other than CS; construction of the purine ring system; preparation of 4,5-dianinopyrimidine; and preparation of functionalized linker phosphonate.
A
N N
R
4 fL.2.a) Demrotection of Phosohonare Ester Phosphonic acids may be prepared from phosphonate esters using known phosphate and phosphonate ester cleavage conditions. For example, alkyl phosphonate esters are generally cleaved by reaction with silyl halides followed by hydrolysis of the intermediate silyl phosphonate esters. Various silyl halides can be used for this transformation, such as chlorotrimechylsilane (Rabinowitz J. Org. Chem., 1963, 28: 2973), bronmotrimethyisilane (McKenna et al. Tetrahedron Lett., 1977, 155), iodotrimechylsilane (Blackburn et al. 1 Chem.
Soc., Chum. Commun., 1978, 870). Phosphonate esters can also be cleaved under stiong acidic conditions, such as hydrogen halides in acetic acid or water, and metal halides (Moffatt et al.
U.S. Paen 3,524,846, 1970). Phosphonate esters can also be converted to dichlorophosphonates with halogenating agents PCl5, and SOC12, Pelchowicz et al. J. Chem. Soc.. 1961, 238) and subsequently hydrolysis to give phosphonic acids. Reductive reactions are useful in cleaving aryl and benzyl phosphonate esters. For example, phenyl phosphonate esters can be cleaved under hydrogenolysis conditions (Lejezak et al. Synthesis, 1982, 412) or metal reduction conditions (Shafer e al. J. Am. Chem. Soc., 1977, 99: 5118); benzyl phosphonate esters can also be cleaved COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:36 SPRUSON FERGUSON NO. 7146 P. 40/51 84 similarly (Elliott et al. i Med. Chem., 1985, 28; 1208). Electrochemical (Shono et al. J. Orgf Chem., 1979, 44: 4508) and pyrolysis (Gupta et al. Synth. Commun., 1980, 10: 299) conditions have also been used to cleave various phosphonate esters.
II.2.b)Modification of CS-Substituted Purinnrmia 8-Substituted purines are useful intermediates in the preparation of compounds of formula 10l. 8-Halopurines, which are particularly useful intermediates, are readily prepared using chemistry well deseibed in the literature. For example, N 9 -alkyladenines are halogenated at CS position using known halogenating agents Br2, NBS). 8-Alkylpurine can be prepared through direct lithiation of putinc followed by trapping with electrophiles alkyl halides, Barton et al. Tetrahedron Lett., 1979, 5877).
Functionaliztion of 8-halopurines can be accomplished under substitution reaction conditions with nucleophiles such as amines, alcohols, azides, sulfides, and alkylthiols. It is advantageous to have the phosphonate moiety as pan of the nuclcophiles. For example, substitution of 8-bromopurine with arninoalkylphosphonates give compounds of formula m where X are alkylarmino groups.
A 0
A
N Br N-Z-P(OH)
N
E N Z alkyt
Y
8-Halopurines can also be transformed into other 8-substituted purines using palladium catalyzed reactions (Heck, Palladium Reagents in Organic Synthesis; Academic Press: San Diego, 1985). For example, palladium catalyzed carbonylation reactions of 8-bromopurine in the presence of alcohol give 8-alkoxycarbonylpurines. Using known chemistry the 8-carboxylate group can be converted into other functional groups, such as hydroxymethyl. halomethyl, formyl, carboxylic acid, carbamoyl, thiocarbonyl groups, and these are useful intermediates for the synthesis of compounds of formula Il. For example, 8-alkyl and 8-arylpurines can be prepared from 8-halopurines via palladium catalyzed coupling reactions with organotin (Moriarty et al.
Tetrahedron Lett., 1990, 41: 5877), organoborane (Yatagai, Bull. Chem. Soc. Jpn., 1980, 53: 1670), and other reagents known to couple with aryl halides. When the coupling reagents contain COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:37 SPRUSON FERGUSON NO. 7146 P. 41/51 the dialkylphosphonate group, the reaction is useful for preparation of compounds of formula where X is alkyl, alkenyl, alkynyl, and aryl. For example, 8-bromopurine can be coupled with diethyl 1-tributylstannyl-3-allylphosphonate to give compounds of formula 5 where X is CH=CHCH2- and subsequent hydrogenation reaction give compounds of formula 5 where X is.
CH2CH2CH 2 The phosphonate group can also be introduced by further modification of the 8substituents. Substitutions of 8-haloalkyl or B-sulfonylalkylpurine with nucleophiles containing the phosphonate group amruseful for the preparation, of compounds of formula 5 where X is alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl. For example, compounds of formula 5 where X is CH20CH2- can be prepared from 8-bromomethylpurine using hydroxymethylphosphonate esters and a suitable base. It is possible to reverse the nature of the nucleophiles and electrophiles for the substitution reactions, i.e. haloalkyl- and/or sulfonylalkylphosphonate esters can be substituted with purines containing a nucleophile at the CS position (such as 8-hydroxyalkyl, 8thioalkyl, 8-aminoalkylpurines). For example, diethyl phosphonomthylriflare can be substituted by alcohols such as 8-hydroxymethylpurine to give compounds of formula 5 where X is CH20CH2- (Phillion et aL Tetrahedron Let. 1986, 27: 1477). Known amide formation reactions are useful for the synthesis of compounds of formula 5 where X is alkylaminocarbonyl, alkoxycarbonyl, alkoxythiocarbonyl and alkylthiocarbonyl For example, coupling of 8purinecarboxylic acids with aminoalkylphosphonate esters gives compounds of formula 5 where X is alkylaminocarbonyL For compounds of formula 5 where X is alkyl, the phosphonate group can also be introduced using other common phosphonate formation methods, such as Michaelis- Arbuzov reaction (Bhanacharya et al. Chem. Rev., 1981, 8: 415), Michaelis-Becker reaction (Blackburn et al. J. Organomet. Chem., 1988, 348: 55), addition reactions of phosphorus to electrophiles (such as aldehydes, ketones, acyl halides, imines and other carbonyl derivatives).
Compounds of formula III, where X is carboxypropyl or sulfonopropyl can be prepared from the reaction of 8-(2-iodoethyl)purine and corresponding phosphonomethylcarboxylate or phosphonomethylsulfonai (Carrescro et al., Terrahedron 1987, 43, 5125) in presence of base (eg. NaH) in polar aprotic solvents (eg. DMF). Substituted 8-(2-iodoethyl)purines arc prepared using know indole chemistry. For the preparation of a-phosphosulfonic acids see Magnin, D. R.
et al. J. Med. Chem. 1996, 39, 657.
Following well-reported literature procedures, other modification of 8-substituent of purines can be used to synthesize various compounds of formula III. For example, compounds of COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB-2004 18:37 SRSN&FRUO O 16 P SPRUSON FERGUSON NO. 7146 P. 42/51-- 86 formula III where X is varbonylalkyg Can be prepared from 8-carboxyallcylpurinea via conversion of 8-carboxyalkylpurines to their corresponding acid chloride and followed by Arbuzoy reaction (Chem- Rev- 1984, 84: 577) wit an alkyl phosphite to give 8-(2dialkylphosphonrarb~fylethyl)puies These a-ketophosphonares can be convened to the ahydroxyphosphonates and aa-dihalophosphonate-s (Smyth, Ct Tet:. Lett., 1992, 33, 4137). For another way synthesizng these aa-dihalophoaphomatcr.see Martin et at. Tert. Leit, 1992, 33, 1839.
8-Azidopurines arq usefta for the preparation for compounds of formula 5 where X is alkylamino and allcylcarbonylamnn groups. For example, carboxylic acids CROflP(O)alkyI-CO2H) can be directly coupled to 8-azidopurines to give S-alkylcarbonylauilnoprtne (Urpi or al. Tetrahedron Lett., 1986, 27: 4623). Alterntively, S-azidopurines can also be convened to S-aminopurincs under reductive conditions, and subsequently converted to 8alkylamixocasbonyj- and 8-aikylaminopunines using known chempistry.
111.6 Modification of Purines at putions le hnC Compounds of formula 5 can be fturther modified to give intermediates useful for the synthesis of compounds oftformula I11. For examnple, substitution reactions of 6-chioropurina by aranoa or allcylamines are useful for the preparations of compounds of formula 5 where A is amino and alkylamino groups.
E groups can be introduced by modifyig existing 2-substituents of purine. For example, 2-halopurines, readily accessible from 2 -axninopurines via chemistry well described in the literature, can be convened to other 2-substituted purines by for example nucleophilic substitution reactions; transition metal catalyzed reictions, etc. Mcd. Chem., 1993, 36: 293 8: Iffetera cycles, 1990, 30: 43 It is envisioned that N 9 -subsnited purines can be readily prepared from compounds of formula 5 where Y is H using for example standard alkylation. reactions (with ailcl halide, or sulfonare), of Mitswiobu reactions. Further elaborations of subsiTuents on Y are also possible.
11L2.d) Conswuogf- n ieSse Purine ring system of compounds o( fornula Mf can be constructed using diaminopyrimidines and carboxylates or their derivatives (such as aldehycdes, amides, uiiziles, orcho eaters, irnidates, etc.) (Townsend Chamisny of Nucleogfdesg and Nucleoddes, Vol I1; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:37 SPRUSON FERGUSON NO. 7146 P. 43/51 87 Plenum Press, New York and London, page 156-158). For example, alkyl and aryl aldehydes can be cyclized with 4 ,5-diaminopyrimidines as shown below.
A
A
N -CHo
N
A NHY E N
Y
Intramolecular cyclization reactions of pyrintidine derivatives can also be used to construct the purine ring system. For example, 5-acylarnino-4-alkylaminopyrimidines are treated with phosphorusoxychloride and cyclized under basic conditions to give purine derivatives. This transformation can also be achieved using other reagents SiCl4-Et3N, Desaubry et al, Tefrahedron Lett., 1995, 36: 4249). Imidazole derivatives are also useful for the construction of purine ring systen via cyclization reactions to form the pyrimidine ring (Townsend Chemisty of Nucleosides and Nucleoides, Vol I Plenum Press, New York and London, page 148-156).
II.2.e) Preoaration of Diaminoyvrimidine Compounds of formula 4 are useful for the construction of purine ring systems, and such compounds can be readily synthesized using known chemistry. For example, the Y group can be introduced using a nucleophilic substitution reaction involving an amine and 4 -halopyrimidines (Tetrahedrn, 1984, 40: 1433). Alternatively, palladium catalyzed reactions (Wolfe et al. J. Am.
Chem. Soc., 1996, 118: 7215) can also be used. Reductive anination reactions (Synthesis, 1975, 135) and alkylation with electrophiles (such as halides, sulfonates) are useful for the preparation of compounds of formula 4 from 4 -aminopyrimidincs. The 5-amino group can be introduced using amine formation reactions such as nitration followed by reduction (Dhainant et alt J. AMed.
Chem., 1996, 39: 4099), arylazo compound formation followed by reduction (Lopez et al.
Nucleosides Nucleondes, 1996, 15: 1335), azide formation followed by reduction, or by rearrangement of carboxylic acid derivatives Schmidt, Curtius, and Beckmann reactions).
Il,2,f) Preparation of Functionalized Linker Phoshonate.
Coupling of aromatic or aliphatic aldehydes, and carboxylic acid derivatives with attached phosphonate ester are particularly suited for the preparation of compounds of formula COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:38 SPRUSON FERGUSON NO. 7146 P. 44/51 88 III as described in section II.2.d. Such phpsphonate esters are prepared by the methods described earlier in section I.2.a.
A second lithiation step can be used to incorporate the aldehyde functionality, although other methods known to generate aromatic aldehydes can be envisioned as well Vilsmeier- Hack reaction, Rcimar-Tcimann reaction etc.). In the second lithiation step, the lithiated aromatic ring is treated with reagents that either directly generate an aldehyde DMF, HCO2R, etc.) or with reagents that lead to a group that subsequently transformed into an aldehyde group using known chemistry alcohol, ester, cyano, alkene, etc.). It is also envisioned that sequence of these reactions can be reversed, i.e. the aldehyde moiety can be incorporated first followed by the phosphorylacion reaction. The order of the reaction will be dependent on reaction conditions and protecting groups. Prior to the phosphorylation it is also envisioned that it may be advantageous to protect the aldehydes using a number of well-known steps (hemiacetal, hemiaminal, The aldehyde is then unmasked after phosphorylation. (Protective groups in Organic Synthesis, Greene, T. 1991, Wiley, New York).
II1.3) BENZIMIDAZOLE BASED INHIBITORS: Synthesis of the benzimidazole compounds encompassed by the present invention typically includes some or all of the following general steps: deprotection of phosphonate ester; substitution of the heterocycle; substitution or modification of 2-substituent; (d) cyclization to generate benzimidazole ring system; synthesis of the substituted 1,2phenylenediamine precursors; and f) preparation of functionalized linker phosphonate. A detailed discussion of each step is given below.
A
A
M-X-P-OR'
I>-X-P-OR'
J R R 6 7 Formula IV 2 OR 9 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:38 SPRUSON FERGUSON NO,7146 P. 45/51 89 IIL.3.a Deorotection ofbPhosphonate Ester Deprotection of phosphonate esters is effected as described in section II.2.a.
TII.3.b) Substitution of the Heterocycle The benzimidazole ring system of formula 8, may require further elaboration to provide desired compounds of formula 9.
i) Substitution of the Phcnvl Ring Electrophilic and nucleophilic substitution reactions enable incorporation of the desired substitutions encompassed by the formula 9. (March, Advanced Organic Chemistry by, Wiley- Intcrsicnce. 1992, 501-521; 641-654). For example treatment of the compounds of formula 8, where A is NH2, L and J are hydrogens with NBS, NCS or NIS in halogenated solvents such as carbon tetrachloride or chloroform gives halo-substituted compounds of formula 9 (L and/or J are halogens). Compounds of formula 9, where A is N02, L and/or I are alkenyl, alkynyl, alkyl, or aryl groups, and Y is H or alkyl, may be prepared from the formula 8, where A is N02, R is H or alkyl, and L and/or J are halogens, preferably bromide or iodide, through Stille coupling(Stille, Angew. Chem. Int. Ed. Engl. 1986, 25: 508-524). Treatment of the compounds of formula 8, where A is NO2, and L and/or J are bromides, with coupling reagent (e.g.
tributyl(vinyl)tin, phenylboronic acid, propargyl alcohol, NN-propargyl amine etc.) in presence of palladium catalyst bis(triphenylphosphine)palladium (II)chloride, tetrakis(triphenylphosphine) palladium(0), etc.] in solvent, such as DMF, toluene, etc. provides the coupling products. The compounds thus obtained can be modified as needed. For example vinyl or propargyl alcohol derivatives can be hydrogenated to give the ethyl or propyl alcohol derivatives respectively. These alcohol can be further modified as required via alkyl halides (ref.
Wagner et al. Tetrahedron Lett 1989, 30, 557) or alkyl sulfonates etc. to a number of substituted alkyls such as amino alkyl compounds by subjecting them to nucleophilic substitution reactions (March, Advanced Organic Chemistry, Wiley-Interacience, Fourth Edition, 1992, 293-500).
Alternately, these substitutions can also be done by metal exchange followed by quenching with an appropriate nucleophile (Jerry March, Advanced Organic Chemistry, Wiley-Interscience.
1992, 606-609). Nucleophilic addition reactions can also be usefol in preparing compounds of formula 9. For e.g. when A is N02, L and/or J are halogens nucleophiles such as alkoxides, thiols, etc. provides the halogen displacement products. (March, Advanced Organic Chemistry COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:38 SPRUSON FERGUSON NO. 7146 P. 46/51 Wiley-Intencience, Fourth Edition, 1992, 649-676). Another example is the addition reactions for example cyclopropanarion (Vorbruggen et al,Tetrahedron Lea,. 1975, 629) on the olefins(e.g.
styryl type) synthesized through Stille coupling.
If required, these substituted compounds can be further modified to the desired products.
For example reduction of the N02 to N142 may be done in many different ways, e.g. Pd/C, H2, aq. Na2S204, etc. (Larock, Comprehensive Organic Transformations, VCH, 412-415). These primary aromatic amines can also be modified as needed. For example N-acetyl derivatives can be prepared by treatment with acetyl chloride or acetic anhydride in presence of a base such as pyridine and mono-, or di-alkylamines can be synthesized by direct alkylation, using a base such as NaH in polar solvents such as DMF or by reductive alkylation (ref. Abdel-Magid et al.
Tetrahedron Lett. 1990, 31, 5595; also see ref. March, Advanced Organic Chemistry, Wiley- Intcrsoience, Fourth Edition, 1992, 898-900 for more methods).
ii AlkviJation of the Imidazole Ring Alkylation of the heterocycle of formula 8, (where R and J are both H) is obtained through two distinct methods that are amenable to a large number of electrophilcs.
Mitsunobu Alkylation Alkylation of the benzimidazole ring system of formula 8, is achieved by treatment of an alcohol, triphenylphosphine and diethylazodicarboxylate with heterocycle and a nonnucleophilic base such as Hunigs base in polar solvents such as CH3CN (Zwierzak et al, Liebigs Ann. Chem. 1986,402).
Alternately, the benzimidazole ring system of formula 8 can be deprotonated with a suitable base, preferably cesium carbonate in a polar aprotic solvent such as DMF, and the resulting anion is alkylated with an appropriate electrophilic component where L' is a leaving group preferably bromide or iodide.
IIL 3.cSubstitution or Modification of 2-Substitucnt Another key intermediate envisioned in the synthesis of compounds of formula 8 are substituted 2-methylbenzimidazoles. These compounds are readily prepared by condensing COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:39 SPRUSON FERGUSON NO.7146 P. 47/51 91 with the appropriate 1,2-phenylenediamine (Phillips, J. Chem. Soc., 1928, 29: 1305).
These compounds are useful in the synthesis of formula IV, wherein X is CH2ZCH2(ZtO,S,NH). For example, compounds where Z=O are readily prepared by treatment of the 2-methylbenzimidazolc with a halogenating agent such as NBS followed by reaction with the hydroxymethyl phosphonate ester (also see section 6, Synthesis of the Linker-PO3R2).
Alternately, a heterosubstituted methyl phosphonatcs can also be prepared by displacement reactions on phosphonomethyl halides or sulfonates (Phillion et al., Tetrahedron Left., 1986, 27: 14774) with an appropriate nucleophile e.g. 2-hydroxylmethyl benzimidazole compound which can be prepared using a variety of methods, including oxidation of the substituted 2-methyl benzimidazoles.
Similarly, compounds of formula IV, where X is carboxypropyl or sulfonopropyl can be prepared from the reaction of 2-(2-iodoethyl) benzimidazole and corresponding phosphonomethylcarboxylate or phosphonomethylsulfonate (Carretero et al., Tetrahedron, 1987, 43, 5125) in presence of base such as NaH in polar aprotic solvents such as DMF. The substituted 2-(2-iodoethyl) benzimidazole can be prepared from condensation of the corresponding substituted diamine and 3-halopropanaldehyde. Also see ref. Magnin, D. P, et al.
J. Med. Chem,. 1996,39, 657 for the preparation of a-phospbosulfonic acids.
The compounds of formula 8 where X is all carbon e.g. -(CH2)3- can be prepared by StiUll coupling (Stille Angew. Chem. Int. Ed. Engl. 1986, 25: 508-524) of the dialkylphosphopropenyl tributylstanne Org. Chem. 1993, 38:, 1986, 27: 1051).
The compounds of formula 8 where X is an amide linker e.g. -CONHCH2- can be synthesized using the following two steps. Treatment of the appropriate 1,2-phenylenediamine with trihalomethylacetamidate preferably trichloromethylacetamidate in polar solvent such as acetic acid followed by hydrolysis of the trihalomethyl group with strong aqueous base KOH) gives the benzimidazole-2-carboxylic acid (Eur. J Med. Chem., 1993, 28: 71). Condensation of the acid with amino phosphonate e.gdicthyl(aminomethyl)phosphonate in presence of condensing agent pyBOP) in a polar solvent such as methylene chloride to provide the amide linker phosphonato.
The compounds of formula 8 where X is an amide linker c.g. -NHCOCH2- can be synthesized using the following two steps. Treatment of the appropriate 1,2-phenylenediamine with cyanogen bromide (Johnson, et al, J. Med. Chem., 1993, 36: 3361) in polar solvent such as McOH gives the 2-amrino benzimidazole. Condensation of the 2- COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:39 SPRUSON FERGUSON NO. 7146 P. 48/51 92 aminobenzimidazole with a carboxylic acid e.g. diethyl(carboxymethyl)phosphonatc using standard coupling conditions (Klausner, et al, Synthesis, 1972, 453) to provide amide linker phosphonate. The 2-amainobenzimidazoles can also be prepared from the 2-bromobenzimidazotc via 2-azidobenzimidazole using known methods (Chem. Rev. 1988, 88: 297).
[II.3.dLCvclization to.Generate Benzimidazole Ring Svstem The benzimidazole ring systems of formula 8, is preferably assembled by condensation of substituted 1,2-phenyleneliamines with an aldehyde (RCHO, where R is e.g. aliphatic, heteroaliphatic, aromatic or heteroaromatic etc.) using known methods; in presence of Fc 3 salts preferably FeCl3 in polar solvents such as DMF, EtOH etc., reflux in non polar solvents such as toluene followed by oxidation, preferably with iodine (Bistocchi ct al. Collect. Czech.
Chem. C, 1985, 50(9): 1959.).; in cases of protected aldehydes the first condensation can be achieved in presence of an dilute inorganic acid preferably 10 H2SO4 in polar solvents such as THF, followed by oxidation with 12. Alternately, this coupling can be achieved with anhydride (RCOOCOR), carboxylic acid (RCOOH) or with the nitrile (RCN) by methods reported by Hein, et al, J. Am. Chem. Soc. 1957, 79. 427.; and Applegate, et al, US 5,310,923.
A
A
L NH 2 RCHO, FeCI3 N T NH, RCOOH. PPA 11.3.c) Substituted L2-.Phenvlenediamine 1,2-Phbnylencdiamines utilized in the preparation of compounds of formula IV, can be synthesized using methods well known in the art.
Compounds of formula 6, where R is H, can be synthesized from simple aromatic compounds. Most aromatic compounds, whether of high or low reactivity can be nirrated, because a wide variety of nitrating agents are available (March, Advanced Organic Chemistry, Wilcy-Intcrscience, 1992, 522-525). The primary aromatic amines are often protected as the Nacetyl, before nitration by treatment with acetyl chloride or acetic anhydride. Nitration of the COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:39 SPRUSON FERGUSON NO-7146 P. 49/51 93 these acetanilides derivatives using 60 HN03 and H2S04 (Monge et al, J. Med. Chem.. 1995, 38: 1786; Ridd Chem. Soc. Rev. 1991, 20: 149-165) followed by deprotection with'strong acids H2S04, HC1, etc.) and hydrogenation H2, Pd/C; Na2S204; etc.) of the resulting 2nitroanilines to provide the desired substituted 1,2-phenylenediamines. Similarly substituted arylhalides(F,CI,Br,I) can also be nitrated provides a-halonitroaryl compounds followed by nucicophilic addition NH3. NH20H, etc) and reduction to generate the diamines.
Diamines of formula 6, where A is N02 and R is H, can be produced using the method of Grivas et. al., Synthesis 1992, 1283 and Tian et a J. Chem. Soc. Perkin Trans 1, 1993. 257 and an appropriate o-nitroaniline. A variety of reactions can be used to substitute the o-nitroaniline.
For example halogenation of the nitroaniline Br2 .Cl2. etc.) gives the corresponding 4,6disubstituted or monosubstituted nitroanilinc which can be further modified at a later stage. The nitro group can be reduced with number of reagents preferably sodium dithionite to provide the corresponding diaminc. This diamine is then subjected to nitration conditions by first generating the 2,1,3-benzoselenadiazole with selenium dioxide followed by nitric acid. Substituted nitro- 1,2-phenylenediamines are generated by treatment of the nitro 2.1,3-bcnzoselenadiazole with aqueous hydrogen iodide or NH3/H2S (Nyhammar et al, Acta. Chem. Scand. 1986, B40: 583).
Other methods to simultaneously protect the diamine are also envisioned.
The compounds of formula 6, where R is alkyl or aryl, can be synthesized using the method of Ohmori et al, J. Med. Chem. 1996. 39: 3971. Nucleophilic substitution of the ohalonitrobenzenes by treatment with various alkylamines followed by reduction Na2S204) of the nitro group provides the desired compounds. Alternately, the compounds of formula 6, where R is H, can be synthesized from these o-halonitrobenzenes via o-azidonitrobntenes followed by reduction of the nitro group to provide the desired compound.
Alternately, diamines of formula 6 where R is not H are prepared by reductive alkylation of the o-nitroanilines with various aldehydes akyl, aryl, etc.) in the presence of a reducing agent preferably NaB(OAc)3 followed by reduction(e.g. Na2S204 Pd/C, H2, etc.) of the nitro group (Magid et al Terrahedron Lett. 1990, 31: 5595).
11 .3f. Prearation of Functionalized Linker Phoshonate.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20,FEB-2004 18:40 SPRUSON FERGUSON NO.7146 P. 50/51 94 Functionalizcd linker phosphonaces arc synthesized as described in section 11.2.f 11A) INDOLE AND 9-AZAINDOLE BASED INHIBITORS; Synthesis of indole and 9-azaindole compounds encompassed by present invention typically includes some or all of the following steps: deprotection of phosphonate ester (b) ring substitution of heterocycle; modification of 2-substituent to introduce X group; (d) synthesis of phosphonate substituted heterocycle by ring closure; synthesis of 2-nitro or 2amino alkylbenzene derivatives; and preparation of finctionalized linker phosphonate.
A
X-P-ORt E Y *OR J Y III.4.a) ODerotection of Phoshonate Ester Deprotection ofphosphonate esters is effected as described in section II.2.a.
III.4.b) Rine Substitution ofIndole Heterocycle i Introduction of Y Groun on Heterocvcle Introduction of Y group on the pyrrole ring of the heterocycle is selectively achieved either at the carbon or on the nitrogen depending on the reaction conditions employed. This selective substitution of the Y group also defines regiochemistry of A, L, E, J substituents on the benzene ring. Substitution at carbon of the indole base can be achieved using palladium mediated chemistry (Heck, R, Palladium Reagents in Organic Syntheses, Academic Press, New York, 1985). In general, these reactions entail coupling C3-iodo or -bromo indoles with boronic acids (Pure Appl.Chem. 1991, 63: 419) and stainanes (Stille, J. et al, J. Am. Chem.
Soc., 1984, 106: 4630) in the presence of a palladium catalyst. Terminal acetylenes also react in the presence of copper chloride and a palladium catalyst in a modified Stephens-Castro reaction (Sonogoshira, cc al, Tetrahedron Leu.. 1975, 4467; Sakamoto, T. et al, Synthesis, 1983, 312). These alkynyl or alkenyl groups can be further transformed to alkenyl or alkyl COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:40 SPRUSON FERGUSON NO. 7146 P. 51/51 substitution in a hydrogenation reaction by selection of a specific catalyst (Hutching in Patai, The Chemistry of Functional groups, Wiley, New York, 1983, 571; Lindlar, et al, Org. Synth Coll. vol. V, 1973, 880). Precursors for these coupling reactions can be made by halogenation at C-3 position of indole using reagents such as N-halosuccinimide (Mistry, A. ct al, Tetrahedron Lett., 1986, 27: 1051) or pyridinium bromide perbromide (Erickson, K. et al, Syn. Commun., 1981, 11: 253).
Introduction of a Y group at the N-I position of the indole in compounds of formula can be obtained by base-promoted alkylation with halides or sulfonates. Suitable bases include cesium carbonate or sodium hydride in an aprotic solvent (Guida, W. et al, J. Org. Chem., 1981, 46: 3172; Kikugawa. Synthesis, 1981, 124). Palladium catalyzed N-alkylation of aryl iodides is also an applicable method to introduce Y groups (Wolfe, J. et al, J. org. Chem., 1996, 61: 1133). Alternatively, Mitsunobu reaction conditions can be used for N-1 substitution of the heterocycle (Mitsunobu, synthesis, 1981, 1) using a variety of alcohols.
ii Substitution of the Bnenene Ring of the Heterocycle Substituents A, L, E and J in formula 10 can be introduced through reactions on indole or indole precursors, For example, substituents can be introduced on the heterocycle by substitution reactions (Hegedus, L. Angew. Chem., Int, Ed. Engl., 1988, 27: 113) and further converted to required functional groups at this stage. Functional groups on the benzene ring are transformed after addition of the linker phosphonate and before the deprotection of the phosphonate diester.
Amino groups can be incorporated from nitro groups introduced through nitration reaction of the heterocycle (Masuda, et al, Heterocycles, 1987, 26, 1475). Nitration reaction of indoles results in a mixture of 4- and 6- regio isomers. Selectivity is obtained based on the other substituents on the benzene ring. The reduction of the nitro functional group is accomplished utilizing methods such as catalytic hydrogenation or a chemical reduction Sn/HCI). Alternatively, selective nitro group reduction is obtained by aqueous sodium dithionate reaction. These conditions avoid hydrogenolysis of double bonds or reductive elimination of halogens (Org. Syn. Coll. vol 3, 1955, 69). Amines can be used to introduce other groups by diazotization reactions (Wulfman, in Patai The Chemistry ofDiazonium and Diazo Groups, Wiley, New York, 1978, 286-297). Amine groups are also expected to facilitate other substitution reactions. Halogenation reaction of the heterocycle results in A, L, E, J substitution with 4- and 6-amino indole isomers. Bromo or iodo substituents can be further transformed into COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:43 SPRUSON FERGUSON NO. 7147 P. 2/51 96 various substitunts by transition metal chemistry (Heck, R. Palladium Reagents in Organic Synthe es, Academic Press, Nw York, 1985). The metillaon, strategy devised by Mayor e al (I Org Chem, 1986, 51: 5106) can be used to substitute different groups CO2R, COR, SMe, alkcyl, aryl) at the IT4.g) Modificon of 2 -Substmen to Grou with Ph honate 2-Substituted indole heterocycles can be converted to intermediates useful for the synthesis of compounds o&formula 10. For example, compounds of formula I where X is methyleneaminocarbonyl may be obtained through a two-step procedure as shown below.
Indole-2-carboxylic esters are hydrolyzed using standard basic conditions g. NaOH, K2CO 3 The resulting carboxylic acids are coupled to form amide linkage (Klausner, et al, Synthea, 1972, 453; Bodansky, The Practice ofPeptide Synthesis, Springer, New York, 1984) with amino substituted phosphonate utilizing known coupling agents such as Pyr-BOP (Tetrahedron Lett., 15991, 32: 6387). Substituted indole-2-carboxyiic esters can be prepared, e. by Reissert indole synthesis (Rosenmond, et al, Ber., 1966, 99: 2504). The reaction involves condensation of 2nitro toluene with ethyl acetoacetat in presence of a mild base followed by a reductive cyclization.
A
A
0 1. INNaOH N 0 E OEt 2. PyBOP SJ
Y
Z=alkyl, aryl Compounds of formula 10, where X is carboxypropyl or sulfonopropyl can be prepared from the reaction of 2 -(2-iodoethyl)indole and corresponding phosphonomethylcarboxylate or phosphonomethylsulfonate (Carretero et aL, Tetrahedron, 1987, 43, 5125) in presence of base (eg. NaH) in polar aprotic solvents (eg. DMF). Substituted 2 -(2-iodoethyl) indoles are prepared using know indole chemistry (eg. Fischer indole synthesis). For the preparation of czphosphosulfonic acids see Magnin, D. R et al. J Med. Chum. 1996, 39, 657.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20,FEB-2004 17:43 SPRUSON FERGUSON 20.FEB 204 7:3 SRUON FRGUONNO. 7147 P. 3/51 97 Following well-reported literature procedures, other Modification of 2-*ubsticucnc of indoles can be used to synthesized various compounds of formula 10. For example, comnpound of formula 10 where X is carbonylalkyl can be prepared from 2 -carboxyaikylindoles via conversion of 2-carboxyalkylindoles to their corresponding acid, chloride and followed by Arbuzov reaction (Cherm. Rev. 1984, 84: 577) wich an alkyl phosphite to give 2-(2dialkylphosphonocarbonylethyl)indoles. These ot-kctophcsphonates can be convented to the abydroxyphosphonatcj and ax,ct-dihalophosphonates (Smyth, et al. Taft. Lore, 1992, 33, 4137), For another way synthesizing these a,a-dihalophosphonaces see Martin et al.Ter:. Let., 1992, 33.
1839.
3-Substituted indoles can be brominated selectively at the 2-position (Misizy, A- et al, Tet rahedron Let., 1956, 27: 105 These intermediates ame useflul in the preparation of compounds where X is alkyl, aryl, alkylamino, arylarnino, alcylihic, and azylzhio. For examiple, the bromo calm be replaced by such groups through a nucleophilic substitution reaction.
Alternat ively, phosphonate containing aromatic boronic acids, alkenyl stannaes or alkynyl X groups can be introduced in palladium mediated chemistry (Heck Palladium Reagents in Organic Syntheses, Academic Press, New York, 1955). In an alternate metallation route, Nsubstituted or protected indoles undergo litidation reaction ax the 2-position which is useful in reactions with various electropies (Synthsis, 1991. 1079; Mv: eracyclts, 1992, 33:173).
Compounds of Formiula 10 containing alkoxyalkyl as X group can be synthesized from indole-2caibinol intennediates obtained from the metallatlon reaction by quenching with an aldehyde (e.
g. formaldehyde). The phosphonate groups are introduced by 0-alkylation of hydroxyl with dialkoxy phosphonomethyl halide.
Compounds of Formula 10 where X and Y substituents are fused to give annulated indoles can be made in two general methods. Alicyclic fused compounds cant be made by Dials- Alder reaction of propargyl phoaphore with 3-vinyl indole derivatives (Pindur, U..
ileserocyci es, 1988, 27, 1253). Heterocyclic annulated indoles are synthesized from Indole-2methylene amnites by Heck type reactions (Tetrahedron Left., 1996, 3 7: 2659) and also by ring closure reaction of tryptumnine derivatives with aldehydes (Pens. S. Q. at al, .UebIg-s. Ann Chem., 1993, 2: 141; Pcelcegini, at at Terrahedrmn-Asynametry. 1994 .1979). Phosphonate ester on the annulated heterocycle can be substituted by dialkoxyphosphonomethyl triflate (Tetrahedron lett., 1986, 27: 1477).
COMS 10 No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:44 SPRUSON FERGUSON 2- -NO. 7147 4/51 98 In another rnthetic route, compounds of formula 10 are assembledbya ring closur; reaction (Sundberg, R. Indoles; Academic press: San Diego, 1996).
One of sch synthetic sequoncs involve the use of a phosphonat substituted aryl aldehyde. This aldehyde is condensed with a 2 irobcnzyl ylide, which is generated in situ by treardng 2 -nitrobenzykriphenyl phosphonium chloride with a base, a. potassium t-butoxide.
The Wittig salt is made under usual conditions by reaction of 2-nitrobeanzyl halide with triphaylphosphine (Murphy, P. et al, Chaem Soc. Ae. 1988, 17: 1. Maryanoff, B. et al., Chem. Rev. 1989, 89: 863).
A 0 NO2
NO,
Y tuOK E i Y pE Or (Eto) 3 P PdCl 2 (Ph 3
P)
2 SSnCI 2
CO
A
-X-PO^
J Y X=ary The diastereomeric mixture obtained from the condensation is then treated with triethylphosphite under refluxing conditions. This key step involves reductioh of the nitro group and consequent addition of nitrene into the styryl double bond resulting in a substituted indole heterocycle as in formula 10 (Gelmi, et al, J.Chem. Soc. Perkin I, 1993, 969). 2- Vinyitrobenzenes can also be prepared using other known methods, such as transition metal catalyzed coupling reaction between 2 -halonitrobenee and vinyl tin reagents, The above sequence can be used in the synthesis of compounds of formula 10, where X is an aryl group.
Various phosphonate substituted aryl aldehydes can'b prepared and used in this condensation.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:44 SPRUSON FERGUSON NO. 7147 P. 5/51 99 These types of reductive cyclizations can also be achieved in the presence of a catalytic amount of PdC2-SnC 2 under carbon monoxide atmosphero (Akazome, et at, Chem. Len.
1992, 769). Another transition metal catalyzed synthetic approach by Larock, R. et. al, (1 Am. Chem. Soc.. 1991, 113, 6689) is also suitable to obtain compounds of formula 1 by a ring closure reaction.
Another ring closure method useful for indolc synthesis is the palladium catalyzed cyclization reaction between 2 -haloaniline and an alkyne, alkene or ketone Org. Chem., 1997, 62(9), 2676; 62(19), 6464, P507). More importantly, this approach has been adopted for combinatorial synthesis of indoles on solid-phase which can be applied for the synthesis of indole FBPase inhibitors (Tetrahedron Leu., 1997, 38(13), 2307).
Compounds of formula 10 are also prepared from o-toluidine trisubstifuted amide cyclization, known as the Madelung indole syntheais (Brown. R. Indoles. Wiley New York 1972 Part 1; Houlihan, W. at al, J Org, Chem, 1981, 46: 4511)- The amide is cyclized under modified Madelung reaction conditions in the presence of Potassium ethoxide. The cyclization precursor is prepared by N-alkylation of amide followed by treatment with a non'nucleophilic base such as LDA and quenching the hcteroaryl anion with chlorodialkylphosphonate. The starting amide is an addition product of substituted o-toluidine and acid chloride.
A 1. YBr, KOH 0X 2. LDA, CIPO(OEt) 2
KOEO
J H j y X: furan, thiophene, oxazole. thiazole
J
j Y COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:44 SPRUSON FERGUSON 2NO. 7147---P. 6/51 100 2-Acyl nobnzylidenephosoane also lead to oles by an raolecuar Wiig ction with the amide carbonyl group (Le Corre, et al, Tetrahedron, 1985, 41; 5313; Capuano, i Chem. Ber., 1986, 119: 2069).
Altemrnatively, compounds of formula 10 can be obtained from silylated 2 -amino benzylie bromide by treating o-toluidines with 2 equivalents of lithiating agent n-BuLi) and TMSCI followed by bromination. Mixed organome
C
talli intermediates are then prepared by reactions with Zn and a soluble copper complex (CuCNs2LiCI), This reactive intermediate undergoes cyclization with an acyl chloride to give highly substituted compounds (Chen, t al, Tetrahedron Lett. 1989, 36: 4795; Bartoli, ct al, J. Chem. Sac. Chem Commun., 1988, 807).
A
1.N(1iMe), Zn S(SiM) 2. CuCN.2UCI (SiMe YE
V
J Br J Cu(CN)ZnB
A
L
N
L X-PEt 2
E
J
Alternatively, C-2 and C-3 substituted heterocycles of formula 10 can be made by condensation of a carboxylic acid ester with an organo dilithium intermediate of N-trimethylsilyl toluidine. Inverse addition of this organodilithium intermediate to a solution of aryl or alkyl ethyl ester results in a substituted indole heterocycle. Smith, A, et a, Terrahedron Lett. 1985. 26 3757; Li, IJ. et al, Synthesis, 1988, 73).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:44 20. EB. 004 7:44 SPRUSON FERGUSON N.74 ~.75 NO, 7147 7/51
NHTMS
1. 2 .2eq. n-BuLi \E-XO Et In another classical method known as the Fischer imdoc sythesis, compounds of fomula can be synthesized from aryl hydrazine wit an aldehyd;, or ketone via hydrazone formation.
Lewis acid catalyzed 3 .3lsigmatropic reanrngement of the bydxazone followed by cyclization of the enannhlo rcslib in substituted Lndolc (Robinson, The Flicker Indoke Synthesis; Wiley: New York, 1983). Zinc chloride is the most frequently used reagent among many known Conditions, however, various metal halides or acids (og. acetic acid. suilfle acid) also promote the reaction (Synthesis, 1980, 2222). Mild acids ane used in syuntheuis of C-2 and C-3 fibed indoles known (Sixnuzu, et al, Chem. Pharm. Bull., 1971,19:2561).
A
N.HNH
2 0 Y%%kXrPO -3N j ZnCi2 4
V
Y-4i, aikyl, cyclosiy, aryl Phosphonate substituted 9-azaindolo (also known as imidazopyridine) can also be synthoslzed via ring closure reations (Heterocycles, 1997, 45(3), 897; Synthesis, 1996, 927).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:.45 Date 2004-02-20 FEB. 2004 17:45 SPRUSON FERGUSON NO. 7147 P. 8/51 102 One method useful for 9-azaindole synthesis is the cyclization reaction between 2 -aminopyridine and a-haloketones (eg. a.bromoketone, a-chloroketone) and ketone derivatives as shown below' Heterocyc. Chem., 1989, 26, 1875).
A BNr txsP(Et)2 A t N L J J Y It is advantageous to have phosphonate ester presence in the a-bromoketone segment, however phosphonate can also be introduced to existing 9-azaindole. For example, 2phosphonomethylaminocarbonyl-9-azaindole can be prepared from 2-ethoxycarbonyl-9azaindole (available via cyclization reaction between 2-aminopyridine and ethyl bromopyruvatc) as described in secrion L1.4.b (Modification of 2-substituent to Introduce X Group with Phosphonate). 2-Phosphonomethoxymethyl-9-azaindole can also be synthesized from 2ethoxycarbonyl-9-azaindole by the following sequence: reduction of 2-ethoxycarbonyl group to 2-hydroxymethyl group, followed by alkylation with dialkylphosphonomethyl halide (referably iodide) as described in section II.4.b. Other modifications of 9-azaindole can be conducted as described early.
II.4.e) Synthesis 2-Nitro or 2-Amino Alkvl Benzene Derivatives: Building blocks for substituted benzene nuclei are obtained by nitration of alkyl benzenes. These compounds can be further transformed to 2-amino alkyl benzenes. 2-Amino alkyl benzenes can also be obtained from alkylation of aniline derivatives. A variety of substitutions on these groups can be made following known chemistry (March, Advanced Organic Chemistry, J. Wiley, New York, 1992, 501-568). N-Acyl and N-alkyl precursors can be obtained by methods mentioned earlier.
II.4.f) Synthesis ofLinker fX arou Phosphonate Diester Functionalized linker phosphonates are synthesized as described in section II.2.f COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 17:45 SPRUSON FERGUSON NO. 7147- P. 9/51 103 FormiuiatiQns Compounds of the invention are administered orally in a total daily dose of about 0. 1 mg/kg/dose to about 100 mg/kg/dose, preferably from about 0.3 mg/kg/dose to about mg/kg/dose. The most preferred dose range is from 0,5 to 10 mg/kg (approximately 1 to nmoles/kg/dose). The use of tirne-release preparations to control the rate of release of the active ingredient may be preferred. The dose may be administered in as many divided doses as is convenient. When other methods are used intravenous administration), compounds are administered to the affecteq tissue at a rate from 0.3 to 300 nmol/kg/mi, preferably from 3 to 100 nmoles/kg/min. Such rates are easily maintained when these compounds are intravenously administered as discussed below.
For the purposes of this invention, the compounds may be administered by a variety of means including orally, parenteally, by inhalation spray, topically, or rectally in formnnulations containing pharmaceutically acceptable carriers, adjuvants and vehicles. The term parenteral as used here includes subcutaneous, intravenous, intramuscular, and intraanerial injections with a variety of infusion techniques. Intraanerial and intravenous injection as used herein includes administration through catheters. Oral administration is generally preferred.
Pharmaceutical compositions containing the active ingredient may be in any form suitable for the intended method of administration. When used for oral use for example, tablets, troches, lozenges, aqueous or oil suspensions, dispersible powders or granules, emulsions, hard or soft capsules, syrups or elixirs may be prepared. Compositions intended for oral use may be prepared according to any method known to the art for the manufacture of pharmaceutical compositions and such compositions may contain one or more agents including sweetening agents, flavoring agents, coloring agents and preserving agents, in order to provide a palatable preparation. Tablets containing the active ingredient in admixture with non-toxic pharmaceutically acceptable excipient which are suitable for manufacture of tablets are acceptable. These excipients may be, for example, inert diluents, such as calcium or sodium carbonate, lactose, calcium or sodium phosphate; granulating and disintegrating agents, such as maize starch, or alginic acid; binding agents, such as starch, gelatin or acacia; and lubricating agents, such as magnesiumn stearate. stearic acid or talc. Tablets may be uncoated or may be coated by known techniques including microencapsulation to delay disintegration and adsorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. For example, a time COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:45 SPRUSON FERGUSON -NO. 7147" P. 10/51 104 delay material such as glyceryl monostcarate or glyceryl distearate alone or with a wax may be employed.
Formulations for oral use may be also presented as hard gelatin capsules where the active ingredient is mixed with an inert solid diluent, for example calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, such as peanut oil, liquid paraffin or olive oil.
Aqueous suspensions of the invention contain the active materials in admixture with excipients suitable for the manufacture of aqueous suspensions. Such excipients include a suspending agent, such as sodium carboxynethylcellulose, methylcellulose, hydroxypropyl methylccelluose, sodium alginate, polyvinylpyrrolidone, gum tragacanth and gum acacia, and dispersing or wetting agents such as a naturally occurring phosphatido lecithin), a condensation product of an alkylene oxide with a fatty acid polyoxyethylene stearate), a condensation product of ethylene oxide with a long chain aliphatic alcohol heptadecaethyleneoxycetanol), a condensation product of ethylene oxide with a partial ester derived from a fatty acid and a hexitol anhydride polyoxyethylene sorbitan monoolcate).
The aqueous suspension may also contain one or more preservatives such as ethyl or n-propyl phydmrxy-benzoate, one or more coloring agents; one or more flavoring agents and one or more sweetening agents, such as-sucrose or saccharin.
Oil suspensions may be formulated by suspending the active ingredient in a vegetable oil, such as arachis oil, olive oil, sesame oil or coconut oil, or in a mineral oil such as liquid paraffin.
The oral suspensions may contain a thickening agent, such as beeswax, hard paraffin or cetyl alcohol. Sweetening agents, such as those set forth above, and flavoring agents may be added to provide a palatable oral preparation. These compositions may be preserved by the addition of an antioxidant such as ascorbic acid.
Dispersible powders and granules of the invention suitable for preparation of an aqueous suspension by the addition of water provide the active ingredient in admixture with a dispersing or wetting agent, a suspending agent, and one or more preservatives. Suitable dispersing or wetting agents and suspending agents are exemplified by those disclosed above. Additional excipients, for example sweetening, flavoring and coloring agents, may also be present.
The pharmaceutical compositions of the invention may also be in the form of oil-in-water emulsions. The oily phase may be a vegetable oil, such as olive oil or arachis oil, a mineral oil, such as liquid paraffin, or a mixture of these. Suitable emulsifying agents include naturally- COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 17:46 SPRUSON FERGUSON NO. 7147 11/51 105 occurring gums, such as gum acacia and gum tragacanlt, naturally occurring phosphatides, such as soybean lecithin, esters or partial esters derived from farty acids and bexitol anhydrides, such as sorbican monooleatc, and .condensation products of these partial esters with ethylene oxide, such as polyoxyethylene sorbitan monooleate. The emulsion may also contain sweetening and flavoring agents.
Syrups and elixirs may be formulated with sweetening agents, such as glycerol, sorbitol or sucrose. Such formulations may also contain a demulcent, a preservative, a flavoring or a coloring agent.
The pharmaceutical compositions of the invention may be In the form of a sterile injectable preparation, such as a sterile injectable aqueous or oleaginous suspension. This suspension may be formulated according to the known art using those suitable dispersing or wetting agents and suspending agents which have been mentioned above. The sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenteraly acceptable diluent or solvent, such as a solution in 1,3-butane.diol or prepared as a lyophilized powder. Among the acceptable vehicles and solvents that may be employed are water, Ringers solution and isotonic sodium chloride solution. In addition, sterile fixed oils may conventionally be employed as a solvent or suspending medium. For this purpose any bland fixed oil may be employed including synthetic mono- or diglyceridos. In addition, fatty acids such as oleic acid may likewise be used in the preparation ofinjectables.
The amount of active ingredient that may be combined with the carrier material to produce a single dosage form will vary depending upon the host treated and the particular mode of administration. For example, a time-release fomnnulation intended for oral administration to humans may contain 20 to 2000 pmol (approximately 10 to 1000 mg) of active material compounded with an appropriate and convenient amount of carrier material which may vary from about 5 to about 95% of the total compositions. It is preferred that the pharmaceutical composition be prepared which provides easily measurable amounts for administration;'. For example, an aqueous solution intended for intravenous infusion should contain from about 0.05 to about 50 gmol (approximately 0.025 to 25 mg) of the active ingredient per milliliter of solution in order that infuision of a suitable volume at a rate of about 30 mL/hr can occur.
As noted above, formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 17:46 SPRUSON FERGUSON NO. 7147 12/51- 106 suspension in an aqueous or non-aqueous liquid; or as an oil-in-water liquid emulsion or a waterin-oil liquid emulsion. The active ingredient may also be administered as a bolus, electuary or paste.
A tablet may be made by compression or molding, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free flowing form such as a powder or granules, optionally mixed with a binder povidone, gelatin, hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegramt sodium starch glycolate, cross-linked povidone, cross-linked sodium carboxymctbyl cellulose) surface active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropyl methylcellulose in varying proportions to provide the desired release profile.
Tablets may optionally be provided with an enteric coating, to provide release in pans of the gut other than the stomach. This is particularly advantageous with the compounds of formula I when such compounds arc susceptible to acid hydrolysis.
Formulations suitable for topical administration in the mouth include lozenges comprising the active ingredient in a flavored base, usually sucroc and acacia or tragacanth; pastilles comprising the active ingredient in an inert base such as gelatin and glycerin, or sucrose and acacia; and mouthwashes comprising the active ingredient in a suitable liquid carrier.
Formulations for rectal administration may be presented as a suppository with a suitable base comprising for example cocoa butter or a salicylate.
Formulations suitable for vaginal administration may be presented as pessaries, tampons, creams, gels, pastes, foams or. spray formulations containing in addition.to the active ingredient such carriers as are known in the art to be appropriate.
Formulations suitable for parentaral administration include aqueous and non-aqueous isotonic sterile injection solutions which may contain antioxidamnts, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampoules and vials, and may be stored in a freeze-dried (lyophili2ed) condition requiring only the addition of the sterile liquid carnier, for example water for injections, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 17:46 SPRUSON FERGUSON NNO, 7147 7P. 13/51 107 immediately prior to use. Injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
Preferred unit dosage formulations are those containing a daily dose or unit, daily subdose, or an appropriate fraction thereof, of a drug.
It will be understood, however, that the specilic dose level for any particular patient will depend on a variety of factors including the activity of the specific compound employed; the age, body weight, general health, sex and diet of the individual being treated; the time and route of administration; the rae of excreion; other drugs which have previously been administered; and the severity of the particular disease undergoing therapy, as is well understood by those skldled in the art.
EXAMLEN
The prodrug compounds of this invention, their intermediates, and their preparation can be understood further by the examples which illustrate some of the processes by which these compounds are prepared. These examples should not however be construned as specifically limiting the invention and variations of the compounds, now known or later developed, are considered to fall within the scope of the present invention as hereinafter claimed.
Compounds of formula UI are prepared according to the literature procedures with modifications and additions well understood by those skilled in tie an. In general, these compounds are synthesized by the method ofSrivastava, jLje4hM.s 19, 1020 (1976). Other methodology is described by Wood et at. LMed.QCh1an. 28: 1198-1203 (1985); Sagi ec al., L Mdf.r m35: 4549-4556 (1992); Paul, Jr. iHiJsirh 28: 1704-1716 (1985); Cohen et al., IAm..C Soc. 95.4619-4624 (1973).
Compounds of formulae III-V are prepared according to the procedures described in scction 1I, supra.
Compounds of formula I arc prepared using procedures detailed in the following examples.
Example L* Genera rocedurefor 1 '.-cycloliexvl armulated ndrus by th vl chloride reaction.
A suspension of 1 mmol ofphosphonic acid in 5 mL of thionyl chloride was heated at reflux temperagre for 4 h. The.reaction mixture was cooled and evaporated to dryness. To the COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:47 20. EB. 004 7:47 SPRUSON FERGUSON O 17 P NO. 7147 P. 14/51 108 resulting residue was added a solon of 1 mMol Of alcohol and 2.5 minol pyridine in 3 ml. of Methylene chloride, After stirring at 25 0 C for 4 hithe reaction was subjected to work up and chromatography.
The following Compounds were prepared in this manmer: 1-1: 6-mn--5-ymy-'3.ylhey~htRoouay--hniy purine. Anal. Cald.
for C23 [1241N$ 05 P OAS H120: C. 57.06 H1: 5.06 N: 14.47. FouncL C: 56.84; H: 4.83; N: 14.38.
1-2: 6 -Amino-8-(5'-hydwoxyc$.3 ccoay~hshnouay--epny purine, minor isomer. RfM0. 10% MCOH-CX-2C12. nip 248 250 Anal. CaM,. for C20 [H26 N5 05 P 0-.H[20: C: 52.63; 5.96; N; 15.34. Found: C: 52.62; 5.70; N: 15.32- 1.3: 6-Aniino-8-(5'-hydroxyll',3' cycloexyl)phosphonoanyl9n.,.pcntl purine, major isomer. RNO.35 10% MeOH-CH2cl. nip -225 230Oc; Anal. Cald. for C20 [126 NS 0S P 0.5 [H20: C: 52.63; H; 5.96; N: 15.34. Found: C: 52.74; Hi: 5.80); N: 15.32.
1.4: 6-Chioro-4S.dimerhyl.] -cyclopropylncty[-24 I-hYdroxy-3'S'Cyc1QhexY1P1VsphoDo-5S fiurany1benimnid1e. nip =211 21SC; Anal. Cald. for C23 [126 Cl N2 05 P 2/31120:
C:
56.50: H: 5.64; N: 5.71 Found: C: 56.65; H-1 5.54 N: 5.64.
6-Chloro-4,5-dimehyu.l-ceyclopropymedlyl-2z..r '-acntylhydroxy-3y,s'.
cYclohxyphosponos5Ruaytbimidzle, minor isomer. Rf-0.35 iW 10% MCOH--CH2C12.
Anal. CakE, for C25H2 SCIN206?p+ 1.51H20: C: 55.00 H: 5.72; S.13. Found: 55.19; H: 5.3 1; N: 4.65.
1.6: 6 -Ch~oro-4,5-dimnethyl.. 1-cyelorropylmethyl..2-4l -acetylhydraxy.J',s'cyihxlhshn--uay~czziaoe major isomer. RNO.4 in 16% MeOH-CR2U Anal. Cald. for C25H28C1206P4+0.7512o+o 1EtOAc; C: 56.37; H: 5.64; 5.18. Found: C: 56.68; H:!5.69; N: 4.80.
6-Chloro- 1-laobinyl.2- cyclhexyl)phoaphonolfur.nyl) beazimidazole, miinor isomer. Rf-0.60 in 10% MCOH.CH2CI2.
Mip >220*C; Anal. Oak!. for 021 H24 Cl N2 05 P 1/31420: C: 55.21; H: 5.44; N: 6.13.
Found: C: 55.04; [H:5.50; N: 6-Chloro- I-isobutyl-2- I 'hydroxcy-3',S'cycohCxy)piosphono~furanyl~be~mjide, major isomer. R1-O.55 in 10% Me0H-CH2Co2 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:47 SPRUSON FERGUSON 20.FEB 204 1:47SPRSON& FRGUONNO. 7147-'P. 15/51- 109 nip >220aC; AntL Cald. for C21 H24 Cl N2 05 P: C: 55.94; H; 5.37; N: 6,2 1. Found: C, 55.73; H: 5.34; N: 6.13.
Prrosrationl of 1' 1-subat td Cc ic-1'3' t~g~ The following compounds were prepared by procedure described for Emple 1: 2.1: 6-Chloro- I -isobutyJ-2'(2-(s 4 1 '-R-phenyl3-p y~hshn~uanlbniiaoe major isomer. in 10% MOOH-CH2C12. nip 204 206 9 C; Anal. Cal&, for 024 IJ24 CI N2 04 P: C: 61.22; HI: 5 14; N: 5.95. Found; C: 60.95; H: 5.01; N: 5.88.
2.2; 6-Chloro- 1-isobutyl-2-(2-(5-( I'-R-phcnyl-.3-rp pophn~uay~bniiao wmorisomen. M-0.72 in 10% MeOH-CH2c12.Anal. 081± for C24liz4ClNm4p+Hao.
C:
58.96; H: 5-36, N: 5.73. Found: C: 58.85; H: 5.48; N. 5.55.
2.4: 6 -Chloro-1-isobuyl.2 (5-[lS-(4-nitropbcnyl) .2R-acec,1ariao>propo-1 .3-yl]phosphonc.2fulranyl)benzimnidazole. major isomer. P1-0.35 3% MeOH-CH2c12, Mass 04±d for C26H26C1N407p- MH-'473, Found: MH4573.
6-Chioro-lI-iaobutyl-2- (S-f lS-(4-ni ophenyl}..2R..aoetynino-ropanz 3 -yljphosphono-2.
fiway)bziiiazil miinor. Rf-0.35 3% MeOR-H2c12. Anal. Cald. for C26H26C2N4O7P.6H20e-o 250H2012. C; 50.61; H: 4.81; N: 8.99. Found: 50.25; H: 4.37; N: 9.01.
2.6; 6-Chloro- 1-isobuzyl-2- (5-f lS.( 4 -merhyltiopheny)2S-cetylumino.propanl ,3- YIJPhhob-f2.tbrmyIlbenzirnidazole- Anal. Caid. for
C
2 7H29CIN3OSPS+1H20+0.35CH2C12: C: 52.83;, H: 5.14; N: 6.76. Found: C: 52.44; 11: 4.76; 6.59.
Preartio o 1'-tra susttued cclic-I .]-gopy1l estrs &WA To a'solution of 2-fiuradehyd. (3 g, 3?1.2 mnmol) in THF (60 mL) was added I M vivyl magnesium bromide in THlE (34 mnL) at 0 03C. Aftr stirring for an hour, a solution of 1 M BH,.THF cfnmplex in THF was added. The reaction was quenched with 3N NaOH (20 tnt) mnd COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:47 SPRUSON FERGUSON NO.7147 P. 16/51 110 hydrogen peroxide (10 mL) at 0 0 oC The organic fraction was scparated and concentrated.
The crude product was chromatographed by eluting with 5% methanol-dichlorornethane to give 2-( 3 -furyl)propane- 1,3-diol (1 g, 22%).
51mThe prodrug was made following the procedure as described in Example 1.
3.1: 6-Chlor- -lisobutyl-2- 3 -furyl)-propan-l ',3'-y]phosphono-2-furyl}benimidale.
mp 160 162 0 C. Anal. cald. for C22H22ClN205P+0.4H 2 0: C: 56.45; H: 4.91; N: 5.99.
Found- C: 56.67; H: 4.82 N: 5.68.
Examnle 4; Preiaration of I'-ovridvi substituted cyclic-I'.3'-royl esters: Step Org. Chem., 1957, 22. 589) To a solution of 2-pyridine propanol (10g, 72 .9mmol) in acetic acid (7SmL) was added hydrogen peroxide slowly. The reaction mixture was heated to 80" C for 16h. The reaction was concentrated under vacuum and the residue was dissolved in acetic anhydride (I OOmL) and heated at 110" C overnight. Acetic anhydride was evaporated upon completion of the reaction.
Chromatography of the mixture by eluring with methanol-methylene chloride resulted in 10.5g of purn diacetate.
To a solution ofdiacetae (Sg, 21. mmol) in methanol-water 40mL) was added potassium carbonate (14.6g, 105.5mmol). After stirring for 3h at room temperature, the reaction mixture was concentrated. The residue was chromatographed by eluing with methanol-methylene chloride to give 2.2g of crystalline diol.
The procedure for coupling is the same as described in Example 1.
The following compounds were prepared in this manner: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:48 SPRUSON FERGUSON 26 FB.264 7:8 PRSO &FEGUONNO 714 7 P. 17/5i 4.1: 6 -Aminao-9.neopeniyis.. 2 -pyridyl)p ropane. YlDPhoSphnoJfijfnyijpurjne.naIj Cald. for C22H25N64p+o.jsHo+1Hci. C: 50.97; H: 5.3 5; N: 16.21. Found: C. 51.19; H: 5.02 K: 15.9 1.
4.2: 6-Chloro.. 1-isobucyl2- -C2-pyridyl)-propan. l,S3'y!I hshn--uanibmmdme Anal. Cald. for C23*i23ClzqaQ4p+l.5H20-I-.3 Cfl2Q2: C: 53.37; H: 5.11; N: 8.01. Found: C 53.23; H. 4.73 N: 7.69.
4.3: 6-Chiora- I -isobutyl-2- (5-1 P-(4-pyridyO)-propan- 1 ',3'-yI]phosphono.2 furanyl) benzimiclnolc. m? 165.0' C(dcc.). Mass Cald. for C23H23clN3o4p: NIH+ 454: Found: MH 4 454 4.4: 4 ,5.67-Tecrunezhyl- I -isobutyl-2. (S-fl 4 -pydidyl)-propan- 1,.yijpbosphono..2.
flranyl)benzimidazo~e. Anal. Cald. (or C27H32N3O4.1251120: C: 62.84; H: 6.74; N: 8. 14.
Found: C: 62.82; H: 6.8 1; N: 8.48.
S-ChloroA4-methyl. 1-isobucyl.2. 4 -pyridyl)-propan- l,i-yIlphosphong4.ftranyllberizimidazolc. Anal. Cald. for C 2 4H25C1N304P+o 5H20+0 JJEC] C: 56-86; H: 5.24.
N: 8.29. Found: C: 56.97; H: 5.08; N: 8.26.
To a solution of 6 -chloro-1-isobuty-z 2 -pyrdyl)..propan-l1'.i'-yI]phosphono-2.
ftuanyllbeuzimidazoie (172mg, O.36mrnol) irt methylene chloride was added 3chloropewoxybezic acid (252mg, 0OJ2mmoi) at 0* C. The reaction was warmed to room temperature and allowed stir for 3h. The solvent was evaporated under reduced pressure, Chromatography by elutiern with mehaxlmethyenechlon'ac resulted in 100mg of pure N-oxide.
The following compound was prepared In this manner 1'.
3 -ylJphOSphono.2-fbranyl}bcnzimidazoljq np 195.0 t(dec.); Anal. Caid. for C23H23CIN3OSP*i0252j+325CH 2
C
2 C: 54.37; H: 4.71: N: 8.18- Found:, C: 54.77; R: 4.86; N: 7.76.
Prsey COMS ID No: SMBI-00629627 Received by iP Australia: Time 18:45 Date (V-M-ci) 2004-02-20 FEB. 2004 17:48 SPRUSON FERGUSON NO. 7147 P. 18/51 112 A: Org. Chem., 1988, 53, 911) To a solution of oxalyl chloride 5.7 mL, 97 mmol) in dichloromethsne (200 mL) at -78* C was added dimethyl sulfoxide (9.2 mL, 130 mmol). The reaction mixture was stirred at 78° C for 20 min before addition of 3 -(benzyloxy)propan-l-ol (11 g, 65 mmol) in dichloromethane (25 mL). After an hour at -78 C, reaction was quenched with triethylamine (19 mL, 260 mmuol) and warmed to room temperature. Work-up and column chromatography by elution with dichloromedtane resulted in 8 g of 3-(benzyloxy)propan-1-al.
tep To a solution of 3-(benzyloxy)propan-l-al (1 g. 6.1 nmol) in THF at 0" C was added a IM solution of4-fluorophenylmagnesium bromide in THF (6.7 mL, 6.7 mmunol). The reaction was warnned to room temperature and stirred for Ih. Work-up and column chromatography by elution with dichloromethane resulted in 0.7 g of alcohol.
To a solution of benzyl ether (500 mg) in ethyl acetate (10 mL) was added 10% Pd(OH)2-C (100 mg). The reaction was stirred under hydrogen gas for 16h. The reaction mixture was filtered through celite and concentrated. Chromatography of the residue by elution with ethyl acetate.
dichloromethane resulted in 340mg of product.
The procedure for. coupling is the same as described in Example 1.
The following compounds were prepared in this manner: 6-Chloro- I -isobutyl-2- 4 -fluorophenyl)-propan-l ',3'-yl]phosphono-2furanylj benzimidazole, minor isomer. Rff-0.45 in 5% MeOH-CH2CI2. mp 207 208 Anal.
Cald. for C24H23CIFN204P: C: 58.96; H: 4.74; N: 5.73. Found: C; 59.20; H: 4.64; N: 5.59.
5.2: 6-Chloro-1-isobucyl-2- {5-[1'-(4-fluorophenyl)-propan-l',3'-yl]phosphono-2fihranyl}benzimidazole, major isomer. Rf-0.4 in 5% MeOH-CH2C12. mp 176 179'C; Anal.
Cald. for C24H23ClFN204P+0.SH20: C: 57.90; H: 4.86; N: 5.63. Found. C: 57.60; H: 4.68; N: 5.54.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB.2004 17:48 SPRUSON FERGUSON NO.7147 P. 19/51 113 xami 6: Preparation of 1'-phenvl substituted cyvclical'.3'-oropvl esters: S A: Org. Chem., 1990, f 54744) To a solution of diisopropylamine (4.1 mL, 29.4 mmol) in ether (40 mL) at -78 °C was added n-butyl lithium (11.8 mL, 29.4 mmol). The reaction was stirred for 15 mmin before adding rbutyl acetate (4 mL, 29.4 n}mol) in ether (10 mL). After 20 min, aldehyde (3g, 14 mmol) in ether mL) was added and warmed to room temperature where it was stirred for 16h. Work-up and column chromatography by clution with ethyl acetate-dichloromethane resulted in 3.3 g of addition product.
To a solution of t-butyl ester (1.5 g, 4.5 mmol) in THF (20 mL) was added IM lithium aluminum hydride at 0"C, The reaction mixture was warnned to room temperature and stirred for 2h. The reaction was quenched with ethyl acetate and saturated aqueous sodium sulfate was added to.
precipitate the salts. Filtration and concentration of solvent resulted in a crude diol. Column chromatography by elution with ethyl acetate-dichlorometbae gave 970 mg of pure diol.
St C: The procedure for coupling is the same as described in Example 1 The following compounds were prepared in this manner 6.1: 6-Chloro-l-isobutyl-2- 3 -bromo-4-methoxyphenyl)-propan-l',3'-yl]phosphono-2firanyl)benzimidazole, major isomer. Rf-0.35 in 70% EtOAc-CH2CI2. mp 167 169 "C; Anal. Cald. for C25H25BrCIN205P: C: 51.79; H: 4.35; N. 4.83. Found: C: 31.77; H: 4.25; N: 4.73.
6.2: 6-Chloro- 1-isobutyl-2- 3 -Bromo-4-mcthoxyphenyl)-propan. ',3'-yl]phosphono-2furanyl)benzimidazole, minor isomer. Rf*0.3 in 70% EtOAc-CH2C12. Anal. Cald, for COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:48 SPRUSON FERGUSON NO. 7147- P. 20/51 114 C25H25BrCIN205P+0.55CHC13: C: 47.54; H: 3.99; N: 4.34. Found: C: 47.50; H: 3.89; N: 3.99.
Example 7: Prcvaration of 2'-ubstituted cyclic-1 '.3.'-ronvl esters: Monoacetvlation of 2-(hvdtrxvmethyl-13-propanedial: To a solution of 2-(hydroxymethyl)-1,3-propanediol (1 g, 9.4 mmol) in pyridine (7.5 mL) at O'C was added acetic anhydride (0.89 mL, 9.4 mmol) slowly. The resulting solution was warmed to room temperature and stirred for 16h. The reaction was concentrated under reduced pressure and chromatographed by eluting-with methanol-dichloramethane to give 510 mg of pure acetate.
Methvlcarbonate formation of 2-(hvdroxvmethyl)-1.3-aropanediol: To a solution of 2-(hydroxymethyl)- 1,3-propanediol (1 g, 9.4 mmol) in dichloromethane (20 mL) and pyridine (7.5 mL) at 0eC was added methyl chlorofornate (0.79 mL, 9.4 mmol) slowly. The resultinS solution was warmed to room temperature and stirred for 16h. The reaction was concentrated under reduced pressure and chroimatographed by eluting with methanoldichloromethane to give 650 rmg of pure carbonate.
The procedure for coupling is the same as described in Example 1.
The following compounds were prepared by step B or by step A and B: 7.1: 6-Chloro-1 -isobutyl-2- {5-[2'-(hydroxymethyl)-propan-1 ',3'-yl]phosphono-2furanyl}benzimidazole. rnp 164 165 Anal. Cald. for C19H22CIN205P: C: 53.72; H: 5.22; N: 6.59. Found: C: 53.62; H; 5.18; N: 6.42.
7.2: 6-Chloro- -isobutyl-2- (5-[2'-(acetoxymethyl).propen-l',3'-yl]phosphono-2furanyl)benzimidazole. mpn 132 134 oC; Anal. Cald. for C21H24CIN206P: C: 54.03; H: 5.18; N: 6.00. Found: C: 54.17; H: 4.99; N: 5.81.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:49 SPRUSON FERGUSON 20.FEB 204 1:49 SPRSON& FRGUONNO. 7147P. 21/51 115 7.3: 6-Chloro-lI-isoburyl-2- {S-(2'4medhoxycrbanyloxymethy)-prpan.. I',3'-yllphoShhcno-z.
furanyl)benzirnidazole. nip 138 140 0 C; Anal. Cald. for C2124C1IN207P: C; 52.24; H: 5.01; N: 5.80. Found. C: 52.13; H; 5.07; N: 5.51.
7.4: 4-Amnino-5-fluoro-7-ethyl-..isobuyL-2.(S-C 21 -(aceroxymelbyi)-propan-.I ',3'-yljphosphono- 2-furanyl~benzimidazole. Anal. Cald. for C23H29FN306p+0.3H20: C; 55.38; H: 5.98-; N: 8.42. Found: C: 55.60; H: 6-31; N; 8.02.
6-Aminc-9-ncopentyl-8- {$-(2'-(acetoxyrnErhyl)-propan- -yljphosphv-no-2fbr-anyl Ipurine. nip 164 165 Anal, Ca. for C2OH26NSO6P: C 51.84; H: 5.65; N; 15.11 Found: C: 52.12; h.,5.77 N: 14.59.
7.6 4-Amino-5-fluoro-7-ethiyl- I1-isobucyl-2- (S-t2'-(vyclohexanecurbonyloxymethyl)-propan- I '.3'-y[]phosphono-2-ftrauyl benzimidazole. rup -62 63 Anal. Cald. for C28H37FN306P: C: 59.89; H: 6.64; N: 7.48. Found: C: 59.97; H: 6.60; 7.33.
4-Aniina-5-tluoro-7-cthyl-1-isobutyl-2- {5-12'-(hydroxymethyl)-propan. I',3'-yl~phospktono- 2-furanyl)benzimidazole. Anal. CulcL Lhr C21H27N305P+o.6 MtAC: 55.73; H: 6.36; N: 8.33. Found: C: 5S.81; H: 6.08 N: 3.02.
7.8: 4,5 ,6.7.Tetramethyl-I -isobutyl-(2-(5 -[2-methoxycarbonyloxymethy)propa-1 .3yllphosphona))furany1]bcnzirnidnote. minor isomecr. Rib;0.53 in 5% MeOH.CH42C12. Anal. Cald.
for CZSH33N2O1P+0.25H20: C: 58.99; H: 6.63; N: 5.50. Found: C: 59.21; H: 6.73; N: 5.48.
7.9: 4.S,6,7-Tetramnethyl- l-isobutyl-[ 2 -(S-(2-(nzethoxyca~onyloxymrihy>.propan-i ,3.
yljphosphono)fisranyl]bcnzimidazole, major isomer. Rf=0S4 in 5% McOH-CHZCI2. Anal. Cald.
fbr C25H33N207P+fM0: C: 57.47; H: 6.75; N: 5.36. Found: C: 57.72; H: 6.86; N: 5.22.
7.10: 5-Chloro-4-mcthyl-l1-isobury-f2-(5[2-(mtoxycarbonyloxymthyl)-pwopan-...3.
yllpbosphono)furayl~bcnzimidazole, mainor isomer. RP=".59 in 100%/ EtOmc. Anal. Cald. fbr C22H26C1N207P+0.7SH2O: C: 51.77; H: 5.43; N: 5.49. Found: C: 51.80; H: 5.35; N: 5.39.
7.11: 5-Chloro-4-niethyl-l .isobury1.42.(5-[2.(rnethoxycarbonyloxymthy).popn-1,3yl~phasphono)fuanyllbenzimidaizole, major isomer. Rt'-0.54 in 100% EtOiAc. Anal. Cald, for C22H26C1N207P+eH20: C: 51.32; H: 5.48; N: 5.44. Found; C: 51.36; H; 5.25; N: 5.25.
8.1: 5-Bromo- 1-(B-D-ribofumraosyl)-imnidazolt-4-carboxsznjde 8.2: 5-Bromo-1 ,5-tri-O-acety1-j-D-ribo-firanosy)imaidazole-4-carbgnamide COMS ID No: SMBI-00629627 Received by IP Australia: Time (1-tm) 18:45 Date 2004-02-20 FEB. 2004 17:49 SPRUSON FERGUSON NO. 7147 P. 22/51 116 A stirred mixture of AICA riboside (200 g, 0.774 mol) in pyridine (1200 mL) was cooled in an ice bath. Acetic anhydride (310 mL, 2.80 mol) was slowly added over 25 minutes. The ice bath was removed and the solution stirred at room temperature for 2 1/2 h. TLC (silica gel, 9/1 merhylene chloride/ methanol) indicated the reaction was complete. The solvent was evaporated to give a pale orange oil. Diethyl ether (600 mL) was added to the oil and the mixture vigorously stirred. The upper ether phase was decanted. The thickened tar was riturated/ decanted three times with 300 mL of ether. The resulting orange tar was dissolved in warm ethanol (600 mL), The solution was stirred overnight at room temperature and the resulting solid filtered, washed with cold ethanol (75 mL) and vacuum dried to yield AICA riboside triacetate, 203 g (68 [melting point -126.5-128.5 TLC (silica gel, 9/1 methylene chloride/ methanol): rf 0.4.
The diethyl ether washings (from above) were combined and stored at room temperature overnight upon which a white crystalline solid founrmed. The solid was filtered, washed with cold ethanol (50 mL) and vacuum dried to give an additional 26.5 g (8.9 AICA riboaide triacetate (50.0 g, 130 mmol), CuBr2 (14.5 g, 64.9 mmnol), LiBr (45 g, 0.52 mol) and acetonitrile (500 mL) were combined under an atmosphere of argon and cooled to [sobutylnitrite (19.3 miL, 162 mmol) was added dropwise over 10 minutes. The cooling bath was removed and the solution stirred for 20 h. The solvent was evaporated and the residue partitioned between methylene chloride (600 mL) and 10 NaHS03 solution (150 mL). The organic phase was separated, evaporated to 200 mL and diluted with ethyl acetate (250 mL). The solution was extracted twice with mL portions of saturated NaHCO 3 Silica gel (175 g) was added to the organic phase and the mixture stirred for 15 minutes. The mixture was filtered through a pad of Celite and the pad washed with ethyl-acetatc (400 mL). The combined filtrate was evaporated to give 39.6 g of a tar which was dissolved in warm water (400 mL) and stirred at room temperature overnight. A white precipitate formed and the mixture was refrigerated for several hours. The solid was filtered, washed with cold water (100 mL) and vacuum dried to yield 20.5 g of an off-white powder (35 [mp 133-135 dC, TLC (silica gel, EtOAc): rf 0.75].
The appropriate chloro and iodo analog were made by using this method with the substitution of copper(II) chloride or copper(H) iodide for the copper(II) bromide.
Example 9: 9.1: 5-Bromo-l-(p-D-ribofuranosyl)-imidazole.4-carboxamide-5'-monophosphate COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:49 SPRUSON FERGUSON NO., 7147 P. 23/51 117 To a cold (0 solution of 5-bromo- l-(P-D-ribofuranosyl)-inidazole4-carboxamide (0.03 s) (from example 9) in 0.2 rmL of triethyl phosphite was added phosphorous oxychloride (0.026 The mixture was allow to warm to room temperature over 3 hours and diluted with I M aqueous sodium hydroxide solution until the pH reached 8. The mixture wae stirred for hours and passed through Dowex® ion exchange resin, The resin was washed first with water and then with 6 M formic acid solution cluting the product whose NMR spectra is consistent with its structure and a satisfactory elemental analysis.
Example 10.1: 5-Trifluorornethyl-l-(B-D-ribofuranosyl)-imidazole-4-carboxamide To a solution of 3.5 g of AICA riboside in 65 mL of 50% aqueous tetrafluoroboric acid was. added a solution of 1.71 g of sodium nitrite in 2 mL ofwater. The mixture was irradiated with a medium pressure lamp in a quartz tube for 18 hours and cooled to 0 The pH was adjusted to -5 with sodium hydroxide solution and the mixture extracted with ethyl acetate. The organic layer was dried over magnesium sulfate and the solvent removed under reduced pressure.
The residue was chromatographed on silica and eluted with methanol/methylene chloride to MeOH) to give ethyl $-fluoroimidazole-4-carboxylatc, melting point 153-154 *C.
This compound was dissolved in methanol and the solution saturated with ammonia in a steel bomb. The bomb was heated at 100 OC for 48 hours. The bomb was carefully opened and the solvent removed under reduced pressure, and the residue chromatographed on silica to methanol in methylene chloride) to give S-fluoroimidaole-4-carboxamide, melting point 253-254 "C.
This compound (500 mg) was dissolved in hexamethyldisilizane (5 mL) and trimethylehlorosilane (0.9 mL) was added. The mixture was heated to 130 "C for 2.5 hours, cooled and the solvent removed under reduced pressure, The residue was dissolved in 2.4 mL of methylene chloride and added to a solution of l-0-acetyl- 2 ,3,5-tri-O-benzoyl-D-ribose (1.96 g) in methyene chloride. The mixture was cooled to 0 *C and a solution of tin tetrachloride (0.6 mL) in 3.4 mL of methylene chloride was added. The mixture was stirred overnight, diluted with ethyl acetate and extracted with saturated aqueous sodium bicarbonate and with water. The organic layer was dried over magnesium sulfate and the solvent removed. The reside was COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:50 SPRUSON FERGUSON NO. 7147 P. 24/51 118 chromatographed on silica (methylene chloride to 5% methanol/mcthylenc chloride) to give 561 milligrams of coupled product.
The compound was dissolved in methanol and the solution saturated with ammonia and stirred for 18 hours. The solvent was removed under reduced pressure and the residue triturared with ether. The solid was chromatographed on silica (10% methanol/rethylene chloride) to give 150 milligrams of final product.
Other 5-substituted analogs were made by this method of coupling the appropriate substituted imidazole with 1-0-acetyl-2,3.5-tri-O-benzoyl-D-ribose. For example, the compound (mp 179-180 and 5-trifluoromethyl compound (mp 255 *C (decomposition]) were prepared in this manner. The substituted imidazoles were made by the general method of R, Paul, J. Mad. Chem. 1985, 28, 1198-1203.
Preparation of N 9 Step A. A solution of 5-amino-4,6-dihloropyrimidine (1 mmol) in nBuOH was treated with Et3N (1.2 mmol) and neopentylamine (1.05 mmol) at 80 oc. After 12 h, the cooled reaction mixture was evaporated under vacuum and the residue was chromatographed to give anmino-4-(neopentylamino)-pyrmidine as a yellow solid.
icLB..The 6 -chloro-5-amino-4-(2-neopentylamino)pyrimidine (I mmol) in DMSO was treated with 5-diethylphosphono-2-furaldohyde (1.5 mmol) and FeCI3-silica (2.0 mmol) at 80 OC for 12 h. The cooled reaction mixture was filtered and the filtrate was evaporated under vacuum.
Chromatography afforded 6-chloro-N 9 -neopentyl-8-(2(5-diethyl-phosphono)fuanyl)purine as a yellow solid.
StsmiL6-Chloro-N9-neopentyl-8-(2-(5,diethylphosphono)franyl)purne (I mmol) in THF- DMSO was treated with liquid ammonia (2 mL) in a steel bomb. After 12 h, the reaction was evaporated under vacuum and the residue was purified through chromatography to give N 9 neopentyl-8-(2-(S-dithylphosphono)fur-nyl)adenine as a yellow solid.
A solution of N 9 -neopentyl-8-(2-(5.diethylphosphono)uranyl)-adenine (1 mmnol) in acetonitrile was treated with bromotrimethylsilane (10 mmol). After 12 h, the reaction was evaporated under vacuum and the residue was treated with a mixture of water and acetonitrile.
The solid was collected through filtration.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB.2004 17:50 SPRUSON FERGUSON NO. 7147 P. 2 5/51' 119 11.1: N9-neopentyl-n-(2-(i-phosphono)furanyl)adenine. mp 230 OC; Anal. caled. for C14.HIgN$O4P: C; 47.87; H: 5.16; N: 19.94. Found: C: 47,59. IH: 4.92; N: 19.53.
11.2; 2- 2 -Phenylethyl)-S-adeninyljj furanylphosphonic acid. rnp 242 244 OC; Anal.
calcd. for C17HI6N504P 1.3 7H20: C: 50.16; H: 4.64; N: 17.21. Found: C: 48,95; H. 4.59; N: 16.80.
Exam=ees 12.
Preparation af N 9 cylohexvieth y.-84Choshonomhomethoilsdnine Sse&. A mixture of N9-cyclohexylethyl-8-bromoadenine (1 mmnol), cetrakis (rriphenylphosphine)palladium (0.05 mmol), and triethylamine (5 mmol) in DMF in a scaled tube was warmed at 110 oC under 50 psi of carbon monoxide. After 24 It the cooled reaction mixture was evaporated and purified through chromatography to give N 9 -cyclohexylethyl-8methoxycarbonyladenine as a yellow solid.
Steu B.A solution of N 9 -cyclohexylethyl-8-methoxycarbonyladenine (I nnol) in tetrahydroffran was treated with lithium aluminum hydride (1 mmol) at 0 CC for I b. Extruction and chromatography give N 9 -eyclabexylethyl--hydroxymethyladenii as a white solid.
&ZCu.A solution of N 9 -cyclohexylethyl-.-hydroxymethyladnine (1 mmol) in methylene chloride was treated with PBr3 (1 mmol) at 25 oC for I b. Extraction and chromatography give N9-cyclohexylethyl-8-bromomethyi-adenine as a white solid.
Sa2 A solution of N9-cyclohexylethyl-g-bromomethyladenine (I mmol) in IDMF was treated .with a solution of diqthyl hydroxymethylphosphonate sodium salt (1 mmol) in DMF at oC for I h. Extraction and chromatography gave N 9 -cyclohexylethyl-4diethylphosphonomcthoxy-methyladenine as a white solid.
SteR E.N 9 -cyclohexylethyl-8diethylphosphonornethoxymethyladenine was subjected to Step F in Example 1.
12.1; N9-cyclohexylethyl-8-(phosphonomethoxymethyl)adeninc as a white solid. mp 250 oC; Anal. calcd. for C15H24NSQ 4 P 1120: C: 46.51; H: 6.76; N: 18.08. Found: C: 46.47; H: 6.71; N: 17.91.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:50 SPRUSON FERGUSON NO. 7147 P. 26/51' 120 Prearation of 29-isouv--(2-5-hono)fuanvl)uin Stce A: 2 -Methylthio-4,5,&criainopyrimidine and 5-diethYlphosphono-2-furaldehyde was subjected to the procedures of Step B in Example 1 to give 6 -amino-2methylthio-8-(2-(5diethylphosphono)fbranyl)purine as a yellow solid. TLC: Rf- 0.27, 80 EtOAc hexane.
SMLLA solution of 6-amino-2-methylthio--(2-(5-dicthylpphono)-furanyl)purine (1 mmol) in DMF was treated with cesium carbonate (2 rnmol) and isobutyl bromide (1.5 mmol) at 80 OC for 12 h. The cooled reaction mixture was subjected to extraction and chromatography to give 6amino-N 9 -isobutyl-2-mehylthio-8-(2-(S-diethylphasphono)furanyl)purine as a yellow solid.
.TLC: Rf 0.27, 80 EtOAc hexamn.
Se W-AminSN-isobuSyl-2-methylthio-8-(2-(S-diSthyl-phosphono)-anyl)purine was subjected to Step F.
13.1: 6-amino-N 9 -isobutyl-2-methylthio--(2-(5-phosphono)-furanyl)purine as a white solid. mp 220 oC: Anal. calcd. for C14H ISN504PS 0.25 HBr 0.25 EtOAc; C: 42.33; H: 4.8; N: 16.45.
Found: C: 42.42; H: 4.53; N: 16.39.
Eam=1e 14.
Preparadon f To a solution of 168 g(l.75 mol) 2-ftraldehyde in 500 mL toluene was added 215 mL(1.75 mol) ofNN'-dimehylethylene diamine. The solution was refluxod using a Dean Stark rap to remove H20. After 2 hours of reflux, the solvent was removed under reduced pressure.
The resulting dark mixture was vacuum distilled (3 nu Hg) and the fraction at 59-61 'C was collected yielding 247.8 g(85%) of clear, colorless oiL A solution of 33.25 g (0-2 mol) furan-2-(N.N'-dimethyimidazolidine) and 30.2 mL (0:2 mol) tetramethylcnediarnine in 125 rnL THF was cooled ir a dry ioe/IPA bath. A solution of 112 mL n-BuLi in hexane(0.28 mol,2.5M) was added dropwise, maintaining temperature between and -40 'C during addition. The reaction was allowed to warm to 0 'C over 30 minutes and was maintained at 0 'C for 45 minutes. The reaction was then cooled in a dry ice/IPA bath to 0 C. This cooled solution was transferred to a solution of 34.7 mL (0:24 mol) COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:51 SPRUSON FERGUSON NO. 7147" P. 27/51-- 121 diethylchlorophosphate in 125 mL THF and cooled in a dry ice/IPA bath over 45 minurtes maintaining the reaction temperature between .50 'C and -38 The reaction was stirred at rt overnight. The reaction mixture was evaporated under reduced pressure. Ethyl acetate and were added to the residue and the layers separated. The H20 layer was washed with ethyl acetate. The ethyl acetate layers were combined, dried over magnesium sulfate and evaporated under reduced pressure yielding 59.6 g of brown oil.
To a solution of 59.6 g 5-diehylphosphonofuran-2-(N,N'.dimethylimidazolidine) in mL H20 was added I 1.5 raL of cone. H2SO4 dropwisc until pH I was obtained. The aqueous reaction mixture was extracted with ethyl acetate. The ethyl acetaie layer was washed with saturated sodium bicarbonate, dried over magnesium sulfate and evaporated to a brown oil. The brown oil was added to a silica column and was cluted with hexane/ethyl acetate. Product fractions were pooled and evaporated under reduced pressure yielding a dark yellow oil, 28.2 g(62%).
The following general procedures are used in the synthesis of the benzimidazole parent drugs: Examl General methods for the oreparation of substituted 1.2-ohenvyenediamines Step A Brominaion ofnitroanilines.
To a solution of 1.0 mmol of sustituted nitroanline in 10 mL of CHCI3 or a mixture of
CHCI
3 and MeOH(7:I) was added a solution of bromine in 5 mL ofCHCI3 over a period of min. After stirring for 2 days at rt, extractive isolation provided the bromination product.
StopMB.
Reduction of nitroanilines To a solution of 1.0 mmol of substituted niutoatiline in 15 mL of MeOH was added of saturated solution of sodium dithionite. Filtration followed by removal of solvent and extraction with EtOAc provided the pure diamine.
StewC Preparation of 2. .3-benzoselenadiazale.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:51 SPRUSON FERGUSON NO. 7147 P. 28/51 122 To a solution of 1.0 mmol of substituted diamine in 3 mrL of 50% aq. ethanol was added a solution of 1.0 mmol of Se02 in 1.5 mnl of H20. The mixture quickly thickened to a slurry. The solid separated out, was fltered, washed with water, and dried.
Niration of benzoselenadizales To a cold(0 C) suspension of 1.0 mmol of substituted 2,1,3-ben oselenadiazole was added dropwise a solution of 2.0 mmol of HNO 3 in I mL of H2SO4. The resultant suspension was stirred for 2 h at 15 0 C. The dark solution was poured onto ice, filtered, washed with water, and dried.
In the case of 5-fluoro-7-bromo-2,1,3-benzoselenadiazole there were two products in 2:1 ratio, major being the required compound, 4 -nitro-5-fluoro-7.bromo-2,1,3-benzoselenadiazole.
This was extracted with hot toluene from the byproduct, 4 -nitro-S-hydroxy-7-bromo-2;1.3benzoselenadiazole.
Substituted 3-nitro.-.2-nhenvlenediamine nrenhration A mixture of 1.0 mmol of substituted 4 -nitro-2,1,3-benzoselenadiazole in 3 mL of 57% HI was stirred at rt for 2 h. Saturated NaHS03 was added and the mixture was neutralized with concentrated NH3 solution. The product was extracted with CHCI3(5xl 0 mL) and the extracts were washed, dried, and evaporated.
Method B: From 2-nitrohalobnzenes: To a solution of 20 mmol of substituted 2 -halonitrobenzenc in 70 mL ofDMF was added mmol of alkyl or arylamine at 0 After 0.5 h TLC (ethyl acetate/hexane 2:1) indicated the completion of reaction. The reaction mixture was evaporated under reduced pressure. The residue was dissolved in ethyl acetate and washed with water. The organic layer was dried, and evaporated to yield the displacement products.
Method C From 2-nitroanilines: To a solution of 10 mmol of substituted 2-nitroaniline, 20 mmol of alkyl or arylaldehyde, and 60 mmol of acetic acid in 30 mL of 1,2-dlchloroethane was added 30 mmol sodium triacetoky borohydride at 0"C. The reaction was stirred overnight under nitrogen atmosphere and COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:51 SPRUSON FERGUSON NO. 7147 P, 29/51 123 was quenched with saturated bicarbonate solution. The product was extracted with EtOAc (3x75 mL) and the extract was washed, dried and evaporated. The residue was chromatographed on a silica gel column cluting with hexmne-erhyl acetate to yield the producL These nomanilincs can be reduced to 1,2-phenylenediamines by the procedure given in the Example 2, Method A, Step 2.
Example 16.
General Drocedures for alklviation Method A: A suspension of 1.5 mmol cesium carbonate, 1.0 mmol of substituted benzimidazole2.
(5-diethylphosphonate)furan and 1.0 mmol of electrophile in 5 mL of dry DMF was heated at for 1-16 h. Extraction and chromatography provided the alkylation product.
Example 17: General procedures for Pd coupling: Method A: A mixture of 1.0 mmol of bromo substituted diethylphosphonate)fumraylJ]bnzimidazole compound, 2.0 mmol of vinyltributyltin or allytributyltin, and 0.1 mmol of Pd(PPh3)2C12 or Pd(PPh3)4 in 4 mL of DMF was stirred and heated at 900C for 1.16 h. Extraction and chromatography provided the coupled compound.
MethLd B A mixture of 1.0 mmol of bromo substituted diethylphosphonate)firanyl]benzimidazole, 2.0 mmol of propargyl alcohol or any terminal acetylenic compound, 0.1 mmol ofPd(PPh3)2Cl2, and 0.1 mmol of Cul in I mL of EtN and mL of CH3CN was stirred and heated at 50-80'C for 1.16 h. Extraction and chromatography provided the coupled.compound.
Method C: A mixture of 1.0 mmol of bromo substituted diethylphosphonate)furanyl]benzimidazole, 5.0 mmol of substituted phenylboronic acid, 0.1 mmol of Pd(PPh3)4, 5 mL of sat. Na2CO3 and 2 mL of EtOH in 10 mL of diglyme was stirred and heated at 80-90"C for 1-16 h. Extraction and chromatography provided the coupled compound.
The compounds thus obtained can be modified as needed. For example vinyl or propargyl alcohol derivatives can be hydrogenated (see Example 7, Method A) to give the ethyl or propyl COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:52 SPRUSON FERGUSON NO 71417 30/51 124 alcohol derivatives respectively. These alcohol can be fuirther modified as required via aikyl halides (see Example 6) or alkyl sulfonates etc. to number of substituted alkyl compoundsi by subjecting them to nueceophilic substitution reactions (March, Advanced Organic Chemistry, Wiley-lncerscienc;, Fourth Edition, 1992, 293-500). See Example S for the cyclopmpmation Of the vinyl derivative.
CvcloproyvOnain pth.4fnifro-7-vinv].lr.. lr--isobu±Y1-2-(2-_dlerhylnhosnhono-.s.
To a suspension of 1 -0 mmol of 4 -nitro-?-vinyl-S-fluoro-d-sobutyl.2-(2.
diethylphosphono-S-furanyl)bepnalmdazole and 0. 1 nunol of Pd(OAc)2 in 8 m. of ether was added an ether solution of diazorrethane(generated from 3.0 g of l-rnethyl-3-nirro.nitrosoguanidine) at 0 After stirring at rt 20 IL. solvent was removed and the residue was subjected to chromatography to give 4 -nitro-7-cyelopropyl-S-fluoro-l1-isobutyl-2-(2furznvflHenziMidpZo I..
To a cold(0 0 C) solution of 1.0 mmol of 4 -amino-7.(4-hydroxybutyl)-S-fluoru.l in 20 mL. of CH2Cl2 .was added rmp!l of PPh3 and 3.0 mine! of CBC4. After 40 mm.L at rt solvent was removed and the residue was subjected to chromatography to give 4 -amiino-7-(4-broinobutyI)-5-fzuoro-1.isobutyl.2(2.
CC14 gave the corrosponding chaor, compound.
Genral rocedures fouireduction: Am znturc of 1.0 mmol of alkylation product and 20 rug of 10 Pd/C inS5 m. ofIDMF or McOH was hydrogenated using H2 in ballooni for 0.5-16 h. The reaction mixture was filtered through celite and chromnatographad to provide the reduction product as an oil.
COMS 10 No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:52 SPRUSON FERGUSON NO.7147- P. 31/51 125 Method 1: To a solution-of 1.0 nmmol of substituted nitroaniline in 15 mL of MeOH was added of saturated solution of sodium dithionite. Filtration followed by removal of solvent and extraction with EtOAc or CHCI3 provided the pure diamine.
These primary aromatic amines can'also be modified as needed. For example N-acetyl derivetives can be prepared by tr6atment with acetyl chloride or acetic anhydride in presence of a base such as pyridine and mono-, or di-alkylamines can be synthesized by direct alkyladon or by reductive alkylation. General procedures for hoshonate hydrolysis: Examrnple 2 1: TMSBr hydrolysis: To a solution of 1.0 mmol of substituted 2 -((5-diethylphosphonatc)furanyl]benzimida2ole in 5 mL of anhydrous CH2C2 was added 10.0 mmol TMSBr at 0 C. After 16 h stining at rt the solvent and excess TMSBr were removed under reduced pressure. The residue was taken into mL of a 1/5 mixture of acetone/water and was stirred for 16 h at rt. The resulting solid was filtered, washed with water, EtOAc. and MeOH and was dried under vacuum at The following compounds were prepared in this manner: 21.1; 4 -Amino-l-( 3 -carbomethoxybenzyl)-2-[2-(5-phosphono)furanyl] benzimidazole. mp 198-202 °C Anal. Cald. for C20HISN306P: C: 55.55; H: 4.39; N: 9.63. Found: C: 55.12; H; 4.29; N: 9.18, 21.2: 4-Amino-l-( 3 -chloropropyl)-2-(2-(5-phosphono)furanyl] benzimidazole. mp >>250 *C; Anal. Cald. for Cl4HISN304CIP 0.7H20: C: 44.83; H: 4.61; N: 10.37. Found: C:44.50; H:4.29; N:I0.96.
21.3: 4-Amino- l-(3-furnylmcthyl)-2-[2-(5-phosphono)furanyl] benzimidazole. mp >>230 C Mass. Cald. 358; Obs. 358.
21.4: 4-Amino-5-ethyl-l-isobutyl-2-(2-phosphono.5-furanyl) benzimidazoe, mp 220-225 C; Anal. Cald. for C: 51.34; H: 5.95; N: 10.21.
21.5: 4-Amino-5-fluoro-7-chloro- 1-isobutyl-2-(2-phosphono-5-furanyl)benzimidazole. mp 220-225 C; Anal. Cald. for C 5HI6N304FCIP 0.9HBr; C: 12; H: 3.70; N: 9,12, Found: C: 39.15; H: 3.46; N: 8.77 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:52 SPRUSON FERGUSON NO. 7147 P. 32/51 126 21.6: 4-Amino- -l(2-ethyl)penryl]benzimidazol-2-yi-methylenoxymethyI phosphonic acid. np C; Anal. Cald. for CISH24N304P 1/2 H20 2 HBr 1/3 toluene: C: 38.05; H: 5.49; N: 7.78. Found. C: 38.30; H: 5.45; N: 7.34.
21.7: 4-Amino-5-fluoro- Icyclopropylmethyl-2-(2-phosphono-5-furanyl)benzimidazol mp 0 258- 260 C; Anal. Cald. for CISHI5N30 4 PF+0.3H 2 0: C: 50.51; H: 4.41; N: I.78.Found: C: 50.21; H: 4.28; N; 11.45.
21.8: 4-Amino-7-ethyl-5-fluoro-I-isobutyl-2-(2-phosphono-5-furanyl)benjimidazole. np 245- 246 C; Anal. Cald. for CI7H21N30 4 FP+0.4H 2 0: C: 52.55; H: 5.66; N: 10.81. Found: C: 52.40; H: 5.79; N: 10.47.
21.9: 4-Amino-7-(propane-3-ol)-5-fluoro-I-isobutyl-2-(2-phosphono-5-furanyl)benzimidazole.
rnp= 170-173 C; Anal. Cald. for CIgH23NsOSFP+1.0H 2 0: C: 50.35; H: 5.87; N: 9.79.
Found: C: 50.3 1; H: 5.80; N: 9,62.
21.10: 4 -Amino-5-fluoro-7-(3-bromopropyl)-1-isobucyl-2-(2-phosphono-5furanyl)benzimidazole. mp 190-195 Anal. Cald. for CISH22N304FBrP: C; 45.59; H: 4.68; N: 8.86. Found C: 45.87; H: 4.87; N: 8.70.
21.11: 4-Amino-5-fluoro-7-(4-bromobutyl) -isobutyl-2-(2-phosphono-5-furanyl)bmnzimidazolc.
np 200-220 Anal, Cald. for Cl9H24N304FBrP+0.5H20: C: 45.89; H: 5.07; N: 8.45. Found. C: 45.61; H: 5.10; N: 8.20.
21.12: 4-Amino-5-fIloro-7-(3-NN-dimethylpropylamine)-1-isobusyl-2-(2-phosphono-5furanyl)bentimidazole hydrobromide salt. mp 208-212 Anal. Cald. for C2OH28N 4 04FP+1-0 HBr+2.0 H20: C: 43.25; H: 5.99; N: 10.09. Found: C; 43.39; H: 5.74: N: 9.90.
Example 22- HBr hydrolysis: A solution of 1.0 mmcl of substituted 2-(5-dimhylphosphonate)furanyl]benzmidazole in ml of 30 H3r was heazed at 80C for 0.5-3 i. The solvent was removed under reduced pressure and the residue was taken into 3 ml of water- The solid precipitated was filtered washed with water and dried under vaccum at The following compounds were prepared in this manner: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB- 2004 17:53 SPRUSON FERGUSON NO. 7147 P. 33/51 127 22.1: 2-(2,8-Diaza-1.2.3.
4 -tetrahydroacenaphthen-9-yl)furan-5-phosphonic acid. Anal. Cald. for
C
1 4H13N2P0 4 0.5 HBr 0.520: C: 47.54; H: 4.13. N: 7.48. Found: C: 47.33; H: 4.16; N: 7.48.
22.2: 4-Hydroxy-l-isobutyl-2-(2-phosphono-5-faranyl) benzitaidazole. mp 244-245 C; Anal.
Cald. for CISUI7N205P 1.IH20: C: 5059; H: 5.43; N: 7.87. Found: C: 50.33; H: 5.38; N: 7.89.
22.3: 4-Fluoro- I-rcopentyl-2-(2-phosphonofrayl)benzimidazole. Anal. Cald- for C1611N2PO4F 0.1 H20 0.3CH3CO2H: C: 53.58; H: 5.25; N: 7-53. Found: C: 53.84; H: 5.12, N; 7.05.
22.4? 5-Phosphonomethylenoxy- 1,2,3, 4 -tettahydropyrido(1,2-a] benzimidazola. mnp 218-222 0 C; Anal. Cald. for C12HI5N2FO 4 H20 0.9HBr: C: 38.63; H: 4.84; N: 7.51. Found: C: 38.96; H: 4.46; N: 7.41.
22,5: 6-Chloro-.-isoburyl-2-(2-phosphona-5-furanyl) benzimidaole. np 195-200 C; Anal.
Cald. for C15H16CIN204P 0.5HBr C: 45.59; H: 4.21; N: 7.09; Cl 8.97;; C: 46.02; H: 3.86; N; 7.01; Cl; 8.63.
22,6; 5-Chloro--isobutyl-4-methyl-2-(2-phosphono-5-furanyl) benzimidazole". mp 193 1960C Anal. Cald. for C16 HIS Cl N2 04 P 1.67 H20: C: 48.19; H: 5.39; N: 7.02; Found: C: 48.24; H: 5.19; N: 6.85.
22.7: N-(Phosphonomethyl)benzimidazole-2-carboxamide. rp 258-2609C. Anal. Cald. for CgHlON304P 0.15 AcOH: C: 42.28; H: 4.04; N: 15.91; Found. C: 42.60; H: 4.02; N: 15.70.
Preparation of l-isobutyl-4-Amino-5-fluoo7m o -2-r3-Rhosoho(metho=CehYUl btenzimidazole.
£a Svnthesisofdichlhospho methyl aetaldbyde dimethw actal ether To a solution of 1.0 mmol dicthyl (hydroxymethyl)phosphonate, 1.5 mmol of sodium 0 -hydride in 2 mL DMF at 0 C was added a solution of 1.2 mmol of bromoacetaldehyde dimethyl acetal. After 3 h. at r. t. the mixture was diluted with 5 mL of water and extracted with cthar(4 x 15 mL). The combined ether layers were concentrated. The residue was chromatographed on a silica geL column eluting with hexane-erhyl acetate to yield the product.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:53 SPRUSON FERGUSON NO. 7147-P. 34/51--- 128 Ste B.
Preation of -isobutyl4-nitr..fluoro.7.brono.2r3diethvlphoshomthoxvnthvl) 1 benzimidazole- To a solution of 1.0 mmol of 2-nitro-3-fluoro-5-bromo-6-isobutylaminc aniline and mmol of diethylphospho methyl aceraldehyde dimothyl acetal ether in 5 mL THF at 0 "C was added 0.5 mL of 10 H2504 and the mixture was heated at 75 °C for 40 min. Solvent wa removed under reduced pr-ssurc, diluted with water and exracted with EtOAc. The combined EtOAc layers were concentrated. The residue was chromatographed on a silica gel column yield the product.
StP.C A solution of 1.0 mmol of this coupled product and 1.0 mmol of 12 in 5 mL of ethanol was stirred at rt for 1-16 Extraction and chromatography provided the title compound as an orange solid.
Prcoaration of 1 -isobutvl-4-amino-5. -luoro- 7 -bromo-2.r3-dithvlh ho methoxvmethyl)l benzimidazole- Followed the procedure given in the Example 20. Method B.
Followed the procedure given in example 21.
23.1: 4-Amino-5-floro-7-bromo-l-isobutyl-2-(l.mcthoxymethyl-3-phosphono)benzimidazole.
mp 200-202 Anal,. Cald. for Cl3HIgN304FBrP: C: 38.07; H: 4.42; N: 10.24.
Found: C: 37.87; H: 4.36; N: 10.15.
Exarnple 24.
Prenaration ofdiethvl 2 4 -methyvvalervlyfiranhosphonate, sL.A A solution of 2 -tributylstannylnjran (I runol), 4-methylvaleroyl chloride (1.1 mmol), and PdCl2(PPh 3 2 (0.05 mmol) in THF was sdrred at 25 oC for 24 h. Extraction and chromatography gave 2 -(4-methylvaleryl)furan.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:53 SPRUSON FERGUSON NO. 7147- P. 35/51 129 atLA solution of 2 -(4-methylvaleryl)furan (1 mmuol) and N,N-dimethylhydrazine (1.5 mmo) in EtOH was heated at reflux for 24 h. Evaporation and distilation give 2-(4--rnthylvakeryl)furandimethylhydrazone as a brown oil.
Steo!C.A solution of 2-(4-mcthylvalery1)furan dimethylhydrazone (I mmol) in THF was cooled to -78 OC and treated with LDA (1.2 mmol) dropwisely. After 1h diechyl chlorophosphate (1.2 mmol) was added, and the resulting misture was stirred at -78 OC for th. The reaction was quenched withbrine, and subjected to extraction and chromatography to give diethyl methylvalcryl))fhranphosphonate dimethylhydrazone.
Step D. A solution of diethyl 2 -(S-(4-methylvaleryl))furanphosphonate dimethylhydrazone (1 nmol) in THF-pH -7 phosphate buffer was treated with CUCI2 tnmol) at 25 OC for 24h. Extraction and chromatography gave diethyl methylvaleiyl))furanphosphonate as a brown oil.
EZample Preoarationof5-chloro-3-isobvI-2-(2-hobosonourunv1linMlL A.pA A mixture of 4-chlorophenylhydrazine hydrochloride (1.5 mmol), diethyl methylvakryl))furanphosphonate (1 mmol), and two drops of concentrated sulfuric acid in glacial acetic acid was heated at reflux for 4 h. The cooled reaction mixture was evaporated to dryness, and the residue was subjected to extraction und chromatography to give 5-chloro-3isobutyl-2-(2-(5-dicthylphosphono)furanyl)iidole as a yellow sticky solid. TLC: Rf 0.30, 50 EtOAc-hexane.
&Srp..D.,A soluion of 5-chloro-3-isobutyl-2-(2-(5-diethylphosphono)-furany)indole (I nmol) in acetonitrile was treated with bromocrimethylsilane (10 imol). After 12 h, the reaction was evaporated under vacuum and the residue was treated with a mixture of water and acetanitrile.
The dark green solid was collected through filtration.
25.1: 5-chloro- 3 -isobutyl-2-(2-(5-phosphono)futranyl)indole.mp 135 139 o0 Anal. cald. for Cl6H17NO4PCI 0.75H20: C: 52.33; H:5.08; N:3.33. Found: C: 51.96; H: 4.93; N:3.81.
meolso 26, PEmarationof9ar--sb l-meh-22(5hoholuavinoe COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:54 SPRUSON FERGUSON NO. 7147 P. 36/51 130 Step A A mixture of dicthyl 2.(5-(4-methylvaleryl))furanphosphonate (1 unnoL) and CuBr2 (4 mmol) in EtOAc-CHCl3 was stirred at 25 0C for 24 h. The reaction was quenched with saturated ammonium chloride. Extraction and chromatography gave 2-(5-(2-bromo-4riethylvaleryl))furanphosphonate as a yellow oil.
St= A solution of 2-amino-6-methylpyridino (1 unmol) and 2-(5-(2-bromo-4methylvalryl))furanphosphonate (1.2 mmol) in n-butanol was heated at reflux for 16 h- The cooled reaction mixture was evaporated to dryness, and the residue was subjected to extraction and chromatography to gite 9-aza-3-isobutyl-7-methyl-2-(2-(5-diethylphosphono)uryl)indole as a brown solid.
_9-Aza-3-isobutyl-7-methyl-2-(2-(5-diethylphosphono)furanyl}-indole was subjected to procedure given in Example 21.
26.1: 9-azn-3-isibutyl-7-mithyl-2-(2-(5-phosphono)furanylindole. mp 225 227 OC. Anal.
cald. for CI.6Hi9N204P IHBr: C: 46.28; H: 4.85; N:6.75. Found: C: 46.23; H: 4.89; N.6.57.
Example 27: Preparation of 2'-azidomethylene and 2'-amingoetylen susiue yci '3-rXI cemr: 6-Chloro-1-isobutyl-2- {542'.(bydrxymethy)-propan-l',3'-ylphosphono-2ftranyl}bcnzimidazole was prepared as described in Example 7.
SxM.To a solution of 6-Chloro-1-isobutyl-2- {5-[2'-(hydroxymethyl)-propan-l',3'yl]phosphono-2-furanyl)bcnzimidazole (300 rg. 0.70 mmol) in dichloromethane (5 mL) was added pyridine (0.12 rnL, 1,4 rtunol) and methanesulphonyl chloride (0.066 nL, 0.84 nnot).
The reaction was stirred overnight and concentrated under vacuum. Chromatography by elution with 5% methanol-dichloromothane resulted in 340 mg (95%)of pure mesylated product.
$M To a solution of mesylate (100 mg, 0.19 mmol) in DMF (2 mL) was added sodium azide mg. I nmol). The mixture was heated to 55* C for 5h, The reaction was concentrated and diluted with ethyl acetate (50 rmL) washed with water and dried. Chromatography by clution with 5% methanol-dichloromethane gave in 35 mg (39%)of pure product.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:54 SRSN&FRUO O 17 P SPRUSON FERGUSON NO.7147 P. 37/51 131 27.1: 6-Chlaro-lI-isobutyl-2- {5-[2'-(azidomethyl)-propan-lI',3'-yllphosphono-2fiuranyl~bcnzirnidazole. mp 167 168 0 C, Anal. cad. for CI9H21INSO4P: C: 50.73; H: 4.71;' N: 15.57. Found: C 50.74; H: 4.72 N; 15.22.
Step D: Azide (100 mg., 0.22 nol) was dissolved in ethyl acetate (5 rnL) and 10% Pd-C mg) was added. The mixture was stirred wider hydrogen (or l16h, Catalyst was filtered off through a celite pad. The filtenate was concentratd and chromatographed by eluting with methanol-dicbloromethane to give pure amine (45mg, 48%).
27.2: 6-Chloro- 1 -isobutyl-2- {5-(2'-(aminomethyl)-propant',3-y]phosphono-2-furmnyl) benzimidazole. rap 158 160 0 C. Anal. cald. for Cl9H23C1N3O4P+1-25H20 C: 51.13; H: 5.76; N: 9.41. found: C: 51.35; 5.48; N; 9.05.
Bxqjnple 28: Prenaration oficyclic 1'Y3-DrouvI esters of 9-r(2-o~hon ohXvlethvll adenine (PMEA), STAU 66: To aL solution of cis.cis-l1.3,5-cylohexanetuiol (1.65 8g 10 mmol) in DM50 niL) was added 60% sodium hydride mnineral oil dispersion (400 mug, 10 ninol). The reaction was stirred at room temperature for 12h. After work-up, the mixture was chrornatographed by eluting with ethyl acetate-mechylene chloride (1 to yield 800mg (3 of monobenzylated, product 2Sp..flTo a suspension of 9-[(2-phosphonomethoxy)ethyl]adenine (Collect. Czech. Chern.
Commun., 1990, 55, 808) (1 g, 3.5 mmcl) in dichloromethane (10 niL) was added trirnethylsiidiethyl amine (3 rut. The reaction ws stirred at room temperature for 2 h and evaporated to dryness. The residue was dissolved in dicbloromcthane (10 rut). DMF (0.05 ruL) followed by oxalyl chloride (0.9 nt) were added at 01C, The reaction was stirred at O0C for lb and an additional lbh at room temperawure. The mixture was concentrated under reduced pressure and the residue was dissolved in pyridine (20 mU). Monobeuzyl cyclohexyl trio] was added and stirred for 1 6h at room temperature. The reaction mixture -was concentrated and chromatographed by eluding with methanol-netbyle chloride to give $00 mg of pure prodrug.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-2-20 FEB. 2004 17:54 SPRUSON FERGUSON NO. 7147 38/51 132 Step C:To a solution of monobenzyl prodrug (500mg) in DMF (10mL) was added i0%Pd(OH)2- C (100mg). The reaction was stirred under hydrogen gas for 16h. The catalyst was filtered off through celite and the reaction mixture was concentrated. Chromatography of the residue by eluting with aqueous NH3-MeOH-dichloromethane (1:20:80) resulted in 200mg of product.
The following compound was prepared in this manner: 28.1: 9-(2-(l'-Hydroxy-3',5'.-cyclohexylphosphonomcthoxy)cthyl] adenine. mp 171 174 'C; Mass Cald. for C 14H20NOS5P: M*1'i 370 Found: MH-370 The following compounds were prepared following the procedure of step C: 28,2: 9-[2-(2'-Hydroxymncthyl- l',3'-propylphosphonomcthoxy)ethyl] adenine. mp 148 151 Mass Cald. for C12H18NSOSP: MH+ 344: Found: MH344 28.3: 9-[2-(l'-phenyl-l',3'-propylphosphonomethoxy)ethyl] adenine. Mass Cald. for C17H20N504P: MH* 390 Found: MH'390.
28.4: 9-[2-(l(4-Pyridyl)-1,3-propylphosphonomethoxy)ethyl]adenine. Anal. Cald. for C16H19N604P: C: 49.23; H: 4.91; N: 21.53. Found: C: 49.01; H: 5.01; N: 19.37.
Example 29: General procedure for formation of 1homshate Drodrues from chlorophosoholane: (Bloorg. Med. chem. Let., 1997, 7. 1577) Step A Variety of substituted 1,3-diols arc commercially available. Diols which are not available are made by procedures described in examples 3 (step 4 (step A and 5 (step A, B and 6 (step A and B) and 7 (step A).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:54 SPRUSON FERGUSON NO. 7147 P. 39/51 133 Cyclic phospholanes are prepared by equimolar addition of IM phosphorus trichloride in dichloromethane to propane-l,3-diols at 0" C. The resulting mixture was warmed and stirred at room temperaure for 3h The reaction was concentrated under vacuum and dried to give crude chlorophospholane which is used for addition in next step without further purification.
To a solution of ara-A or oehcr nucleoside or other alcohol containing drug(1 mmol) in DMF mL) was added triethylamine (2 mmol) at -40' C. To this mixture was added crude cyclic chlorophospholane (1.5 mmole) in 2 mL of DMF. The mixture was warmed to room temperature and stirred for two hours. The reaction was cooled back to -400 C and t-butylhydroperoxide (2mmol) was added and left at room temperature overnight. The reaction was concentrated and the crude mixture was chromatographed on a silica gel column to give pure cyclic prodrug product.
The following compounds were prepared in this manner: 29.1: Adenine-9-bea-D-arabinofuranoside-S'-[2'-acetoxymethyl-1',3'-propyl]monophosphate.
mp v 118 120 Anal. Cald. for C16H22N509P+i.0H20: C: 40.26; H: 5.07; N: 14.67.
Found: C: 40.08; H: 4.84; N: 14.67.
29.2 Adenine-9-beta-D-arabinofianoside-5'-[I'-phenyl-l',3'-propyl]monophosphate. nip 122 125 Anal. Catd. for C19H22N507P+1.5H20+0.15CH2Cl2: C; 45.71; H: 5.07; N: 13.92.
Found: C: 45.43; H: 4.64 N: 13.92.
Example General procedure for formation of rodniru by chliorhosohoramidite method; Substituted diols are obtained as described in step A of example 29.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:55 SPRUSON FERGUSON N0.7147 P. 40/51 134 Preparation of cyclic nhosphoramidite fronm substituted iols.
(Ter., 1993, 49, 6123) To a solution of commercially available diisopropyl phosphoramidous dichloride (lmmoo) in THF (SmL) was added 1,3-diol (lmmol) and triethylamine (4mmol) in THF (5mL) at -780 C over 30 min. The reaction was slowly warmnned to room temparamure and left stirring overnight.
Reaction mixture was filtered to remove salts and filterate was concentrated to give crude product. Silicagel column chromatography provided pure cyclic diisopropyl phosphoramidite of 1,3-diol.
Addition of cyclic phospboramidite and oxidation: Org. Chem., 1996, 61, 7996) To a solution of nucleoside (Immol) and cyclic phosphoramidite (Immol) in DMF (10 mL) was added benzimidazolium triflate (1mmol). The reaction was stirred for 30 min at room temparature. The mixture was cooled to -40° C before addition of t-butylhydro peroxide (2nmmol) and left at room temperature overnight. Concentration under reduced pressure and chromatography of crude product resulted in pure cyclic propyl prodrug.
The following compounds were prepared in this manner 30.1: Adenine-9-beta-D-arabinofuranoside-5'. I -(4-pyridyl)-1,3-propyl]monophosphate.
p>220 Anal. Cald. for C18H21N607P+1H20+0.25PhCH3; C: 46.93; H: 4-99; N: 16.63.
Found: C: 47.42 H: 4.27; N: 16.74.
30.2: 4 -pyridyl)-1,3-propyllphosphoryl}. 3'-dideoxyinosme. mp=133 135 Anal.
Cald. for C18H20N506P+1 H20: C: 47.90; H: 4.91; N: 15.52, Found: C: 48.06; H: 4.64; N: 15.49.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:55 SPRUSON FERGUSON NO. 7147 P. 41/51 135 30.3: {[l1-(4-pyridyl)-1,3-propyl]phospharyl) ribavirin. mp-162 164 OC; Anal. Cald. for CI6H20NSO5 P+1.5H20+0.2CH2Cl2; C; 40.09; H: 4.86; N: 14.43. Found: C: 40.42; H: 4.63; N:' 14.63.
30.4: [l-(4-pyridyl)-1,3-propyl]phosphoryl }-2-fluoroadenine-9-b-D-arabinofuranoside.
mp>210 Anal. Cald. for C18H20FN607P+1.5H20+0.iPrOH+0.2CH2CI2: C: 41.74; H: 4.58; N: 15.79. Found: C: 41.55; H: 4.15; N: 15.76.
30.5: -(4-pyridyl)- 1,3-propyl]phosphoryl)-3'- 1-(4-pyridyl)-1,3-propyl]phosphoryl}-5fluoro- 2'-deoxy uridine. mp=200 204 Anal. Cald. for C25H27FN401 l1P2+l.5H20+03CH2Cl2+0.1i-PrOH: C: 43.99; H: 4.53; N: 8.02. Found: C: 43.56; H: 4.06; N; 7.97.
30.6: -(4-pyridyl)-1,3-propyl]phosphoryl)- 3'-dldeoxyadenosine. mp=98 100 Anal.
Cald. for CISH21N605P+1.6H20: C: 46.88; H: 5.29; N: 18.22. Found: C: 47.21; H 5.11; N: 17.76.
30.7: [-(4-pyridyl)-1,3-propyl]phosphoryl -5-fluoro-2'-deoxy uridine. mp=120-124 *C.
Mass. Cald. for CI7H19N308PF. MNa+: 466. Foumd: 466.
30.8: 9-{[l-(4-pyridyl)-1.3-propyl]phosphoryl) [2(methylenoxyethoxy)J-guanine. Mass. Cald.
for Cl6H19N606P. MNa+; 445. Found: 445.
Examples of use of the method of the invention includes the following. It will be understood that these examples are exemplary and that the method of the invention is not limited solely to these examples.
For the purposes of clarity and brevity, chemical compounds arc referred to as synthetic example numbers in the biological examples below.
Besides the following Examples, assays that may be useful for identifying compounds which inhibit gluconeogenesis include the following animal models of Diabetes: i. Animals with pancreatic b-cells destroyed by specific chemical cytotoxins such as Alloxan or Streptozotocin the Streptozotooin-treated mouse, -rat, dog, and -monkey).
Kodama. Fujita. Yamaguchi, Japanese Journal of Pharmacology 1994, 66, 331-336 (mouse); Youn, Kim, Buchanan, Diabetes 1994, 43. 564-571 (rat); Le Marchand, Loten, Assirnacopoulos-Jannet, et al., Diabetes 1978, 27, 1182-88 (dog); and Pitkin, ILM., Reynolds, Diabetes 1970, 19. 70-85 (monkey).
COMS ID No: SMBI-00629827 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:55 SPRUSON FERGUSON NO. 7147 P. 42/51 136 ii. Mutant mice such as the C57BL/Ks db/db, C57BLKs ob/ob, and C57BL/6J ob/ob strains from Jackson Laboratory, Bar Harbor, and others such as Yellow Obese, T-KK, and New* Zealand Obese. Coleman, D.L, Hwnmel, Diabetologia 1967, 3,238-248 (C57BL/Ks db/db); Coleman, Diabetologia 1973, 14, 141-148 (C57BL/6J ob/ob); Wolf Pitot, Genetics 1973, 73, 109-123 (Yellow Obese); Dulin, Wyse, Diabeiologia 1970, 6, 317-323 and Bielschowsky, Bielschowsky, F. Proceedings of the University of Otago Medical School, 1953, 11, 29-31 (New Zealand Obese).
iii. Mutant rats such as the Zucker fa/fa Rat rendered diabetic with Streptozotocin or Dexamethasone, the Zucker Diabetic Fatty Rat, and the Wistar Kyoto Fatty Rat. Stolz, KJ., Martin, RJ. Journal ofNutridon 1982, 12, 997-1002 (Streptozotocin); Ogawa, Johnson, Ohnbeda, McAllister, Inman, Alam, Unger, The Journal of Clinical Investigation 1992, 90,497-504 (Dexamethasane); Clark, Palmer, CJ., Shaw, W.N., Proceedings of the Society for Experimental Biology and Medicine 1983, 173, 68-75 (Zucker Diabetic Fatty Rat); and Idida, Shino, Matsuo, et al., Diabetes 1981, 30, 1045-1050 (Wistar Kyoto Fatty Rat).
iv. Animals with spontaneous diabetes such as the Chinese Hamster, the Guinea Pig, the New Zealand White Rabbit, and non-human primates such as the Rhesus monkey and Squirrel monkey. Gerritsen, ConneJ Blanks, Proceedings of the Nutrition Society 1981. 40, 237 245 (Chinese Hamster); Lang, Munger, Diabetes 1976, 25,434-443 (Guinea Pig); Conaway, Brown, CJ., Sanders, L.L, eta Journal of Heredity 1980, 71, 179-186 (New Zealand White Rabbit); Hansen, Bodkin, Diabetologia 1986, 29, 713-719 (Rhesus monkey); and Davidson, Lang, CM.. Blackwell, Diabetes 1967, 16. 39S-401 (Squirrel monkey).
v. Animals with nutritionally induced diabetes such as the Sand Rat, the Spiny Mouse, the Mongolian Gerbil, and the Cohen Sucrose-Induced Diabetic Rat. Schmidt-Nielsen,
K.,
Hainess, Hackel, D.B, Science 1964, 143, 689-690 (Sand Rat); Gonet, Stauffacher, Picter, R, et al., Diaberologia 1965, 1, 162.171 (Spiny Mouse); Boquist, Diabetologia 1972, 8, 274-282 (Mongolian Gerbil); and Cohen, Teitebaum, Saliternik, R., Metabolism 1972, 21,235-240 (Cohen Sucrose-Induced Diabetic Rat).
vi. Any other animal with one of the following or a combination of the following characteristics resulting from a genetic predisposition, genetic engineering, selective breeding, or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:56 SPRUSON FERGUSON NO. 7147 P. 43/51'" 137 chemical or nutritional induction: Impaired glucose tolerance, insulin resistance, hyperglycemia, obesity, accelerated gluconeogenesis, increased hepatic glucose output.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:56 SPRUSON FERGUSON NO.7147 P. 44/51 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:56 SPRUSON FERGUSON NO. 1147 45/51- -Synthello, Group V No. 7.10 1 245*Cdoro-4- mgthyl. Mefliosyca i noraisomer JisobnlvrbanzImidazai-zivI)I~uan] oIm 4. 2-l5-Chlaro-4-ma-thyl-1. 770idy H F H IisabutvenzimidazoI.2.y~furanm.
7.9 2-J41 -Ipahwty-4.5,6.7. H mtoyfb ni H major isomher tolramutiylbenimildazol-2- xef yllfuran-8-yI a 7.8 '2-11 -4sobuty-4,5.t7'. M nufxyho H f H mner isomr twtrunithyIbsuuimicao-- awl4-n 4.4 2-41-ImbutyI-45,6.7- 4.pyfidy H 16auamlethwlbenimidazaI-2-I 28.3 1Adenine- 9-ethwleneezyrnth (R)henylHH H 28 2 1Adbnine9-quhIylnhokymetnyl 11 hydV~ymedm I H 4A -GAleOnonlullU 2-ylp H Pf 7.7 !2.(4-Aaino.7.ahyl.B4tIueS.1. bnmPrxyeulyl j Hi ;isobutybenzimidazal-2-ylfwan.- 2,3-4lro t H H I1aobutvinunzimIwazei.2.YIitugen.
29.2 lAdia-S-bat (R>Hw,1 II H F f aabnumwnwaos- 1 1Y 29.3 Aduing-beu-o- H "Mutwnl H NaWifurwd,4*
IY
7mw- k-414-Amiiia-7-ouwIy-w-kacro-1- H eymelahanocb H H COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:56 SPRUSON FERGUSON NO. 7147--P. 46/SFsnhtcOopvZ w Oa~steremgcs 37.2 2-IG-Chlao-1. H Iamflarmmyl H H isobut~ylbwimiidazol-2-yIlfurmn- 271 Z.-Chlorg-- II 3Sionwh-iI I H
L
7.4 2.(4.AAna-7-ulhyl-5-Fhno-1i- H mcauwrmty H H Isqtltmnzmidaal-2-vyluuran.1 73 12-I&Oic-1. 2 y~l r H m thaxy~t bnyt 8 H- Vi1
I
712 2.l-&Ch~ro-1. H H -kolsm iQburybehwizoi.2vlurw%.fion aI hgraznuiyl K 5.V1 7.2- 12.(Chioro-1. K-iMO4 a II N H minor aeK 6ablutylbmnimidazal-2.yflfrw ehxpa) 5.1 2.IG.CNAO-- .bo-d. HH 14 mjriaa lsgbutvlbenzbnliduz*d2-flftam-meixpwj, 246.chloro-1I- N-axo-2pr*dv mio H Hm l*0uabutyeilmldazoI-2-yQ~flhn-- COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-2-20 FEB. 2004 17:57 SPRUSON FERGUSON NO. 7147 P. 47/51 Snteo I Group i V 2 w w O'want Fgample 4.2 Ib(btJWoroflZ.a dzoI-yltumn H H H 1 5 -Co 1 aoI iaobqunylbunzimideol.2.yllfun ImethlthiaphIy i.6 1 2-l5.hor. wanol-2 phinylv 2.4 2-(O-Chiioro-I 4-Ahkfpw wActylamine H I m" mer iS~buYlbiflzlmidaz&l2.Yllfurmin- 2.4 i6-Chlora-l-I (-pnplfyl Hay~ M minor lroner isobtylbnzimlduS.l2.yllttran-I 2.2 te f-6-~or1 H ii minor homer lgabsylbnimlduz-, yfwn COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:57 SPRUSON FERGUSON NO, 7147 P. 48/51 142 BIOLOGICAL EXAMPLES Example A: Chemical Stability The stability of 30.1 was assessed in isotonic saline and in phosphate buffer (pH 3, 7, and 9), and that of 1.3 in phosphate buffer at pH 7.4.
Methods: Aliquots of a 10 Mg/mL solution of 30.1 in isotonic saline and in 100 mM potassium phosphate buffers at pH 3, 7, and 9 were sampled after 1, 2, 5, and 7 days of incubation at room tempeature and analyzed by HPLC. A Beckman Ultrasphere CS column (4.6 x 150 mm) was employed and eluted with a gradient from 0.1% v/v trifluoroacetic acid to 80% methanol at a flow rate of 1.0 mlJmin. Detection was at 254 ranm. Using these conditions, the four isomers of 30.1 were readily separated and quantified. 13 was incubated at 100 pM in phosphate buffer at pH 7.4 and analyzed by HPLC as described in Example I following I hour of incubation at room temperature.
Results: No decomposition of 30.1 was noted either in saline or buffer throughout the 7-day evaluation period. The results demonstrate that 30.1 is stable for a minimum of seven days, 1.3 was found to be fully stable for the 1 -hour incubation period tested. Thus, prodrugs of the invention are stable under a broad pH range.
Example B: Stability to Esterases and Phosphatases The stability of select prodrugs to cleavage by purified esterase and phosphatase preparations is assessed.
Methods: Carboxylesterase (porcine liver) and alkaline phosphatase (calf intestinal mucose) are purchased from Sigma Chemical Co. (St. Louis, MO). Carboxyl esterase activity is measured in 0.1 M Tris-HCI buffer at pH 8.0. Activity towards p-nitrophonyl acetate, a known substrate and positive control in the reactions is measured as described for example by Matsushima at al. (FEBS Lett. (1991)293(1-2): 37-41]. Alkaline phosphatase activity is measured in a 0.1 M diethanalamina buffer, pH 9.8, containing 0.5 mM MgCI2. Activity towards p-nitrophenyl phosphate, a known substrate and positive control in the reactions, is measured as described Brenna et at (1975) Biochem J. 151(2): 291-6]. 1.3, 28.4, and 30.1 are incubated at a concentration of, for example, 250 jIM in appropriate reaction mixtures containing carboxylesterase or alkaline phosphatase. Parallel reactions are run with known substrates of the enzymes as described above. Aliqouts are removed from the COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:57 SPRUSON FERGUSON NO, 7147 P. 49/51- 143 reaction mixture at various time points and the reaction stopped by addition of methanol to Following centrifugation and filtration, the aliquots are analyzed for generation of parent compound by HPLC. Ara-AMP and PMEA, are analyzed as described in Example G. 6- Amino-9-neopenryl-8-(2-phosphonofuranyl)purine is quantified as described in Example I.
Results: Insusceptibility to cleavage by carboxylesterase or alkaline pbosphatase is indicated by the absence of parent compound and presence of intact prodrug in the samples.
Example C: Stability in Plasma The stability of 30.1, 1.t.and 1.3 was assessed in freshly prepared rat plasma.
Methods: Compounds were incubated in plasma at 37.C. and aliqouts removed at appropriate time points over the course of 5-8 hours. The aliquots were extracted with 1.5 volumes of methanol and clarified by centrifugation. Supernatants were then evaporated to dryness and the residues reconstituted with 100 gL of isotonic saline and then analyzed by reverse phase HPLC.
Results: There was no evidence of metabolism of 30.1, 1.1, or 1.3 during the incubation period. These findings demonstrate that these prodrugs are not susceptible to cleavage by plasma esterases, adenosine deamrinase, or other plasma enzymes.
Example D: Activation by Rat Liver Microsomes 7.1 was, and 30.1 and 28.4 are tested for activation to their respective parent compounds in reactions catalyzed by the microsomal fraction of rat liver.
Methods. The microsomal fraction was prepared from fresh, saline-perfused rat liver. Liver tissue was homogenized in three volumes of 0.2 M KIHzPO, buffer pH 7.5, containing 2mM MgCI 2 and I mM EGTA. The homogenate was centrifuged at 10,000 g for 1 hour and the supernatant recovered. The supematant fraction was then recentritged at 100,000 g to pellet the microsomal fraction. The pellet was resuspended in homogenization buffer and recentrifuged.
This process was repeated twice to ensure complete removal ofcytosolic enzyme activities. After the last centrifugation, the microsomal pellet was resuspended in homogenization buffer at a final procein concentration of about 14 mg/ml.
For 7.1, reaction mixtures (0.5 ml) consisted of 0.2 M KH 2 PO0 pH 7.5. 13 mM glucose-6phosphate, 2.2 mM NADP*, I unit of glucose-6-phosphate dehydrogenase, 0-2.5 mg/mn microsomal protein and 100 1M 7.1. Reactions were carried out at 370C. Aliquots were removed from the reaction mixtures at appropriate time points, and extracted with COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:58 SPRUSON FERGUSON NO. 7147 P. 50/51 144 methanol. The methanolic extracts were centrifuged at 14,000 rpm, and filtered (0.2 4M) prior to analysis by HPLC as described in Example I. Eluted peaks were quantitated relative to authentic standards of 6-Chloro-l-isobutyl-2-(2-Phosphono-5-furanyl)benzimidazole of known concentration.
28.4 and 30.1 arc evaluated essentially as described for 7.1 above. The formation of parent compounds PMEA and ara-AMP is monitored as described in Example G. Alternatively, the activation of 28.4 and 30.1 can be monitored by the depletion of NADPH, an essential cofactor in the reaction. This assayis performed in reactions mixtures consisting of 0.2 M KH 2 POQ, 0.22 mM NADFH, 0-2.5 mg/ml microsoral protein, and 100 pM 28.4 or 30.1. Reactions are monitored spectrophotometrically at 340 nm. A decrease in absorbance is indicative of cofactor depletion and thus of the enzymatic oxidation of prodrug to parent compound.
Raesuts: 7.1 was converted to parent compound in the presence, but not in the absence of, NADP* (this cofactor is enzymatically reduced to NADPH by the dehydrogenase present in the reaction mixtures). This result indicates that an oxidative step was involved in the activation of the prodrug. The rate of activation of 7.1 to parent compound was found to be linearly dependent on microsomal protein concentration, confirming that activation occurs by an enzymedependent mechanism. Similar results are found for 30.1 and 28.4.
Example E: Activation by Human Liver Microsomes 7.1, 13, and 30.1 were tested for conversion to their respective parent compounds by the microsomal fraction of human liver.
Methods: Reaction mixtures (0.5 ml 37°C) consisted of 0.2 M KH 2 PO4, 13 mM glucose- 6-phosphate, 2.2 mM NADP', I unit of glucose-6-phosphate dehydrogenase, 0-2.5 mg/ml human microsomal protein (In Vitro Technologies, Inc.), and 250 IM of 7.1, 1.3, or 30.1- The activation of 7.1 and 1.3 to parent compound was monitored by HPLC as described in Example I. Ara-AMP generated by activation of 30.1 was detected by reverse-phase, ion-pairing HPLC as follows. Reactions were stopped by addition of methanol to a concentration of 60%, filtered (02 pM filter), and lyophilized. Samples were resuspended in HPLC buffer (10 mM phosphate pH 2.5 mM otyl-triethyl-anummonium), loaded onto a YMC CS HPLC column (250 x 4.6 numm), and eluted with a methanol gradient to 80%, Formation ofara-AMP was confirmed by coelution with an authentic ara-AMP standard.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:58 SPRUSON FERGUSON NO, 714) P. 51/51 145 Results: The rate of formation of parent compound from 1.3 and 7.1 was determined to be 0.55 and 0.85 nmoles/mgjnicrosomal protein/minute, respectively, The reaction rate observed for the formation of ara-AMP from 30.1 was 0.12 nmoles/min/mg of microsomal protein. All three prodrugs were thus transformed to their respective parent compounds by an NADPHrequiring microsome-catalyzed reaction.
Example F: Identification of the p450 Isozyme Involved In the Activation 30.1, 1.3, and 7.1 were evaluated for human microsomr-catalyzed conversion to parent compound in the absence and presence of specific inhibitors of three major p450 isozymes: ketoconazole (CYP3A4), furafylline (CYPIA2), and sulfaphonazole (CYP2C9).
Methods: Reactions (0.5 ml 37C) consisted of 0.2 M KH?0 4 13 mM glucose-6phosphate, 2.2 mM NADP*, I unit of glucose-6-phosphate dehydrogenase, 0-2.5 mg/ml human microsomal protein (In Vitro Technologies, Inc.), 250 pM prodrug, and 0-100 gM p450 isozyme inhibitor. Ara-AMP, parent compound of 30.1, was detected by reverse-phase, ion-pairing HPLC. Reactions were stopped by addition of methanol to a concentration of 60%, filtered (0.2 pM filter), and lyophilized. Samples were resuspended in HPLC buffer (10 mM phosphate pH 5.5,2.5 rmM octyl-triethyl-ammonium), loaded onto a YMC C8 HPLC column (250 x 4.6 rmm), and cluted with a methanol gradient to 80%. Formation of ara-AMP was confirmed by coelution with an authentic ara-AMP standard. Generation of parent compound from 1.3 and 7.1 was determined as described in Example I.
Results: 30.1 was converted readily to ara-AMP in human liver microsomes. The reaction rate observed for the formation of am-AMP was 0.12 nmoles/min/mg of microsomal protein.
Ketoconazole inhibited the formation of ara-AMP in a dose-dependent fashion; 95% inhibition was observed at I jM, a concentration known to specifically inhibit CYP3A4. The other inhibitors, furafylline and sulfaphenazole, showed no significant inhibition. The results indicated that CYP3A4 is the primary p450 isoform responsible for 30.1 activation in human liver.
Similar results were obtained with 1.3 and 7.1 in human microsomes.
Example G: Activation of 30.1 and 28.4 by Recombinant CYP3A4 Activation of 30.1 and 28.4 was evaluated in reactions containing microsomes from baculovirus-infected insect cells co-expressing recombinant CYP3A4 and cytochrome p 4 reductase (Panvera Corp., Madison, WI).
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:59 SPRUSON FERGUSON NO. 7148 P. 2/51 146 Methods: Reaction mixture composition was similar to that described in Example E.
Reactions were terminated by addition of methanol to a final concentration of 60%, and producrg typically analyzed by HPLC with use of a YMC reverse phase CS column (250 x 4.6 nmm).
Samples were loaded onto the column in mobile phase A (10 mM phosphate pH 5.5, 2.5 mM octyl-triethyl-ammoniwn) and eluted with a gradient of mobile phase B (methanol) to 60% over minutes. The effluent was monitored at 254 nm.
Results Activation of 30.1 and 28.4 to their respective parent compounds was found to be dependent upon the presence of NADPH, and to be linear for 30 minutes at 37°C. The rate of formation of araAMP from 30.1 (250 gM) was 3.3 0.5 nmoles/ min/ nmole CYP3A4, whereas the rate of formation of PMEA from 28.4 (100 pM) was 3.1 0.1 nmoles/ min/ nmole CYP3A4.
These studies confirmed that p450 isoform CYP3A4 catalyzed the oxidation of 30.1 and 28.4.
Example H: Identification of Active Diastereomers P450 enzyme-catalyzed oxidation of the four isomers of 30.1 was evaluated in reactions containing microsomes from baculovirus-infecced insect cells co-expressing recombinant CYP3A4 and cytochrome p450 reductase (Panvera Corp., Madison, WI).
Methods: Reaction mixtures were similar to those described in Example E. Prodrug stock solutions were prepared in methanol and added to the reaction mixture to a final concentration of 100 pM. Reactions were terminated by addition of methanol to 60% evaporated, and redissolved in methanol. The 30.1 isomers were separated and quantified by HPLC with use of a YMC reverse phase CS column (250 x 4.6 mm) equilibrated with mobile phase A TFA) and eluted with a gradient to 80% mobile phase B (methanol) over 15 minutes. Isomers 1, 2,3, and 4 had retention times of 11.4, 12, 12.1, and 12.4 minutes, respectively.
Results: Reaction rates were linear over the 30 minute reaction period. Oxidation rates for each isomer are shown below (Isomer numbers refer to their elution order from the HPLC column): Isomer nmnoles oxidized/min/nmole CYP3A4 1 0.31 2 0.54 3 0.70 4 0.75 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 17:59 SPRUSON FERGUSON NO, 7148 P. 3/51 147 The results indicate that all four isomers of 30.1 were substrates for CYP3A4. Isomer I was the poorest substrate while isomer 4 was the best. This is in agreement with in vivo results described in Example N. Selection of single purified isomers may allow for optimization of pharmocokinetic and pharmnnacodynamic parameters.
Example 1: Activation in Isolated Rat Hepatocytes 7.1, 7.2, and 1.3 were evaluated for activation to parent compound in isolated rat hepatocytes.
Methods: Hepatocytes were prepared from fed Sprague-Dawley rats (250-300 g) according to the procedure of Berry and Friend (Berry, Friend, D.S. J. Cell Biol. 43. 506-520 (1969)) as modified by Groen (Groen, A.K. et al. Eur J. Biochm 122, 87-93 (1982)). Hepatocytes mg wet weight/ml) were incubated In 1 ml Krebs-bicarbonate buffer containing 10 mM glucose.
and I mg/ml BSA. Incubations were carried out in a 95% oxygen, 5% carbon dioxide atmosphere in closed, 50-ml Falcon tubes submerged in a rapidly shaking water bath (371C).
Prodrugs were dissolved in DMSO to yield 10 mM stock solutions, and then diluted into the cell suspension to yield a final concentration of 100 pM. At appropriate time points over the course of one hour, aliquots of the cell suspension were removed and spun through a silicon/mineral oil layer into 10% perchloric acid. The cell extracts in the acid layers were neutralized, and the intracellular prodrug metabolite content analyzed by reverse phase HPLC. An ODS column was used for analysis. It was eluted with a gradient from 10 mM sodium phosphate, pH 5.5 to acetonitrile. Detection was at 310 nm. Peaks on the chromatograms were identified by comparison to the retention times and spectra of standards of prodrug and parent compound.
Results: Both 7.1 and 7.2 generated peak intraheparocyte levels of 6-Chloro-l-isobutyl-2.(2- (approximately 700 and 500 nmoles/S cells, respectively) within 15 minutes of addition to the cell suspension. 1.3 generated similarly high levels of its parent compound, 6 -Amino- 9 -neopentyl--(2.phosphonofuranyl)purine. The data indicated that the prodrugs diffused rapidly into the cells and that they were readily metabolized to parent compound intracellularly.
Example J fanhibition of Glucose Production in Rat Hepatocytes Select prodrugs of FBPase inhibitors were tested for inhibition of glucose production in isolated rat hepatocytes.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:59 SPRUSON FERGUSON NO, 7148 P. 4/51 148 Methods: Hepatocytes were prepared from overnight fasted Sprague-Dawley rats 2 50-300 g) as described in Example I. Hepatocytes (75 mg wet weight/ml) were incubated in I ml Krebs-.
bicarbonate buffer containing I rng/ml BSA. Incubations were carried our in a 95% oxygen, carbon dioxide atmosphere in closed, 50-ml Falcon tubes submerged in a rapidly shaking water bath (37 After a 10 minute equilibration period, lactate and pyruvate were added to 10 mM and 1 mM concentrations, respectively. Addition of gluconeogenic substrates was immediately followed by that of appropriate concentrations of test compound. After I hour, an aliquot (0.25 ml) was removed, transferred to an Eppendorf tube and centrifuged. The supernatant (50 dl) was then assayed for glucose content using a Glucose Oxidase kit (Sigma Chemical Co.) as per the manufacturer's instructions.
Results: The following table depicts the inhibitory effect on gluconeogenesis for several prodrugs prepared in the Examples. Since none of the prodrugs were found to be inhibitots of FBPase, inhibition of gluconeogenesis in hepatocytes is a measure of the degree of activation of prodrug to the active parent compound. The results indicate that all of the prodrugs wcre converted to their respective parent compounds in hepatocytes.
Coam und inhibition 100 tM ICS0. uM 1.1 77 1.3 13 1,4 36 7.1 48 2.4 4.2 68 5.2 36 5.2 16 7.2 4.3 6.1 11 7.1 65 48 7-3 61 7.4 13 Example K: Biologically Active Drug Metabolites Formed Following Prodrug Activation In Isolated Rat Hepatocytes Metabolism of 28.4 and 30.1 to active antiviral nucleoside phosphates was monitored in isolated rat hepatocytes.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 17:59 SPRUSON FERGUSON NO. 7148 P. 5/51 149 Methods: Hepatocytes were prepared and incubated as described in Example I. 28.4 and 30.1 and their respective parent compounds, PMEA and ara-AMP, were dissolved in DMSO or methanol to yield 10 mM stock solutions, and then diluted into the cell suspension to yield a final concentration of 100 pM. At appropriate time points over the course of 2-4 hours, aliquots of the cell suspension were removed and spun through a silicon/mineral oil layer into 10% perchloric acid, The cell extracts in the acid layers were neutralized by the addition of 0.3 volumes of 3M KOH/3M KHCO 3 and the extract was then centrithged, filtered (0.2 micron filter) and loaded onto an anion exchange HPLC column equilibrated with 70% A (10 mM ammonium phosphate pH 3.5,6% ETOH) and 30% B (1 M ammonium phosphate pH 3.5, 6% ETOH). Ara-ATP and PMEApp were eluted with a gradient to 80% B. Detection was at 254 nm.
Results: The Table below clearly demonstrates that both 28.4 and 30,1 were readily converted to PMEA-diphosphate and ara-ATP in hepatocytes.
The lower levels of ara-ATP generated with 30.1 relative to ara-AMP arc most likely due to the slow rate of activation of the prodrug to parent compound in cells. In vivo, 30.1 was demonstrated to have a clear advantage over ara-AMP as evidenced by the 2- to 5-fold higher levels of ara-ATP generated in liver (Example 0).
Product concentration Compound Dose. uM Product Time Point nnmlesg.sClls 28.4 100 PMEApp 4h 169 PMEA 250 PMEApp 4h 120 30.1 100 ara-ATP 2h 108*4 araAMP 100 ara-ATP 2h 212*0.6 Example L: Identification of Prodrug Cleavage Products 1.1 was evaluated for microsome-catalyzed conversion to parent compound 2- Phenylethyl)-8-adeninyl]} I furanylphosphonic acid and phenol, an expected byproduct of the reaction.
Methods: Rat liver microsomes were prepared as described in Example I Reaction conditions, sample processing, and HPLC analysis were as described in Example Phenol was identified by HPLC by its retention time and spectral characteristics relative to an authentic standard.
COMS ID No: SMI8-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:00 2Q. EB. ~O4 8:00 SPRUSON FERGUSON N.74 .65 NO-7148 P. 6/51 Rens: 2-{(5-[9-(2-Phcnylethyl)-B-adcninyl]) fiuranylphosphonic acid =nd phenol were generated in a linear fatshion over a 5 hour reaction period at a rate of 15 pmolcs 1mg rnkrrosomal protein/rninutc. 2- {S-[9-(2-Phenylediyl)-S-adeninyl} furmnylphosphonic acid and phenol were liberated in an equimolar fashion as expected. These results support an oxidadiva-linination mechanism.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:00 SPRUSON FERGUSON NO. 7148 P. 7/51" 151 Example M: Oral Bloavailability The oral bioavailability (OBAV) of 30.1 was estimated by comparison of the area under the curve of ara-ATP generated in liver following oral administration to that generated following intravenous administration. The oral bioavailability of prodrugs of a variety of phosphonic acid FBPase inhibitors was estimated by a urinary excretion method in rat.
Methods: Fasted rats were dosed orally and intravenously with 30.1 at 3 and 10 mg/kg. Liver samples were obtained, processed, and analyzed for ara-ATP content as described in Example P.
Prodrugs of FBPase inhibitors were dissolved in 10% ethanol/90% polyethylene glycol (mw 400) and administered by oral gavage at doses of 20 or 40 mg/kg parent compound equivalents to 6-hour fasted, Sprague Dawley rats (220-240 The rats were subsequently placed in metabolic cages and urine was collected for 24 hours. The quantity of parent compound excreted into urine was determined by HPLC analysis. An ODS column eluted with a gradient from potassium phosphate buffer, pH 5.5 to acetonitrile was employed for these measurements.
Detection was at 310-325 rnm. The percentage oral bioavailability was estimated by comparison of the recovery in urine of the parent compound generated from the prodrug, to that recovered in urine 24 hours after intravenous administration of usubstituted parent compound at approximately 10 mg/kg. Parent compounds were typically dissolved in dimethyl sulfoxide, and administered via the tail vein in animals that were briefly anesthetized with halothane.
Results: The oral bioavailability of 30.1 was found to be 17.4% and 13.7% by comparison of the 10 mg/kg and 3 mg/kg oral and intravenous doses, respectively, These results suggest that 30.1 may be administered orally for the treatment of viral disease. AraA, the parent compound, has only been useflil as a parenteral treatment because of its low oral bioavailability. ["Adenine Arabinoside: An Antiviral Agent" (1975) D. Pavan-Langston, Editor, Raven Press Publishers, New York].
Results for the FBPase inhibitor prodrugs were as follows Prodrug Parent comnound OBAV Parent OBAYV Prodnru 2.4 6-Chloro- l-isobutyl-2-(2- 0.5 8.7 4.2 6-Chloro-l-isobutyl-2-(2- 0.5 5.2 6-Chloro-l-isobutyl-2-(2- 0.5 2.4 7.2 6-Chloro- 1-isobutyl-2-(2- Phosphono-5-furanyl)benzimidazolc 0.5 12.5 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:00 SPRUSON FERGUSON NO, 7148 P. 8/51 152 4.3 6-Chloro-1-isoburyl-2-(2.
0.5 11.6 7.1 6-Chloro- 1-isobutyl-2-(2- 0.5 10.9 7.3 6-Chloro- 1-isobutyl-2-(2.
0.5 14.1 As is evident from the above, the prodrugs increased the oral bioavailability of 6-Chloro-lisohutyl-2-(2-Phosphono-5-furanyl)benzimnidazole by 2.5- to Example N: Pharmacoldneties of30.1 and Certain Drug Metabolites The pharmacokinctics of 30.1 were evaluated in the rat.
Methods: 30.1 was administered at 20 mg/kg to fasted rats via the tail vein under light halothane anesthesia. At apropriate time points following drug administration, rats were reanesthetized with halothanc. The peritonecal cavity was then opened and a blood sample was obtained from the abdominal vena cava and the liver freeze-clamped and excised. Blood samples were briefly centrifuged and the plasma fraction was then extracted with 1.5 volumes of methanol, processed, and analyzed by HPLC as described in Example A. Liver samples were excised, frozed in liquid nitrogen, and then homogenized in 100% methanol. Liver extracts were then centrifuged at 14,000 rpm and the methanolic supernaants subsequently filtered and evaporated to dryness. Samples were resuspended in isotonic saline and analyzed by HPLC as described in Example A. Phannacokinetic parameters were determined with the aid of WinNonLin version 1.1 (Scientific Consulting, Inc.) software. Renal excretions studies were performed as described in Example Q.
Results: The following pharmocokinetic parameters were calculated for 30.1: clearance 3.34 L/hr/kg plasma half life 0.19 hr volume of distribution 0.70 L/kg mean residence time 0.21 hr liver half life 0.90 hr urinary (reral) excretion (24hr) (57.7 5.9%) The primary route of clearance was renal However, the short plasma half life and low volume of distribution of 30.1, coupled with high concentrations ofprodrug observed in liver 20 nmoles/g at 20 minutes), suggest that 30.1 was rapidly extracted from plasma by the liver as well. Hepatic metabolism of 30.1 to ara-ATP and/or other products could potentially account for clearance of the remaining 42% of the dose.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:01 SPRUSON FERGUSON NO. 7148 P, 9/51 153 The results also indicated that the four isomers of 30.1 underwent hepatic metabolism at different rates. The isomers were separated according to Example H, Isomer 1, for instance, wa cleared in liver significantly slower than isomer 3. The hepatic half-lives for isomers I and 3 were 0.98 and 0.54 hours, respectively.
Example 0: Enhanced Liver Delivery of ara-ATP and Reduced Generation of Ara-hypoxanthine The temporal profile of ara-hypoxanthine and ara-ATP generation following intravenous administration of 30.1 and free ara-AMP was compared in the rat.
Method: 30.1 and ara-AMP (Sigma) were administered to normal, fasted rats intravenously (in saline) via tail vein catheters. At appropriate time points following drug a administration, animals were lightly anesthetized with halothane. The peritoneal cavity was then opened and a blood sample was obtained from the abdominal vena cava and the liver freeze-clamped and excised.
The blood samples were hcparinized and plasma was prepared. Plasma samples (100 ItL) were mixed with 4 N perchloric acid (40 gL), vortexed, and then clarified by centrifugation.
The supematants were analyzed for ara-hypoxanthine by HPLC with use of a Beckman Ultrasphere ODS column (4.6 x 150 mm). The column was eluted with 100 mM acetic acid at a flow rate of 1.0 mrL/min. The eluent was monitored by UIV absorbance at 254 runm.
Livers were immediately homogenized in 3 volumes of 10% perchloric acid and the extracts clarified by centrifugation at 2500 x g (5 minutes). The resulting supernatants were removed and neutralized with 0.3 volumes of 3M KOH/ 3M KHC03. The neutralized liver extracts were then spun in an Eppendorfmicroftge at 10,000 rpm (20 minutes, The supernatnts were analyzed for ara-ATP content by reverse phase HPLC (-IP 1050) using a Whatman Partisphere SA column (4.6 x 125 mm). A gradient from 0,3M ammoniumwn phosphate buffer pH 3.5 to 0.8M anmmonium phosphate pH 3.5 was run (1 tnl/minute flow rate).
Renal excretion of araH following i.v. administration of 30.1 or ara-AMP was determined as described in Example Q.
Results: Ara-hypoxanthine was not detected following administration of 30,1. Nor was arahypoxanthine detected in urine In contrast, ara-AMP at equivalent doses generated pmolar levels of ara-hypoxanthine in plasma, which persisted for 8 hours. In addition, 19.2% of the mg/kg equivalent ara-AMP dose was excreted in the form of ara-hypoxanthine in urine.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:01 SPRUSON FERGUSON NO. 7148 P. 10/51 154 Both 30.1 and ara-AMP readily generated ara-ATP in liver. Based on area under the curve, Ara-ATP levels generated in liver by 30.1 at doses of 10 and 3 mg/kg were 5.2 and 2.1 times higher over 8 hours, respectively, than those generated by a 10 mag/kS equivalent dose of ara- AMP. These studies indicate that 30.1 has an advantage over ara-AMP in vivo because higher levels of the active antiviral metabolite, ara-ATP, were generated in liver, whereas arahypoxanthine, a metabolite associated with the toxicity of ara-AMP, was not detected in plasma or urine.
Example P: Hepatic PMEA-diphosphate and Plasma PMEA Generation Following Intravenous Administration of 28.4 and bisPOM-PMEA to Rats The metabolism of 28.4 and bisPOM-PMEA to the active antiviral nucleaside dipbosphate, PMEApp, and parent compound, PMEA, were compared in rat.
Methods: 28.4 and bisPOM-PMEA were dissolved in DMSO/cthanol and administered intravenously (10 mg/kg, PMEA equivalents) to fasted rats via tail vein catheters under light halothane anesthesia. At appropriate time points, blood and liver samples were obtained and processed as described in Example 0. PMEA-diphosphate in liver and PMEA in plasma were analyzed by HPLC as described for araATP and ara-hypoxanthine, respectively, in Example 0.
Results: Both 28.4 and bisPOM-PMEA generated readily detectable levels of PMEAdiphosphate in liver. The area under the curve for hepatic PMEA-diphosphate was 3-fold greater for bisPOM-PMEA relative to 28.4. Plasma PMEA levels, however, were greater than lower with 28.4 relative to bisPOM-PMEA. The results indicate that peripheral exposure to PMEA can be reduced by using a cyclic prodrug targeted to the liver and, coupled with the findings described in Example Q, suggest that the therapeutic index for PMEA can be increased with 28.4.
Example Q: Renal Excretion of PMEA following Intravenous Administration of 28.4, bisPOM-PMEA, and PMEA in rats The renal excretion profiles of 24, bisPOM-PMEA, and PMEA were compared in rat.
Methods: 28.4, bisPOM-PMEA, and PMEA were dissolved in 56% DMSO/44% isotonic saline and administered intravenously to rats via the tail vein. Rats were subsequently caged in metabolic cages and urine collected over a 24 hour period. Urine samples were extracted with mechanol/ 2% acetic acid, vortexed and clarified by centrifugation. Supernatants were COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:01 SPRUSON FERGUSON NO.7148 P. 11/51 155 analyzed for PMEA content by reverse phase HPLC. A Beckman Ultrasphere ODS (4.6 x 150 mm) column was used with a gradient from 20 mM potassium phosphate pH 6.2 to acctonitrile.
Results: Renal excretion of PMEA and of PMEA generated from bisPOM-PMEA was 83% and 54%, respectively, of the administered dose. In contrast, less than 1% of the 28,4 dose was recovered as PMEA in the urine. This result indicates that the renal exposure and thus the renal toxicity associated with PMEA may be avoided by administration of the compound as the prodrug 28.4.
Example R: Preliminary Toxicological Evaluation of 30.1 In Mice The toxicity of 30.1 was evaluated in the mouse.
Methods: 30.1 and free Ara-AMP were administered intraperitoneally to normal C57 mice for 22 days at doses of 10 and 100 mg/kg Death and other obvious signs of toxicity such as weight loss, tremors, ruffling fur, hunching, prostration, diarrhea. lethargy, and hyperactivity were monitored. On day 22, 3 hours following.the final dosing, mice were sacrificed and livers were removed and homogenized in 3 volumes (to liver weight) of perchloric acid. Liver extracts were neutralized and araATP was quantitated by ion-exchange HPLC as in Example K.
Reauls: There was no mortality during the study nor were significant differences in weight gain observed for any of the drugtreated mice relative to saline treated controls. In addition, no abnormal behaviors were observed during the treatment period. AraATP levels measured in liver at the end of the treatment period are shown in the table below: Compound Dose. myf/k/d araATP, nmoles/e liver araAMP 20 2.5 0.2 30.1 20 6.6 1.6 araAMP 200 19.1 30.1 200 23.1 9 The results demonstrate that 30.1 can be safely administered to mice over a 22 day period at doses up to 200 mg/kg/d while generating liver ara-ATP levels that are equal to or higher than levels generated by the parent compound, ara-AMP.
Example 5: Sustained Drug Levels COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:02 SPRUSON FERGUSON NO.7148 P. 12/51 156 As described in Example 0, the plasma half-life of 30.1 in rat was 12 minutes, whereas that of free ara-AMP, as judged by our inability to detect the compound in plasma, was considerably shorter. The latter is in accordance with studies reported in woodchucks and in man. In woodchucks administered araAMP intravenously, plasma ara-AMP levels dropped by greater than 90% over 2.5 minutes, suggesting a half-life on the order of seconds [Ponzetto, A. et al.
(1991) Hcpatology 14: 16-24]. Following intravenous administration to human patients, ana- AMP was not detected in plasma, also indicating an extremely short half-life Whitley, RJ et al (1980) Antimicrobial Agents and Chemotherapy 18: 709-7151. 30.1, by virtue of its sustained plasma levels, can thus serve as a reservoir of parent compound and can therefore be useful in extending the pharmacoldkneic and pharmacodynamic half-life of the parent drug.
The pharmacodynamic half-life of parent compounds may also be extended by selecting prodrugs with a slow rate of activation. In the study described in Example o it was found that the four isomers of 30.1 were activated at different rates in liver. Isomer 1, for instance, had an intrahcpatic elimination rate half-life of 0.98h whereas isomer 3 had a half-life of 0.54 h. This suggests that the intrahepatic half-life of 30.1 and thus presumably that of the active antiviral nucleoside triphosphate, ara-ATP, is dependent on the drug diastereomer. For example, Isomer I would have a relatively long half-life, whereas isomer 3 would have a shorter half-life.
Example T: Tissue Distribution 30.1 is tested for activation in homogenates from various tissues to assess the specificity of liver activation.
Methods: Rats are anaesthetized under halothane and tissues including liver, kidney, brain, heart, stomach, spleen, muscle, lung, and testis are excised and frozen in liquid nitrogen. Tissues are then homogenized in 1 to 3 volumes of 50 mM Tris/HC1, 154 mM KCi, I mM EGTA pH 7.4.
The homogenate is centrifuged at 10,000 rpm and 40C for 30 minutes and the supematant recovered. Liver cytosol is prepared by centrifuging the liver crude extract for 1 hour at 40,000 rpm and 4"C. Reaction mixtures consist of 50 mM KH 2 P0 4 pH 7.4, 13 mM glucose-6phosphate, 2 mM NADP t 10 units of glucose-6-phosphate dehydrogenase, 100 AM 30.1 and tissue homogenate at a protein concentration of 8.5 mg/ml. Reactions are incubated at 37*C.
Aliquots are taken after 0 and 1 hour of incubation, and extracted with 60 methanol. The methanolic extracts are centrifuged at 14,000 rpm, and filtered prior to analysis by HPLC. The COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:02 SPRUSON FERGUSON NO. 7148 13/51 157 samples are loaded onto a YMC C8 column equilibrated with 0. 1% TFA and eluted with a methanol gradient to 80% The activation of 30.1 to ara-A.MP is monitored at 254 nM Results: Activation of 30.1 is expected in crude rat liver homogenate resulting in depletion of the prodrug and formation of parent compound, ara-AMP. Incubation of 30.1 with liver cytosol, which does not contain microsomes, does not result in activation. Incubation with all of the other tissue homogenates is not expected to result in activation. Results of this nature indicate liver specific activation of 30.1.
Example U: Delivery of 6-Amino-9-neopentyl-8-(2-phosphonofuranyl)purine to liver The generation of parent compound 6-Amino-9-neopentyl-8-(2-phosphonofuranyl)purine in liver following administration of prodrug 1.3 was evaluated in rat.
Methods: 1.3 was dissolved in DMSO and administered intraperitoncally to fasted rats at a dose of 50 mg/kg. At various time points following drug administration, animals were anesthetized with halothane. The peritoneal cavity was then opened and a blood sample was obtained from the abdominal vena cava and the liver freeze-clamped and excised. Blood and liver samples were processed as described in Example P and analyzed for 6-Amino-9-neopentyl- 8-(2-phosphonotfranyl)purine content as described in Example I.
Results' 6-Amino-9-neopentyl-8-(2-phosphonofuranyl)purine was detected both in plasma and liver following 1.3 administration. Peak levels measured at the earliest time point of 1 hour were 5.8 gM in plasma and 5 nmoles/g tissue in liver. 6-Amino.9-ncopentyl-8-(2phosphonofuranyl)purine persisted in plasma and liver for 4 hours (the last time point measured).
The data demonstrate that 1.3 is metabolized to 6-Amino-9-neopentyl-8-(2phosphonofuranyl)purine in vivo and that prodrug administration results in the delivery of 6- Amino-9-neopentyl-8-(2-phosphonofiranyl)purine to the liver.
COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20

Claims (238)

1. A method of enhancing oral bioavailability of a parent drug by administering to an animal a prodrug of formula I: V H w W wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy. and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, hetcroaryl, substituted heteroaryl, 1-alkenyl. l-alkynyl, and Z is selected from the group consisting of-CHROH -CHROC(0)R', -CHR2OC(S)R', -CHRzOC(S)OR', -CHR 2 OC(O)SR', -CHR 2 OCOzR, -OR 2 -SRk, -CHR'N,, -CHiaryl, -CH(aryl)OH. -CH(CH--CR2)OH, -CH(C-CR)OH, -NR -OCOR', -OCO0R, -SCOR., -SCO 2 R, -NHCOR, -NHCOzR', -CH 2 NHaryl, -(CH 2 ),-OR 2 and -(CH2)p-SRz; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:03 SPRUSON FERGUSON NO 7148 P. 15/51 159 R 2 is an R3 or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to POJ PzO6 3 or P 3 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
2. The method of claim I wherein M is attached to the phosphorus in formula I via an oxygen atom.
3. The method of claim 2 wherein M is a nucleoside.
4. The method o f claim 3 wherein said oxygen is in a primary hydroxyl group on a ribofuranosyl or an arabinofuranosyl group. The method of claim 3 wherein said .oxygen is in a primary hydroxy group. 6, The method of claim 5 wherein MH is selected from the group consisting of araA, AZT, d4T, ddl, ddA, ddC, L-ddC, L-FddC, L-d4C, L-Fd4C, 3TC, ribavirin, penciclovir, fluoro-2'-deoxyuridine, FIAU, FIAC, BHCG, yl]cytosine, (-)-b-L-2',3'-dideoxycytidine, (-)-b-L-2',3'-dideoxy-5-fluorocytidine, FMAU, BvaraU, E-5-(2-bromovinyl)-2'-deoxyuridine, Cobucavir. TFT, CDG, DAPD, FDOC, d4C, DXG, FEAU. FLG, FLT, FTC. 5-yl-carbocyclic 2'-deoxyguanosine, Cytallene, Oxetanocin A, Oxctanocin G, Cyclobut A, Cyclobut G, fluorodeoxyuridine, dFdC, araC, bromodeoxyuridinc, IDU, CdA, F-araA. 5-FdUMP, Coformycin, and 2'-dcoxycoformycin. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 18:03 SPRUSON FERGUSON NO. 7148 P. 16/51
7. sugar group. The method of claim 3 wherein said oxygen is in a hydroxyl group on an acyclic
8. The method of claim 7 wherein MH is selected from the group consisting of ACV, GCV, 9-(4-hydroxy-3-hydroxymethylbut-1-yl)guanine, and dihydroxybutyl)guanine.
9. The methodoaf claim 3 wherein M is a compound of formula II: J L wherein E is selected from the group consisting of alkyl, amino or halogen; L and J are independently selected from the group consisting of hydrogen, hydroxy, acyloxy, alkoxycarbonyloxy, or when taken together form a lower cyclic ring containing at least one oxygen; and A is selected from the group consisting of amino and lower alkylarnino; and pharmaceutically acceptable salts thereof. The method of claim I wherein M is attached to the phosphorus in formula I via a carbon atom.
11. The method of claim 10 wherein M-P03 2 is selected from the group consisting of phoapohonoformic acid, and plosphonoacetic acid.
12. The methods of claim 10 wherein M is a compound of formula III: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:03 SPRUSON FERGUSON NO, 7148 P. 17/51 161 A N N Y wherein A is selected from the group consisting of -NR 2, NHSOiR 3 halogen, lower alkyl, -CON(R4)2, guanidine, amidine, and perhaloalkyl; E is selected from the group consisting of halogen, lower alkylthio, lower perhaloalkyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, -CN, and -NR 7 2; X is selected from the group consisting of alkylamino, alkyl, alkenyl, alkynyl, alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonate), alkyl(sulfonatc), aryl, alkylaminoalkyl. alkoxyalkyl, alkylthioalkyl, alkylthio, alicyclic, 1,1-dihaloalkyl, carbonylalkyl, alkylaminocarbonyl, alkylcarbonylamino, aralkyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including cyclic alkyl, heterocyclic, and aryl; Y is selected from the group consisting of-H, alkyl, alkenyl, alkynyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl, -C(O)R 3 -S(0)2R -C(O)-OR 3 -CONHIR, -NR 2 2, and -OR 3 all except H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; R 4 is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; R is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic; R 6 is independently selected from the group consisting of and lower alkyl; R 7 is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, lower aryl, and -C(0)RI0 R' is independently selected from the group consisting of lower alkyl, lower aralkyl, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:04 SPRUSON FERGUSON NO.7148 P. 18/51 162 lower aryl, lower alicyclic, or together they form a bidentae alkyl; RP is selected from the group consisting of lower alkyl, -NH2, lower aryl, and lower perhaloalkyl; RI I is selected from the group consisting of alkyl, aryl, -OH, -NH2 and and pharmaceutically acceptable prodrugs and salts thereof.
13. The method of claim 10 wherein M is a compound of formula IV: A L N J Y wherein: A, E, and L are selected from the group consisting of-NR 8 2, -NO2, -OR 7 -SR -C(O)NR 4 2, halo, -COR 1 -SO2R', guanidine, amidine, -NHSOzR 5 -SO2NR 4 2, -CN, sulfoxide, perhaloacyl, perhaloalkyl, perhaloalkoxy, Cl-CS alkyl, C2-C5 alkenyt, alkynyl, and lower alicyclic, or together A and L form a cyclic group, or together L and E fomn a cyclic group, or together E and J form a cyclic group including aryl, cyclic alkyl, and heterocyclic; J is selected from the group Consisting of -NR 8 2, -NO2, -OR 7 -SR 7 -C(O)NR 4 z, halo. -C(O)R 1 1 -CN, sulfonyl, sulfoxide, perhaloalkyl, hydroxyalkyl, perhaloalkoxy, alkyl, haloalkyl, aminoalkyl, alkcnyl, alkynyl, alicyclic, aryl. and aralkyl, or together with Y forns a cyclic group including aryl, cyclic alkyl and heterocyclic alkyl; X is selected from the group consisting of alkylamino, alkyl(hydroxy), alkyl(carboxyl), alkyl(phosphonate), alkyl, alkenyl. alkynyl, alkyl(sulfonate), aryl, carbonylalkyl, 1,1- dihaloalkyl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthio, alkylarinocarbonyl, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:04 SPRUSON FERGUSON NO. 7148 19/51 163 alkylcarbonylarmino, alicyclic, aralkyl, and alkylaryl, all optionally substituted; or together with Y f6rms a cyclic group including aryl, cyclic alkyl, and heterocyclic; Y is selected from the group consisting of-H, alkyl, alkenyl, alkynyl. aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl, -S(0)2R, -CONHR 3 -NR 2 2. and -OR, all except -H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; R 4 is independently selected from the group consisting of lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; RK is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic; R6 is independently selected from the group consisting of-H, and lower alkyl; R 7 is independently selected from the group consisting of lower alkyl, lower alicyclic, lower aralkyl, lower aryl, and -C(0)R10 R* is independently selected from the group consisting of lower alkyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R 0 o. or together they form a bidentate alkyl; Ri 0 is selected from the group consisting of lower alkyl, -NH2, lower aryl, and lower perhaloalkyl; R 11 is selected from the group consisting of alkyl, aryl, -OH, -NH2 and -OR3; and pharmaceutically acceptable prodrugs and salts thereof; with the provisos that: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:04 SPRUSON FERGUSON NO. 7148 P. 20/51 164 a) when X is alkyl or alkenc, then A is -NR 2; b) K is not alkylamine and alkylaminoalkyl when an alkyl moiety is substituted with phosphonic esters and acids; and c) A, L, E, Y, and X together may only form 0-2 cyclic groups.
14. The method of claim 10 wherein MH is selected from the group consisting of PMEA, PMEDAP, HPMPC, HPMPA, FPMPA, and PMPA. The method of claim I wherein MPO 3 2 MPZO 3 or MPO, 4 is useful for the treatment of diseases of the liver or metabolic diseases where the liver is responsible for the overproduction of a biochemical end product.
16. The method of claim 15 wherein said disease of the liver is selected from the group consisting of hepatitis, cancer, fibrosis, malaria, gallstones, and chronic cholecystalithiasis. 18
17. The methods of claim 16 wherein MH MPO h MP 2 06 or MP 3 O 4 is an antiviral or anticancer agent
18. The method of claim 15 wherein said metabolic disease is selected from the group consisting of diabetes, atherosclerosis, and obesity.
19. The method of claim 15 wherein said biochemical end product is selected from the group consisting of glucose, cholesterol, fatty acids, and triglycerides.
20. The method of claim 19 wherein MH or MPO' is an AMP activated protein kinase activator.
21. The method of claim I wherein M -PO32' is a compound that inhibits human liver FBPase.
22. The method of claim 21 wherein said compound inhibits human liver FBPase with an IC5o of less than 10 pM. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:04 SPRUSON FERGUSON NO. 7148 P. 21/51 165
23. The method of claim 21 wherein M is a group T-X wherein T is selected from the group consisting ofbenzimidazole, indole, purine, and 9- azaindole, all of which contain at least one substituent; X is attached at the 2, 2, 8, and 2 positions of said T groups, respectively; and X is selected from the group consisting of alkylamino, alkyl, alkenyl, alkynyi. alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonate), alkyl(sulfonate), aryl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkylIalkylthio, alicyclic, 1,1-dihaloalkyl, carbonylalkyl, alkylaminocarbonyl, alkylcarbonylamino, aral.kyl, and alkylaryl, all optionally substituted.
24. The method of claim I wherein V is selected from the group consisting of aryl, substitued aryl, heteroaryl, substituted heteroaryl; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing one heteroatom. that is fused to an aryl group attached at the beta and gamma position to the oxygen attached to the phosphorus or together V and W are connected via 3 carbon atoms to form a cyclic group containing 6 carbon atoms mono-substituted with a substituent selected from the group consisting of hydroxyl, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy and aryloxycarbonyloxy three atoms from an oxygen attached to the phosphorus.
25. The method of claim 24 wherein V is selected from the group consisting of aryl, substicued aryl, heteroaryl, and substituted heteroaryl.
26. The method of claim 25 wherein V is selected from the group consisiting of aryl and substituted aryl.
27. The method of claim 26 wherein V is selected from the group consisting of phenyl, and substituted phenyl.
28. The method of claim 27 wherein V is 3-bromophenyl.
29. The method of claim 25 wherein V is selected from the group consisting of heteroaryl and substituted heteroaryl. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:05 SPRUSON FERGUSON NO. 7148 P. 22/51 166 The method of claim 29 wherein V is 4-pyridyl.
31. The method of claim 24 wherein together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta and gamma positions to the oxygen attached to phosphorus.
32- The method of claim 31 wherein said aryl group is an optionally substituted monocyclic aryl group and the connection between Z and the ortho position of the aryl group is selected from the group consisting of CH2, CH2CH, OCH 2 or CH7O. 33, The method of claim 24 wherein together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and mono- substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alikoxycarbonyloxy, alkylthiocarbonyloxy. and aryloxycarbonyloxy three atoms from an oxygen attached to the phosphorus.
34. The method of claim 33 wherein together V and W form a cyclic group selected from the group consisting of-CH 2 -CH(OH)-CH 2 -CHzCH(OCOR)-CH 2 and -CHICH(OCO 2 R)-CH-, The method of claim 1 wherein Z is selected from the group consisting of -CHROH -CHR 2 OCOR', and -CHR 2 OCO 2 R
36. The method of claim I wherein Z is selected from the group consisting of-ORt, -SR 2 -CHRN 3 -NRW, -OCOR', -OCO 2 -SCORW, -SCO 2 -NHCOR 2 -NHCO 2 R', -CH 2 NHaryl, -(CH2)p-OR 2 and -(CH 2 )p-SR 2
37. The method of claim 36 wherein Z is selected from the group consisting of-OR, -R 2 -OCOR, -OC0 2 -NHCOR 2 -NHCO7R 3 -(CH 2 ),-OR 2 and, -(CH 2 )p-SR 2 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:05 SPRUSON FERGUSON N0.7148 P, 23/51 167
38. The method of claim 37 wherein Z is selected from the group consisting of-OR, -H -OCOR. -OCO 2 and -NHCOR 2
39. The method of claim .wherein W and W' are independently selected from the group consisting of H, R3, aryl, substituted aryl, hetcroaryl, and substituted aryl. The method of claim 39 wherein W and W' are the same group.
41. The method of claim 40 wherein W and W' are H.
42. The method of claim I wherein said prodrug is a compound of formula VI: V M-P VI wherein V is selccted from the group consisting of aryl, substituted aryl, heteroaryl, and substituted aryl.
43. The method of claim 42 wherein M is atached to phosphorus via an oxygen or carbon atom.
44. The method of claim 42 wherein V is selected from the group consisting of phenyl and substituted phenyl. The method of claim 42 wherein V is pyridyl.
46. The method of claim I wherein said prodrug is a compound of formula VII: V H VII COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:05 SPRUSON FERGUSON NO. 7148 P. 24/51 168 wherein Z is selected from the group consisting of: -CHR 2 OH, -CHROCOR', -CHR 2 OC(S)R 3 -CHR 2 OCOzRa, -CHR'OC(O)SR, -CHR 2 OC(S)OR, -SR 2 and -CHzay[.
47. The method of claim 46 wherein M is attached to the phosphorus via a carbon or oxygen atom.
48. The method of claim 47 wherein Z is selected from the group consisting of -CHROH, -CHROC(O)R', and -CHRIOCOzR 3
49. The method of claim 48 wherein R 2 is -H.
50. The method of claim I wherein said prodrug is a compound of formula VIII: VIII wherein Z' is selected from the group consisting of-OH, -OC02 and -OC(O)S RK; 0' and D" are independently selected from the group consisting of-H, alkyl, -OH, and -OC(O)R 3 with the proviso that at least one of D' and D" are -H.
51. The method of claim I wherein W'.and Z are W and V are both the same aryl, substituted aryl, heteroaryl, or substituted heteroaryl such that the phosphonate prodrug moiety: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 18:05 SPRUSON FERGUSON NO. 7148 P. 25/51 1 169 V w has a plane of symmetry.
52. The method of claim 1 wherein W and W' are H, V is selected from the group consisting of aryl, substituted aryl, heteroaryl, substituted heteroaryl, and Z is selected from the sroup consisting of OR 2 and -NHCOR 2
53. The method of claim 52 wherein Z is -H.
54. The method of claim 53 wherein said oxygen is in a primary hydroxyl group. The method of claim 53 wherein V is selected from the group consisting of phenyl or substituted phenyl.
56. The method of claim 55 wherein said oxygen is in a primary hydroxyl group.
57. The method of claim 53 wherein V is an optionally substituted monocyclic heteroaryl containing at least one nitrogen atom.
58. The method of claim 57 wherein said oxygen is in a primary hydroxyl group.
59. The method of claim 57 wherein V is 4-pyridyl, The methods of claims 54, 56, or 58 wherein MH is selected from the group consisting of araA, AZT, d4T, ddl, ddA, ddC, L-ddC, L-FddC, L-d4C, L-Fd4C. 3TC, ribavirin. penciclovir, 5-fluoro-2'-deoxyuridine, FIAU, FIAC, BHCG, (-).b-L-2',3'-dideoxycytidine, (-)-b-L-2',3'-dideoxy- FMAU, BvaraU, E-5-(2-bromovinyl)-2'-dcoxyuridine, Cobucavir, TFT, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:06 SPRUSON FERGUSON NO. 7148 P. 26/51 propyDyl-1-arabinosyluracil, CDG. DAPD, FDOC. d4C. DXG, FEAU, FLG, FLT, FTC, carbocyclic 2'-deoxyguanosine, Cytallene, Oxetanocin A, Oxetanocin G, Cyclobut A, Cyclobut 0, fluorodeoxyuridine, dFdC, araC, bromodcoxyuridine, IDU, CdA. F-ara-A, coformycin, and 2'-deoxycoformycin.
61. The methods of claims 54, 56, or 58 wherein MH is selected from the group consisting of ACV, GCV, 9-(4-hydroxy-3-hydroxymethylbut- 1 -yl)guanine, and dihydroxybutyl)guanine. 4
62. The methods of claims 53, 55, OR 57 wherein M is attached to the phosphorus via a carbon atom.
63. The method of claim 62 wherein MPO^' is selected from the group consisting of phosphonoformic acid, and phosphonoacetic acid.
64. The method of claim 62 wherein MH is selected from the group consisting of PMEA, PMEDAP, HPMPC, HPMPA, FPMPA, and PMPA. The method of claim 60 wherein V is selected from the group consisting of phenyl and 4-pyridyl and MH is selected from the group consisting of ribavirin, AZT, penciclovir, araA, 5-fluoro-2'-dcoxyuridine, ddl, ddA. ddC, and F-araA.
66. The methods of claims 54, 56, or 58 wherein M is selected from the group consisiting of: 'A j L II wherein E is selected from the group consisting of alkyl, amino or halogen; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:06 SRSN&FRUO SPRUSON FERGUSON NO. 7148' P. 2 7/5 1 171 L and J are independently selected from the group consisting of hydrogen, hydroxy, ac~ylocy, alkoxycarbonyloxy, or when taken together formi a lower cyclic ring Containing at least- one oxygen;- and A is selected from the group consisting of amino and lower allcylarnino; uad pharmaceutically acceptable prodrugs and salts therof.
67. The methods Of claims 54, 56, or 58 wherein M is selected from. the group consisiring of: A wherein A is selected from te group consislting of -NR 8 2, NHSO 2 R', -OR 3 halogen, lower allcyl, -CON(Rh guanidine, ainidine, and perhaloalkCyl; E is selected from the group consisting of halogen, lower alkltilo, lower perbaloalkyl, lower alkyl, lower alkenyl. lower alkynyL, lower alkoxy, -CN, and -NR 7 2; X is selected from the group consisting of aikyiwnino, aikyl, alkcnyl, alkynyl, alkyl(carboxyl), allcyl(hydnoxy), alkyl(phosphonate), alkyi(sulfonate), ryl, alkylawinoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthia, alicyclic, 1,1-ihAloalkyl, carbonylalkyl, aminocarbonylamino, alkylarninocarbonyl, alkylcarbonylamino, arailcyl. and alkylaryl, all optionally substituted; or together wit Y form a cyclic group including cyclic akyl, heterocyclic, and asql; Y is selected frm the group consisting of alkyl, alkenyl, alkynyl. aryl. alicyclic, aralkyl, az-yloxyaikyl, alkoxysilcyl, .C(O)-ORW, -CONHR 3 -NR 2 2, and -OR3, all except H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic ailkyl, mnd heterocyclic; COMS ID No: SMSI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:06 SPRUSON FERGUSON NO.7148 P. 28/51 172 is independently selected from the group consisting of lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; R3 is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic; R is independently selected from the group consisting of-H, and lower alkyl; R 7 is independently selected from the group consisting of-H, lower alkyl. lower alicyclic, lower aralkyl, lower aryl, and -C(O)R I 0; R' is independently selected from the group consisting of lower alkyl, lower aralkyl. lower aryl, lower alicyclic, or together they form a bidentate alkyl; Rio is selected from the group consisting of-H, lower alkyl, -NH2. lower aryl, and lower perhaloalkyl; R! is selected from the group consisting of alkyl, aryl, -OH, -NH2 and and pharmaceutically acceptable prodrugs and salts thereof.
68. The methods of claims 54, 56, or 58 wherein M is a compound of formula IV: Jl X- IV wherein: A. E, and L are selected from the group consisting of -NR 8 2, -NO2, -OR 7 -SR 7 -C(O)NR 4 2, halo, -COR 11 -S02R', guanidine, amidine, -NHSO 2 -SO2NR 4 2, -CN, sulfoxidc, perhaloacyl, pcrbaloalkyl, perhaloalkoxy, Cl-C5 alkyl, alkenyl, C2-C5 alkynyl and lower alicyclic, or together A and L form a cyclic group, or together L and E form a cyclic group, or together E and J form a cyclic group including aryl, cyclic alkyl, and heterocyclic; J is selected from the group consisting of -NR 2, -N02, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:06 SPRUSON FERGUSON NO, 7148 P. 29/51 173 -OR 7 -SR 7 -C(O)NR42, halo, -C(O)R 1 1 -CN, sulfonyl, sulfoxide, perhaloalkyl. hydroxyalkyl, perhaloalkoxy, alkyl, haloalkyl, aminoalkyl, alkenyl, alkynyl, alicyclic, aryl, and aralkyl, or together with Y forms a cyclic group including aryl, cyclic alkyl and hererocyclic alkyl; X is selected from the group consisting of alkylamino, alkyl(hydroxy), alkyl(carboxyl), alkyl(phosphonate), alkyl, alkenyl, alkynyl, alkyl(sulfonare), aryl, carbonylalkyl, 1,1- dihaloalkyl, aminocarbonylamino, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthio, alkylaminocarbonyl, alkylcarbonylamino, alicyclic, aralkyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; Y is selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl, -S(0)2R. -C(0)-OR 3 -CONHR -NR22, and all except -H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; R 4 is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; R 5 is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic; R is independently selected from the group consisting of and lower alkyl; R is independently selected from the group consisting of lower alkyl, lower alicyclic, lower aralkyl, lower aryl, and -C(0)R R' is independently selected from the group consisting of-H, lower alkyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R t or together they form a bidentate alkyl; Rio is selected from the group consisting of-H, lower alkyl, -NH2, lower aryl, and lower perhatoalkyl; R I l is selected from the group consisting of alkyl, aryl, -OH, -NH2 and and pharmaceutically acceptable prodrugs and salts thereof; with the provisos that: a) when X is alkyl or alkene, then A is -NR 2: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:07 SPRUSON FERGUSON NO. 7148 P. 30/51 174 b) X is not alkylamine and alkylaminoalkyl when an alkyl moiety is substituted with phosphonic esters and acids; and c) A, L, E, 1, Y, and X together may only form 0-2 cyclic groups.
69. The method of claim I wherein said oral bioavailability is at least The method of claim 69 wherein said oral bioavailability is at least
71. The method of claim 69 wherein said oral bioavailability is enhanced by compared to the parent drug administered orally.
72. The method of claim 71 wherein said oral bioavailability is enhanced by at least 100/%.
73. A method of delivering a biologically active drug to an animal for a sustained period usin-compounds of formula 1: wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted beteroaryl, 1-alkenyl, I-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:07 SPRUSON FERGUSON NO. 7148 P. 31/51 175 together V and Z arc connected via 3-5 atoms to form a cyclic group, optionally containing I hteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl. substituted aryl, hetcroaryl, substituted heteroaryl. 1-alkenyl, 1-alkynyl, and Z is selected from the group consisting of-CHRzOH, -CHR 2 OC(O)R', -CHR 2 OC(S)R', -CHR'OC(S)OR, -CHR 2 OC(O)SR', -CHR 2 OCO 2 -SR', -CHR'N3, -CHaryl, -CH(azyl)OH, -CH(CH-CR% 2 )OH, -CH(C-CR')OH, -NR 2 -OCOR', -OCOzR, -SCOR', -SCOzR'. -NHCOR 2 -NHCOZR'. -CH2NHaryl, -(CH 2 ),-OR 2 and -(CH2),-SR; R 1 is an R 3 or -H; R is selected from the group consisting of alkyl, ary), aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to PO, P, 20, or P09 4 is biologically active in vivo, and that islattached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
74. The method of claim 73 whereby therapeutic levels of said drug are maintained for at least one hour longer than the levels achieved by oral administration of the bispivaloyloxymethyl.(bis-POM) ester. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:07 SPRUSON FERGUSON NO. 7148 P. 32/51 176 The method of claim 73 whereby therapeutic levels of said FBPase inhibitors are maintained for at least one hour longer after systemic administration relative to an equivalent molar amount of the parent compound administered by the same route.
76. The method of claim 73 wherein MP0 3 z is an FBPase inhibitor.
77. The method of claim 73 wherein MH or MPO3- is an antiviral or anticancer agent.
78. The method of claim 77 wherein MH is selected from the group consisting of araA, AZT, d4T, 3TC, l-P-D-ribofuranosyl-l,2,4-triazole-3-carboxamide, PMEA, ACV, 9-(4- hydroxy-3.hydroxymethylbut-i-yl)guanine, 5-yl-carbocyclio 2'-deoxyguanosine, dFdC, araC, F- ara-A, and CdA-
79. A method of delivering a biologically active drug to an animal with greater selectivity for the liver using compounds of formula I: V oH 0 M-P Z W' W I wherein: V is selected from the group consisting of-H. alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkcnyl, 1-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:08 SPRUSON FERGUSON NO. 7148 P. 33/51 177 toSether V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroacom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substiment selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted hcteroaryl, 1-alkenyl, 1-alkynyl, and Z is selected from the group consisting of -CHR 2 OH, -CHR 2 OC(O)R 3 -CHROC(S)R, -CHR 2 OC(S)OR', -CHROC(O)SR, -CHWROCOR, -SR 2 -CHRN 3 -CH2aryl, -CH(aryl)OH, -CH(CH=CR'2)OH, -CH(C-CR 2 )OH, -R -NR22. -OCOR 3 -OCO 2 -SCOR, -SCO 2 R 2 -NHCOR 2 -NHCOI 2 R, -CHNHaryl, -(CH2),-OR 2 and -(CHz)p'SR R is an R' or -H; tR is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and Rt is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is then at least one of V and W is not or and M is selected from the group that attached to PO 3 P 2 0 3 or P30 4" is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof. The method of claim 79 whereby the ratio of a parent drug or a drug metabolite concentration in the liver over a parent drug or a drug metabolite concentration in the plasma is two times greater compared to administration of a parent drug. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:08 SPRUSON FERGUSON NO. 7148 P. 34/51 178
81. The method of claim 80 wherein the liver specificity has qMAtt Ma 'latfviU administration of M-PO03=.
82. The method of claim 79 wherein said biologically active drug is a triphosphate generated in the liver.
83. The method of claim 82 wherein MH is selected from the group consisting of araA, AZT, d4T, 3TC, l-0-D-ribofuranosyl-1,2,4-triazole-3-carboxamide, ACV, 9-(4-hydroxy- 3-hydroxymethylbut- -yl)guanine, 5-yl-carbocyclic 2'-deoxyguanosine, dFdC, araC, F-ara-A, and CdA.
84. The method of claim 79 wherein the active drug is MPO32'. The method of claim 84 wherein the drug is selected from the group consisting of FdUMP, 5-Bromo-( 1 D-ribotiranosyl)imidazole-4-carboxamide-5'*monophosphate, 6-Amino- 9-ncopentyl-8-(2-phosphonofuranyl)purine, 6-Chloro-I-isobutyl-2-(2-phosphono-5- furanyl)benzimidazole, and 4-Amino-7-echyl-5-fluoro-l-isobutyl-2-(2-phosphono-5- nfranyl)benzimidazole.
86. The method of claim 79 wherein the active drug is MPO, and M is attached via C.
87. The method of claim 86 wherein the drug is selected from the group consisting of PMEA, PMEADAP, HPMPS, HPMPA, FPMPA, PMPA, phosphonoformic acid, and phosphonoacetic acid.
88. A method of increasing the therapeutic index of a drug by administering to an animal compounds of formula I: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:08 SPRUSON FERGUSON NO. 7148 P. 35/51 179 I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted hateroaryl, I -alkenyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or L together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is ftsed to an.aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbony[oxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, beteroaryl, substituted heteroaryl, 1-alkenyl, I -alkynyl, and Z is selected from the group consisting of-CHRZOH -CHROC(O)R., -CHR'OC(S)R, -CHR 2 OC(S)OR, -CHROC(O)SR, -CHRMOCO 2 -SR 2 -CHR 2 -CH 2 aryl, -CH(aryl)OH, -CHCHHCR22)OH, -CH(C=CR2)OH, -R 2 -NR 2 -OCOR', -OCO2R, -SCOR -SCO 2 -NHCOR 2 -NHCO 2 -CH 2 NHaryl, -(CH2),-OR 2 and -(CH2),p-SR'; R is an R 3 or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is then at least one of V and W is not or -R9; and COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:08 SPRUSON FERGUSON NO. 7148 P. 36/51 180 M is selected from the group that attached to PO P20<, or PO(94- is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
89. The method of claim 88 wherein extrahepatic toxicity is reduced. The methqd of claim 89 wherein M-PO) 2 is excreted by the kidney.
91. The method of claim 89 wherein the M is selected from the group consisting of PMEA, PMEADAP, HPMPS, HPMPA, FPMPA, and PMPA.
92. The method of claim 89 wherein the gastrointestinal toxicity is reduced.
93. The method of claim 89 wherein central or peripheral nervous system toxicity is reduced.
94. A method of bypassing drug resistance by administering to an animal compounds V 00 H ~H w of formula I: I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, I -alkynyl, and -R 9 or together V and Z arc connected via 3-5 atoms to form a cyclic group, optionally containing I heteroarom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:09 SPRUSON FERGUSON NO. 7148 P. 37/51 181 together V and Z are connected via 3-5 atoms to form a cyclic group, optionhily containing I heferoatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form on optionally substituted cyclic group containing 6 carbon atoms and substituted with one substinrent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl. substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 Z is selected from the group consisting of-CHR 2 OH -CHROC(O)R', -CHR'OC(S)R', -CHR'OC(S)OR', -CHR 2 OC(O)SR', -CHR'OCOaR', -OR 2 -SR 2 -CHR -CHiaryl. -CH(aryl)OH, -CH(CH-CR 2 2 )OH, -CH(CuCRZ)OH, -NR'2, -OCOR', .OCO 2 R, -SCOR 3 -SCOR 3 -NHCOR 2 -NHICO 2 -CH 2 NHaryl, -(CH 2 9 -OR 2 and -(CH2),-SR 2 R1 is an R3 or -H; R3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R' is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that; a) V, Z, W. and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R9; and 3 M is selected from the group that attached to P0 3 PzO6, or P3O," is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof. The method of claim 94 wherein said resistance arises from decreased cellular production of M-POa".
96. The method of claim 94 wherein said compound is an anticancer or antiviral agent. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:09 SPRUSON FERGUSON NO. 7148 P. 38/51 182
97. The method of claim 96 wherein M is 5-fluoro-2'-dcoxvuridine and said agent Is (5-fluoro-2'-deoxyuridine), or
98. The method of claim 96 wherein said resistance is to an antiviral agent selected from the group consisting ofaraA, AZT, d4T, 3TC, 1-b-D-ribofihranosyl.l1,2,4-triazole-3- carboxamide, ACV, 9 4 -hydroxy-3-hydroxymethylbut-1-yl)guanine, and 5-yl-carbocyclic 2'- dcoxyguanosine. X
99. The method of claim 96 wherein the resistance or lack of antihepatiis activity is due to a deficiency in thymidine kinase and said antiviral agent is selected from the group consisting of AZT, d4T, ACV.
100. The method of claim 96 wherein said anticancer agent is selected from the group consisting of dFdC, araC, F-araA, and CdA.
101. A method of treating cancer by administering to an animal a compound of formula 1: O H M-P z wherein V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, hetroaryl, substituted heteroaryl, i-alkenyl, I-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to formnn a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:09 SPRUSON FERGUSON NO. 7148 P. 39/51 183 together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the bera and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of allcyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and Z is selected from the group consisting of-CHR 2 OH, -CHROC(O)R', -CHR 2 OC(S)R, -CHR'OC(S)O R -CHR'OC(0)SR', -CHR 2 OCO 2 -OR 2 -SR,. -CHR Ns, -CH2aryl, -CH(aryl)OH, -CH(CH=CR',)OH, -CH(C-CR2)OH, -R 2 -NR. 2 -OCOR;, -OCO, 2 R, -SCOR', -SCO2R', -NHCOR'. -NHCO2R', -CHNHaryl, -(CH2),-OR 2 and -(CHz),-SR 2 R is an R or -H: R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is then at least one of V and W is not or -R9; and M is selected from the group that attached to P02-, P 2 0 6 or P30s' is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
102. The method of claim 101 wherein the active drug is the triphosphate of M-H.
103. The method of claim 101 wherein the active drug is the monophosphate of M-H. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:10 SPRUSON FERGUSON NO. 7148 P. 40/51 184
104. The method of claim 101 wherein said prodrug is administered to patients resistant to the parent drug. 105, The method of claim 101 wherein MH is selected from the group consisting of dFdC, araC, F-araA. and CdA.
106. A method of treating viral infections by administering to an animal a compound of formula I: V 0/0 H M-P Z W I wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, I-alkenyl, l alkynyl, and or together V and Z are connected via 3-5 atoms to fprm a cyclic group, optionally containing 1 heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy. alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:10 SPRUSON FERGUSON NO. 7148 P. 41/51 185 W and W' are independently selected from the group consisting of-H, alkyl, aralkyl. alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, I-alkynyl. and -RI; Z is selected from the group consisting of-CHR 2 OH, -CHR'OC(0)R', -CHR'OC(S)R', -CHR'OC(S)OR 3 -CHR 2 OC(O)SR'. -CHR'OC 2 -OR 2 -SR 2 -CHR 2 -CH 2 aryl, -CH(aryl)OH, -CH(CH-CR' 2 )OH, -CH(CuCR')OH, -R .NR 2, -OCOR 3 -OC0 2 -SCORW, -SCO 2 R 3 -NHCOR 2 -NHCOzR', -CHzNHaryl, -(CH)-OR 2 and -(CHa),-SR 2 R 2 is an R 3 or -H; R3 is selected from the group consisting of alkyl, aryl. aralkyl, and alicyclic; and R9 is selected from the group consisting of alkyl. aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that; a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to P0 3 P20 6 or PO,9 4 is biologically active In vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
107. The method of claim 106 wherein said viral infection is hepatitis.
108. The method of claim 107 wherein MH is selected from the group consisting of AZT, d4T, and ACV.
109. The method of claim 107 wherein said prodrug is administered to patients resistant to the parent drug.
110. The method of claim 107 wherein said hepatitis is hepatitis B.
111. The methods of claim 106 wherein viral kinases produce M-P03'. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:10 SPRUSON FERGUSON NO. 7148 P. 42/51 186
112. The method of claim 111 wherein said viral infection is hepatitis and said viral kinases are kinases from viruses other than the hepatitis viruses.
113. The method of claim 106 wherein the active drug is the triphosphate of M-H.
114. A method of treating liver fibrosis by administering to an animal a compound of formula I: V H W W I wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1 -alkcnyl 1 -alkcynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally conaining I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonytoxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl. aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heeroaryl, 1-alkcnyl, 1-alkynyl, and -R 9 Z is selected from the group consisting of-CHROH -CHROC(O)R 3 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:10 SPRUSON FERGUSON NO. 7148 P. 43/51 187 -CHR'OC(S)R -CHR:OC(S)0R 3 -CHR'OCO)$SR', -CHR 2 OCOzR'. -SR -CHRZNi, -CHzaryl, -CH(aryl)OH, -CH(CH=CR 2 z)OH, -CH(C=CR 2 )OH, -R 2 -NR 2 2 -OCOR'. -OCOI2 3 -SCOR, -SCO 2 -NHCOR 2 -NHCO 2 R, -CHNHaryl, and R' is an R' or -H; R is selected from the group consisting of alkyl. aryi, aralky), and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos tat' a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and 1- 3 M is selected from the group that attached to PO, P206 or P30 9 4 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
115. A method of treating hyperlipidemia by administering to an animal a compound of formula I: V M w w I wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, hetcroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:11 SPRUSON FERGUSON NO. 7148 P. 44/51 188 together V and Z are connected via 3.5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy. and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl], heteroaryl, substituted heteroaryl, I -alkanyl, 1-alkynyl. and Z is selected from the group consisting of -CHR 2 OH, -CHR 2 OC(0)R', -CHR'2C(S)R', -CHR20C(S)OR', -CHR'OC(O)SR', -CHR'OCO 2 -OR 2 -SRI, -CHR' 2 N, -CH 2 aryl, -CH(aryl)OH, -CH(CH=CR'2)OH, -CH(C=CR 2 )OH, -R 2 -NR 2 -OCOR 3 -OC0 2 -SCOR', -SCOR', -NHCOR 2 -NHCO 2 -CH 2 NHaryl, -(CH2)p-OR, and -(CHz)-SR; R 2 is an R or-H; R' is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V. Z, W, and W' arc not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to PO, P 2 aO 6 or P309*' is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
116. The method of claim 115 wherein the hyperlipidernia agent is a squalene synthase inhibitor. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:11 SPRUSON FERGUSON NO.7148 P. 45/51 189
117. The method of claim 116 wherein said squalene synthase inhibitor is selected from the group consisting of 1-hydroxy-3-(methylpentylamino)-propylidene-1,1-bisphosphonic acid, BMS 187745 and analogs thereof.
118. A method of treating parasitic infections by administering to an animals compound of formula I: V H W W I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl. substituted aryl, heteroaryl, substituted beteroaryl, I-alkenyl, 1-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroacom, that is fused to-an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are conncted via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consising of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 Z is selected from the group consisting of -CHR 2 OH, -CHRZOC(O)R3, -CHROC(S)R, -CHR 2 OC(S)OR', -CHR 2 OC(O)SR, -CHROCO 2 zR, -OR 2 -SR 2 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:11 SPRUSON FERGUSON NO. 7148 P. 46/51 190 -CHR 2 N3, -CHiaryl, -CH(aryl)OH, -CH(CH=CR%2)OH, -CH(CwCR)OH, -R 2 -NR 2 -OCOR', -OCO z R 3 -SCOR', -SCO z R, -NHCOR', -NHCOR 3 -CH2NHaryl, -(CHz)p-OR 2 ad R 2 is an R 3 or -H; R 1 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisosthat: a) V, Z. W, and W' are not all and b) when Z is -R 2 then at least one ofV and W is not or -R9; and M is selected from the group that attached to PO, P 2 0 6 1 or P30' is biologically active in viva, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and phurmaceutically acceptable prodrugs and salts thereof.
119. A method of delivering diagnostic imaging agents to the liver comprising administration to an animal of compound of formula I: V H M- Z W' W I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I hoteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20,FEB-2004 18;11 SPRUSON FERGUSON NO.7148 P. 47/51 191 together V and Z are connected via 3-5 axoms to form a cyclic group, optionally containing I hetcroatorn, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and Z is selected from the group consisting of-CHR'OH, -CHR'OC(O)R, -CHROC(S)R', -CHR 2 OC(S)OR', -CHR 2 OC(0)SR', -CHR'OCO 2 R 3 -SR 2 -CHR 2 N 3 -CH 2 aryl, -CH(aryl)OH, -CH(CH-CR'z)OH. -CH(CmCR')OH, -NR 2 2 -OCOR', -OCO 2 R 3 -SCOR 3 -SCO 2 R, -NHCOR 2 -NHCOzR', -CH 2 NHayl, -(CH)p-ORR 2 and -(CH)p-SR; R2 is an R7 or -H; R is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to PO 3 PO"0 6 3 or P309'- is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
120. A method of making a prodrug of a compound drug having a -P02 moiety comprising, a) transforming said phosph(on)ate into a compound of formula I: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:12 SPRUSON FERGUSON NO. 7148 P. 48/51 192 V H 0 W W w w I wherein. V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl. 1-alkenyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substimrent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 Z is selected from the group consisting of-CHR 2 OH, -CHR 2 OC(O)R 3 -CHR 2 OC(S)R, -CHR 2 0C(S)OR 3 -CHRzOC(O)SR', -CHRZOCOzR 3 -OR 2 -SR. -CHR 2 N 3 -CH 2 aryl, -CH(aryl)OH, .CH(CH-CR' 2 )OH, .CH(C-CR 2 )OH, -NR -OCOR', -OCOIR', -SCOR3, -SCO 2 -NHCOR 2 -NHCO 2 R, -CHzNHaryl, -(CH2)-Op, and -(CHz),-SR 2 R' is an R or-H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl. aralkyl, and alicyclic; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 18:12 SPRUSON FERGUSON NO. 7148 P. 49/51 193 p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to PO P206, or P0 9 4 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, H, oxygen, atom; and pharmaceutically acceptable prodrugs and salts thereof. i
121. The method of claim 120 further comprising, a) converting M-PO32" to a compound M-P(0)L" 2 wherein L" is a leaving group selected from the group consisting of halogen; and b) reacting M-P(0)L" 2 with HO-CH(V)CH(Z)CH(Z)-CW(W')-OH.
122. The method of claim 121 wherein HO-CH(V)CH(Z)-CW(W')-OH is chiral.
123. The method of claim 122 further comprising isolating a single diasiereomer.
124. The method of claim 120 wherein a) converting a hydroxyl or amino to a phosph(oramid)ite by reaction with L-P(- OCH(V)CH(Z)-CW(W')O-) wherein L selected from the group consisting of NR' 2 and halogen; b) transforming said phosph(oramid)ite into a compound of formula I by reaction with an oxidizing agent.
125. The method of claim 124 wherein L-P(-OCH(V)CH(Z)-CW(W')O-) is chiral.
126. The method of claim 125 wherein the chiral phosph(oramid)ite is generated using a chiral diol.
127. The method of claim 124 wherein said oxidizing agent produces a mayor stereoisomer at the phosphorus in a ratio of at least 3:1. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 18:12 SPRUSON FERGUSON NO. 7148 P. 50/51 194
128. The method of making a prodrug of formula I comprising converting a hydroxyl or an amino to a phosphate or phosphoramidate, respectively, by reaction with OCH(V)CH(Z)-CW(W')O-) wherein L' is a leaving group selected from the group consisting of -NR2, aryloxy, and halogen.
129. The method of claim 128 wherein is a single stereoisomer. 4t
130. The method of claim 129 wherein said stereoisomer is generated using a chiral diol.
131. A compound, R'sN-P-(-OCH(V)CH(Z)-CW(W')O-) wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy. acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W arc connected via 3 carbon atoms to form a cyclic group containing 6 carbon atoms substituted with hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, hereroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and Z is selected from the group consisting of-.CHR'OH, -CHROC(O)R', -CHR OC(S)R', -CHR 2 OC(S)OR 3 -CHR 2 OC(O)SR. -CHR 2 OCO2R1, -OR 2 -SR, -CHR 2 NR 2 2 -CH 2 aryl, -CH(aryl)OH, -CH(CH=CR 2 2 )OH, -CH(CmCR 2 )OH, .NR2, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:13 SPRUSON FERGUSON N0.7148 P, 51/51 195 -OCOR, -OCO 2 R 3 -SCOR 3 -SCO 2 R 3 -NHCOR, -NHCO 2 R 3 -CH2NHaryl, .(CH 2 )p.-OR and -(CH 2 )pSR:; R 2 is an R or -H; R' is selected from the group consisting of alkyl, aryl, aralkyl, and alcyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not alkyl, or and each R i is independently selected from the group consisting ofalkyl, aryl, and aralkyl or together R' and R' form a cyclic group, optionally containing a hereroatom; with the proviso that R' is not methyl. 132, A compound R'2N-P(O)(-OCH(V)CH(Z).CW(W')O-) wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkcnyl, 1-alkynyl, and -R 9 or together V and Z arc connected via 3-5 atoms to form a cyclic group, optionally containing I heteroarom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form a cyclic group containing 6 carbon atoms substituted with hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-atkynyl, and -R 9 Z is selected from the group consisting of -CHR 2 OH -CHR 2 OC(O)R 3 -CHR 2 OC(S)R -CHR'OC(S)OR 3 -CHR 2 OC(O)SR 3 -CHROCO 2 R 3 -OR 2 -SR', COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:13 SPRUSON FERGUSON NO. 7149 P. 2/45 196 -CHR'NR2,. -CH 2 ary], -CH(aryl)OH, -CH(CH=CR-2)OH, -CH(CaCR 2 )OH, -R -NR 2 -OCOR', -OCO 2 -SCOR 3 -SCO 2 R3, -NHCOR', -NHCO2R, -CHzNHaryl, -(CH 2 ),-OR 2 anid -(CH2)p-SR 2 R 2 is an R' or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V. Z, W, and W' are not all and b) when Z is -R z then at least one of V and W is not alkyl, or and each R' is independently selected from the group consisting of alkyl, aryl, and aralkyl; or together R' and R' form a cyclic group, optionally containing a heteroatom; with the proviso that both R' groups are not benzyl or ethyl at the same time.
133. A method of delivering a compound to heparocytes wherein said compound has a moiety selected from the group consisting of phosph(on)ate comprising: a) converting said compound to a prodrug of formula I: V o0 oH M-P Z W W I. wherein: V is selected from the group consisting of-H, alkyl, aralky), alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to fbrmnn a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxyoarbonyloxy. or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:13 SPRUSON FERGUSON NO, 7149 P. 3/45 197 together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heceroaryl, I -alkenyl, 1-alkynyl, and .R 9 Z is selected from the group consisting of -CHR 2 OH, -CHR 2 OC(O)R', -CHR 2 OC(S)WR, -CiROC(s)OR, -CHR 2 OC(O)SR', -CHR 2 OCOiR 3 -OR 2 -SR 2 -CHR 2 -CHzaryl, -CH(aryl)OH, -CH(CH-CR2)OH, -CH(CmCR)OH, -NR 2 -OOROCOR', -COCO 2 RR -SCORR, -5COR 3 -NHCOR', -NHCO z -CH2NHaryl, -(CH2)-OR, and .(CHz),-SR; R' is an R 3 or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R is selected from the group consisting of alkyl, aralkyl, and alicycli; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or and M is selected from the group that attached to POJ 2 P 2 0 6 3 or Pj09' is biologically active m vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaccutically acceptable prodrugs and salts thereof COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:14 SPRUSON FERGUSON NO. 7149 P, 4/45 198
134. A compound of formula I: V 0 H W w I wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, hcteroaryl. substituted heteroaryl, 1-alkenyl, 1-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclip group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy anached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substitucnt selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl. hetcroaryl, substituted heteroaryl, alkenyl, 1-alkynyl, and Z is selected from the group consisting of-CHROH, -CHR 2 OC(O)R', -CHR'OC(S)R', -CHR 2 OC(S)0R 3 -CHROC(O)SR', -CHR'OCO 2 -SR, -CHR'N), -CH2aryl, -CH(aryl)OH, -CH(CH=CRZ 2 )OH, -CH(CMCR')OH, -R 2 -NR 2 -OCOR 3 -OCO 2 R 3 -SCOR', -SCOzR', -NHCOR', -NHCOzR'. -CH 2 NHaryl. -(CH 2 )p-ORt, and -(CH2),-SR 2 R? is an R 3 or R) is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 18:14 SPRUSON FERGUSON NO.7149 P. 5/45 199 R' is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that; a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or and M is selected from the group that attached to POI P 2 0 6 or P309 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom with the proviso that M-PO3 2 is not an FBPase inhibitor; and pharmaceutically acceptable prodrugs and salts thereof. 135, The compounds of claim 134 wherein MPO 3 MH MP 2 0 6 and MPFO 9 4 is selected from the group consisting of an antiviral, anticancer, antihyperlipidemic, and antiparasitio agents.
136. The compound of claim 134 wherein MPO 2 MH, MP 2 0Q 3 and MPO4- is selected from the group consisting of metalloprotease inhibitor, and TS inhibitor.
137. The compounds of claim 135 wherein MH is selected from the group consisting of araA, AZT, d4T, ddl, ddA, ddC, L-ddC, L-FddC, L-d4C, L-Fd4C, 3TC, ribavirin, penciclovir, 5-fluoro-2'-deoxyuridine, FIAU, FIAC, BHCG, 2'R,5'S(-)-l-[2-(hydroxymethyl)oxathiolan.$- yl]cytosine, (-)-b.L.2',3'-dideoxycytidine, (-)-b-L-2',3'-didcoxy-5-fluorocytidine, FMAU. BvaraU, E-5-(2-bromovinyl)-2'-deoxyuridine, Cobucavir, TFT, CDG, DAPD. FDOC. d4C, DXG, FEAU, FLG, FLT, FTC, 5-yl-carbocyclic 2'-deoxyguanosine, Cytallene, Oxetanocin A, Oxetanocin G, Cyclobut A, Cyctobut G, fluorodeoxyuridinc, dFdC, araC, bromodeoxyuridine, IDU, CdA, F-araA, 5-FdUMP, Coformycin, and 2'-deoxycoformycin.
138. The compounds of claim 135 wherein MH is selected from the group consisting of ACV, GCV, 9-(4-hydroxy-3-hydroxymcthylbut-l-yl)guanine and dihydroxyburyl)guanine.
139. The compounds of claim 134 wherein M is selected from the group consisting of PMEA, PMEDAP, HPMPC. HPMPA, FPMPA, and PMPA. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:14 SPRUSON FERGUSON NO. 7149 P. 6/45 200
140. The compounds of claim 134 wherein M.PO z 2 is selected from the group consisting of phospohonoformnic acid, and phosphonoacetic acid.
141. A method of enhancing the pharmacodynamic half-life of a parent drug by administering to an animal a prodrug of formula I: V O_ H A /H w W I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substitned aryl, heteroaryl, substituted heteroaryl, 1-alkcnyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 beteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one subsituent selected from the group consisting of hydroxy, acyloxy, atkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 Z is selected from the group consisting of-CHRIOH -CHR 2 OC(O)R', -CHR 2 OC(S)R', -CHR 2 OC(S)OR 3 .CHROC(O)SR 3 -CHR 2 OCO 2 R 3 -OR z -SR, IT (AULS COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:15 SPRUSON FERGUSON NO. 7149 P. 7/45 201 -CHRa Nj, -CHzaryl, -CH(aryl)OH, -CH(CH=CR 2 )OH, -CH(CCR)OH, -R 2 -NR2,. -OCORt. -OCOCR3, -SCOR, -SCO 2 R 3 -NHCOR -NHCO 2 -CH2NHaRyl, -(CHZ)p-OR", amd -(CHz)p-SR'; R 2 is an R or -H; R is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that; a) V. Z, W, and W' are not all and b) when Z is then at least one of V and W is not or and M is selected from the group that attached to P 2 0z 3 or P 3 09, is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
142. A prodrug for enhancing the pharmacodynamic half-life of a parent drug comprising a prodrug of formula I: VH M-P Z w' W I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and or together V and Z arc connected via 3-5 atoms to form a cyclic group, optionally containing 1 hctcroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:15 SPRUSON FERGUSON NO,. 7149 P. 8/45 202 together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted 6 cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted hctcroaryl, I -alkenyl, 1 -alkynyl, and Z is selected from the group consisting of-CHR 2 OH, -CHR'OC(O)R 3 -CHR'OC(S)R!, -CHR'OC(S)OR 3 -CHROC(O)SR', -CHR 2 OCO 2 -OR 2 -SR', -CHR 2 Nj, -CHnaryl, -CH(aryl)OH, -CH(CH-CR- 2 )OH, -CH(C-CR 2 )OH. -R 2 NR 2 2 -OCOR', -OCO2R', -SCOR3, -SCOzR', -NHCOR 2 -NHCO 2 R 3 -CH2NHaryl, -(CH2)p-OR 2 and -(CHz),R 2 R z is an R3 or-H; R is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting ofalkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is then at least one of V and W is not or -R 9 and M is selected from the group that attached to F 2 PzO 3 or P,30' is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceuically acceptable prodrugs and salts thereof.
143. The prodrug of claim 141 wherein M is attached to the phosphorus in formula I via an oxygen atom.
144. The prodrug of claim 143 wherein M is a nucleoside. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20.FEB-2004 18:15 20. EB. 004 8:15 SPRUSON FERGUSON N.74 .94 NO. 7149 P. 9/45-' 203
145. The prodrug of claim 144 wherein said oxygen is in a primiary bydraxyl group on a ribofuraosy! or an arabinofliranosyl group.
146. The prodrug of claim 144 wherein said oxygen is in a primary hydroxy group.
147. The prodrug of claim 146 wherein MH is selected fromt the group consisting of artA, AZT, d4T, ddl, ddA, ddC, L-ddC, L-PddC, L.-d4C, L-Fd4C, 3TC, ribavirin, peaciclovir, fluoro-2'-deoxywidine, FIAU. PLACE BHCO, I-[ 2 -(hydroxcymerby1)oxatholan-s- yl]cytosine, .3 -dideoxycytidine, '-dideoxy-5-fluorocyidine,*FA4U, EvaretT, E-5-(2-bromovixiyl)-2'-deoxywidine, Cobucavir, TFT, 5-propynyl-1 -aratinosyluracil, MDG, DAPD, FDOC, d4C, DXC, FBATJ, PLO, FLiT, FTC, S-yl-carbocyvlic, 2'-deoxyguaosine, Cyrallene, Oxetanocin A, Oxetmnocin 0, Cyclobut A, Cyclobut 0, fluorodeoxyuzridine, dIFdC, mrnC, bromodeoxyuridine, IDIJ, CdA, F-IaA, 5-FdTJMP, Coformycin, aimd 2-deoxycoformycin.
148. The prodrug of claim, 144 wherein said oxygen is in a hydroxyl group on an acycl ic sugar group.
149. The prodrug of claim 148 wherein IM is selected from the group consisting of ACV, GCV. 9-( 4 -hydroxy-3-hydroxynethylbut-.1-yflguanne, and dihydroxybwtyl)guanine.
150. The prodrug of claim 144 wherein M is a compound of formula M; EA J L wherein E is selected from the group consisting of alkyl, amino or halogen; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:; 15 SPRUSON FERGUSON N0. 7149 P. 10/45 204 L and J are independently selected from the group consisting of hydrogen, hydroxy, acyloxy. alkoxycarbonylaxy, or when taken together form a lower cyclic ring containing at leat one oxygen; and A is selected from the group consisting ofanuino and lower alkylamino; and pharmaceutically acceptable salts thereof
151. The prodrug of claim 141 wherein M is attached to the phosphorus in formula I via a carbon atom.
152. The prodrug of claim 151 wherein M-PO?) 2 is selected from the group consisting of phospohonoformic acid, and phosphonoacetic acid.
153. The prodrug of claim 151 wherein M is a compound of formula III: A N N -X\ N N Y wherein A is selected from the group consisting of-NR82, NHSO 2 halogen, lower alkyl, -CON(R4), guanidine, amidinc, and perhaloalkyl; E is selected from the group consisting of-H, halogen, lower alkylthio, lower perhaloalkyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, -CN, and -NR 7 2; X is selected from the group consisting of alkylamino, alkyl, alkenyl, alkynyl, alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonate), alkyl(sulfonate), aryl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthio, alicyclic, 1,1-dihaloalkyl, carbonylalkyl, alkylaminocarbonyl, alkylcarbonylamino, aralcyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including cyclic alkyl, heterocyclic, and aryl; Y is selected from the group consisting of alkyl, alkenyl, alkynyl, aryl, alicyclic, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:16 SPRUSON FERGUSON NO. 7149 P. 11/45 205 aralkyl, aryloxyalkyl, alkoxyalkyl, -CONHR3, -NR22, and -OR J all except H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; R is independently selected from the group consisting of lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; Rs is selected from the group consisting of lower alkyl, lower aryl. lower aralkyl, and lower alicyclic; is t R is independently selected from the group consisting of-H, and lower alkyl; R is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, lower aryl, and -C(0)RI; R 6 is independently selected from the group consisting of-H, lower alkyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R 0 o, or together they form a bidentate alkyl; R 10 is selected from the group consisting of lower alkyl, -NH2, lower aryl, and lower perhaloalkyl; R 11 is selected from the group consisting of alkyl, aryl, -OH, *NH2 and -OR 3 and pharmaceutically acceptable prodrugs and salts therof.
154. The prodrug of claim 151 wherein M is a compound of formula IV: A L N J Y wherein: A, E, and L are selected from the group consisting of *NR 2, -N02, -OR 7 -SR 7 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 20-FEB-2004 18:16 SPRUSON FERGUSON NO. 7149 P, 12/45" 206 -C(O)NR42, halo, -COR I 1 4O02R 3 guanidine, amidine, -NHSO 2 R 5 -SO2NR2, -CN, sulfoxide, perhaloacyl, perhaloalkyl, perhaloalkoxy, CI-C5 alkyl, C2-C5 alkenyl, alkynyl, and lower alicyclic, or together A and L form a cyclic group, or together L and E form a cyclic group, or together E and J form a cyclic group including ary), cyclic alkyl, and heterocyclic; J is -selected from the group consisting of -NR 8 2, -NO 2 -OR 7 -SR 7 -C(Q)NR42. halo, -C(O)R I I -CN, sulfonyl, sulfoxide, perhaloalkyl, hydroxyalkyl, perhaloalkoxy, alkyl, haloalkyl, aminoalkyl, alkenyl, alkynyl, alicyclic, aryl, and aralkyl, or together with Y forms a cyclic group including aryl, cyclic alkyl and heterocyclic alkyl; X is selected from the group consisting ofalkylamino, alkyl(hydroxy), alkyl(carboxyl), alkyl(phosphonate), alkyl, alkenyl, alkynyl, alkyl(sulfonate), aryl, carbonylalkyl, 1,1- dihaloalkyl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl, alkylthio, alkylaminocarbonyl, alkylcarbonylamino, alicyclic, aralkyl, and alkylaryl, all optionally substituted; or together with Y forms a cyclic group including aryli cyclic alkyl, and heterocyclic; Y is selected from the group consisting of alkyl, alkenyl, alkyfiyl, aryl, alicyclic, aralkyl, aryloxyalkyl, alkoxyalkyl, -S(0)2R 3 -C(O).OR, -CON,RW, -NR22, and -OR 3 all except -H arc optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyl, and heterocyclic; R4 is independently selected from the group consisting of lower alkyl, lower alicyclic. lower aralkyl, and lower aryl; Rj is selected from the group consisting of lower alkyl, lower aryl. lower aralkyl, and lower alicyclic; R 6 is independently selected from the group consisting of-H, and lower alkyl; R is independently selected from the group consisting of-H, lower alkyl, lower alicyclic. lower aralkyl, lower aryl, and -C(O)R 10 R is independently selected from the group consisting of-H, lower alkcyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R 0 o, or together they form a bidentate alkyl; R I0 is selected from the group consisting of lower alkyl, -NH2, lower aryl, and lower perhaloalkyl; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 18:16 SPRUSON FERGUSON N0.7149 13/45- 207 R is selected from the group consisting of alkyl, aryl, -OH, -NH2 and and pharmaceutically acceptable prodrugs and salts thereof; with the provisos that: a) when X is alkyl or alkene, then A is -NR82; b) X is not alkylamine and alkylaminoalkyl when an alkyl moiety is substituted with phosphonic esters and acids; and c) A, L, E, J, Y, and X together may only form 0-2 cyclic groups.
155. The prodrug of claim 151 wherein MH is selected from the group consisting of PMEA. PMEDAP, HPMPC, HPMPA, FPMPA, and PMPA.
156. The prodrug of claim 141 wherein MPO MP 2 0 6 or MPjOs 4 is useful for the treatment of diseases of the liver or metabolic diseases where the liver is responsible for the overproduction of a biochemical end product.
157. The prodrug of claim 156 wherein said disease of the liver is selected from the group consisting of hepatitis, cancer, fibrosis, malaria, gallsiones, and chronic cholccystalithiasis.
158. The prodrug of claim 157 wherein MH, MPOA MP 2 0s 3- or MP O' is an antiviral or anticancer agent.
159. The prodru g of claim 156 wherein said metabolic disease is selected from the group consisting of diabetes, atherosclerosis, and obesity.
160. The prodrug of claim 156 wherein said biochemical end product is selected from the group consisting of glucose, cholesterol, fatty acids, and triglycerides.
161. The prodrug of claim 160 wherein MH or MPO3 2 is an AMP activated protein kinase activator. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:17 SPRUSON FERGUSON NO. 7149 P. 14/45- 208
162. The prodrug of claim 141 wherein M -PO,3 is a compound that inhibits human liver FBPase.
163. The prodrug of claim 162 wherein said compound inhibits human liver FBPase with an ICso of Jess than 10 pM.
164. The prodnig of claim 162 wherein M is a group T-X wherein T is selececd from the group consisting of benzimidazole, indole, purine, and 9- azaindole, all of which contain at least one subsitucnt; X is attached at the 2, 2, 8, and 2 positions of said T groups, respectively; and X is selected from the group consisting of alkylamino, alkyl, alkenyl, alkynyl, alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonate), alkyl(sulfonate), aryl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyl. alkylthio, alicyclic, 1,1 -dihaloalkyl, carbonylalkyl, alkylamninocarbonyl, alkylcarbonylamino, aralkyl, and alkylaryl, all optionally substituted.
165. The prodrug of claim 141 wherein V is selected from the group consisting of aryl, substimed aryl, beteroaryl, substituted heteroaryl; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing one heteroatom, that is fused to an aryl group attached at the beta and gamma position to the oxygen attached to the phosphorus or together V and W are connected via 3 carbon atoms to form a cyclic group containing 6 carbon atoms mono-substituted with a substituemn selected from the group consisting of hydroxyl, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy and aryloxycarbonyloxy three atoms from an oxygen attached to the phosphorus.
166. The prodrug of claim 165 wherein V is selected from the group consisting of aryl, substitued aryl, heteroaryl, and substituted heteroaryl.
167. The prodrug of claim 166 wherein V is selected from the group consisiting of aryl and substituted aryl.
168. The prodrug of claim 167 wherein V is selected from the group consisting of phenyl, and substituted phenyl. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:17 SPRUSON FERGUSON NO. 7149 P. 15/45- 209
169. The prodrug of claim 168 wherein V is 3 -bromophenyl.
170. The prodrug of claiin 166 wherein V is selected from the group consisting of heteroaryl and substituted heteroaryl.
171. The prodrug of claim 170 wherein V is 4-pyridyl.
172. The prodrug of claim 165 wherein together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma positions to the oxygen attached to phosphorus.
173. The prodrug of claim 172 wherein said aryl group is an optionally substituted monocyclic aryl group and the connection between Z and the gamma position of the aryl group is selected from the group consisting of O0, CH2, CHzCH 2 OCH 2 or CH,0.
174. The prodrug of claim 165 wherein together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and mono- substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkyltiocarbonyloxy, and aryloxycarbonyloxy three atoms from an oxygen attached to the phosphorus.
175. The prodrug of claim 174 wherein todgether V and W form a cyclic group selected from the group consisting of-CH 2 -CH2CH(OCOR)-CH,.-, and -CH2CH(OCO 2 R)-CHz-.
176. The prodrug of claim 141 wherein Z is selected from the group consisting of -CHR 2 OH, -CHR 2 OCOR', and.-CHR'OCO2R 3
177. The prodrug of claim 141 wherein Z is selected from the group consisting of -OR, -SR 2 -CHRNa, -NR 2 -OCORC, -OCO 2 R, -SCOR, -SCOzR 3 -NHCOR 2 -NHCO2R', -CHNHaryl. -(CH 2 )p-OR 2 and .(CH 2 ),-SR 2 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB, 2004 18:17 SPRUSON FERGUSON NO. 7149-P. 16/45- 210
178. The prdrug of claim 177 wherein Z is selected from the group consisting ot- OR R -OCOR 2 -OCOR 3 -NHCOR 2 -NHCOzR, -(CH2)p-OR2, and, .(CH2)o-SR'. 179 The prodrug of clairn 178 wherein Z is selected from the group consisting of OR 2 -OCOR 2 -OCOR 3 and -NHCOR.
180. The prodrug of claim 141 wherein W and W' are independently selected from the group consisting of H, R, aryl, substituted aryl, heteroaryl, and substituted aryl.
181. The prodrug of claim 180 wherein W and W' are the same group.
182. The prodrug of claim 181 wherein W and W' are H.
183. The prodrug of claim 141 wherein said prodrug is a compound of formula VI; V 0 0 wherein V is selected from the group consisting ofaryl, substituted aryl, heteroaryl, and substituted aryl.
184. The prodrug of claim 183 wherein M is attached to phosphorus via an oxygen or carbon atom.
185. The prodrug of claim 183 wherein V is selected from the group consisting of phenyl and substituted phenyl.
186. The prodrug of claim 183 wherein V is pyridyl. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:17 SPRUSON FERGUSON NO. 7149 P. 17/45 211
187. The prodrug of claim 141 wherein said prodrug is a compound of formula VII: 0 0 M P 0 VII wherein Z is selected fromy the group consisting of: -CHROH, -CHROCOR, -CHR 2 OC(S)R 3 -CHR 2 OCOC2R, -CHR'OC(0)SR' -CHRIOC(S)OR 3 and -CHaryl.
188. The prodrug of claim 187 wherein M is attached to the phosphorus via a carbon or oxygen atom.
189. The prodrug of claim 188 wherein Z is selected from the group consisting of -CHR'OH, -CHRzOC(O)R' and -CHR 2 OCOR
190. The prodrug of claim 189 wherein R 2 is -H.
191. The prodrug of claim 141 wherein said prodrug is a compound of formula VIII: M P--P- wherein Z' is selected from the group consisting of-OK, -OCO 2 R, and -OC(O)S R 3 D' and D" are independently selected from the group consisting of alkyl, -OH, and *OC(O)R COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:18 SPRUSON FERGUSON NO. 7149 P. 18/45 212
192. The prodrug of claim 141 wherein W' and Z arc W and V are both the sae aryl, substituted aryl, heteroaryl, or substituted hereroaryl such that the phosphonate prodrug moiety: V W has a plane of symmetry.
193. The prodrug of claim 141 wherein W and W' are H, V is selected from the group consisting of aryl, substituted aryl, heteroaryl, substituted heteroaryl, and Z is selected from the group consisting of OR 3 and -NHCOR'
194. The prodrug of claim 193 wherein Z is -H.
195. The prodrug of claim 194 wherein said oxygen is in a primary hydroxyl group.
196. The prodrug of claim 194 wherein V is selected from the group consisting of phenyl or substituted phenyl.
197. The prodrug of claim 196 wherein said oxygen is in a primary hydroxyl group.
198. The prodrug of claim 194 wherein V is an optionally substituted monocyclic heteroaryl containing at least one nitrogen atom.
199. The prodrug of claim 198 wherein said oxygen is in a primary hydroxyl group.
200. The prodrug of claim 198 wherein V is 4-pyridyl, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:18 SPRUSON FERGUSON NO. /149 P. 19/45' 213
201. The prodrug s of claims 195, 197, or 199 wherein M I isselected from the group consisting of ara.A. AZT, d4T, ddl. ddA, ddC. L-ddC. L-FddC, L-d4C. L-Fd4C, JTC, ribavirin,' peiciovir, S-iluoro.2'-deoxyuhidine, FIAU. FIAC, B1{CO, 2'R 1 0 y 05 jine '-dideoxycytidine, '-dideoxy- 5-tluorocytidine, FMAU, BvaraLJ,.5-(2 bromoviny)2 '-deoxyuridjne, Cobucavy, TFT, propynyl-1-arabinosyluradzl CDG, DAPD, EDOC, d4C, DXG, FE-AT, FLG, FLT, FTC, carbocyclic 2 '-deoxyguanosine, Cytallane, Oxezanocin A, Oxeranocin Gi, Cyclobut A, Cyciobut G, fluorcdeoxyuridine, dFdC. araC, bromodeoxcyudidine. IDU, C±4, F-ama-A, S-FdUMP, coformycin, and 2 '-deoxycoforrycin.
202. The prodrugs of claims 195, 197, or 199 whercin MH is selected from the group consisting of ACV, GCV, 9-4hdoy--yrxmeh u--yi)guanine, and dihydroxybucyi)guanine.
203. The prodnigs of claims 195, 197, or 199 wherein M is attached to the phosphorus via a carbon atom.
204. The prodrugs of claims 195, 197, or 199 wherein MP0 3 2 is selected from the group consisting of phosphonaogmjc acid, and phosphonoacetic acid.
205. The prodrugs of claims 195, 197, or 199 wherein ME is selected from the group cons isting of PMEA, PMEDAP, FtPMPC, NPMPA, FPMAI and PMIPA.
206. The prodrug of claim 201 wherein V is selected from- the group consisting of phenyl and 4 -pyridyl and MH is selected from the group consisting of ribavirin, AZT, penciclovir, artA, S-fluoro-2 '-dccx yuridine, dcii, ddA, ddC, and E-araA.
207. The prodnags of claims 195, 197, or 199 wherein Mv is selected from the group consisiting of: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-2O FEB. 2004 18:18 SPRUSON FERGUSON NO. 7149 20/45 214 0 E J L II wherein E is selected from the group consisting of alkyl, amino or halogen; L and J are independently selected from the group consisting of hydrogen, hydroxy, acyloxy, alkoxycarbonyloxy. or when taken together form a lower cyclic ring containing at least one oxygen; and A is selected from the group consisting of amino and lower alkylamino; and pharmaceutically acceptable prodrugs and salts thereof.
208. The prodrugs of claims 195, 197, or 199 wherein M is selected from the group consisiing of: A 1 Y III wherein A is selected from the group consisting of NR 8 2, NHSOR 3 -OR 5 halogen, lower alkyl. -CON(R 4 guanidine, amidine, and perhaloalkyl; E is selected from the group consisting of-H, halogen, lower alkylthio, lower perhaloalkyl, lower alkyl, lower alkenyl, lower alkynyl, lower alkoxy, -CN, and -NR72; X is selected from the group consisting of alkylamino, alkyl, alkenyl, alkynyl, alkyl(carboxyl), alkyl(hydroxy), alkyl(phosphonate), alkyl(sulfonate), aryl, alkylaminoalkyl, alkoxyalkyl, alkylthioalkyL alkylthio, alicyclic, 1.1-dihaloalkyl, carbonylalkyl, aminocarbonylamino, alkylaminocarbonyl, alkylcarbonylamino, aralkyl, and alkylaryl, all COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:18 SPRUSON FERGUSON NO. 7149- P. 21/45-- 215 optionally substituted;.or together with Y forms a cyclic group including cyclic alkyl, heterocyclic, and aryl; Y is selected from the group consisting of-H, alkyl, alkenyl, aikynyl., aryl, alicyclic, aralkyl. aryloxyalkyl, alkoxyalkyl, -C(O)R 3 S(0)2R, -C(O}-OR 3 -CONHR -NR 2 2, and -OR 3 all except H are optionally substituted; or together with X forms a cyclic group including aryl, cyclic alkyi, and hetcrocyclic; R 4 is independently selected from the group consisting of-H, lower alkyl, lower alicyclic, lower aralkyl, and lower aryl; R' is selected from the group consisting of lower alkyl, lower aryl, lower aralkyl, and lower alicyclic; R 6 is independently selected from the group consisting of-H, and lower alkyl; R 7 is independently selected from the group consisting of lower alkyl, lower alicyclic, lower aralkyl, lower ary], and -C(O)R 10 R' is independently selected from the group consisting of-H, lower alkyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R' Q or together they form a bidentate alkyl; R 1 0 is selected from the group consisting of lower alkyl, -NH2, lower aryl, and lower perhaloalkyl; R 1 is selected from the group consisting of alkyl, aryl, -OH, -NH2 and and pharmaceutically acceptable prodrugs and salts thereof.
209. The prodrugs of claims 195, 197, or 199 wherein M is a compound of formula IV: A j Y IV wherein: A. E, and L are selected from the group consisting of COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:19 SPRUSON FERGUSON NO. 7149- P. 22/45- 2 16 -NR 8 2 SR 7 _C(O)NRt2. halo, -CORI 1 S02R', guanidine. amnidine, -NHSO 2 R 5 'SO2NR42, -CN, sulfoxidc, perhaloacyl, perhaloalkyl, perhaloalkoxy, Cl-C5 alkyl, C2-CS allcenyl. C2-CS alkynyl, and lower alicyclic, or Together A and L form a cyclic group, or together L and B form a cyclic group, or together E and J form a cyclic group including aryl, cyclic alkyl, and heterocyclic; J is selected from the group consisting of -NR 8 2, -NO 2 -OR. 7 -SR 7 ,I -C(O)NR 4 2 halo, -C(O)Pk 11 -CN, sulfonyl, sulfoxide, perhalosilcyl. hydroxyalkyl, perhaloalicoxy, alkyl, haloalkyl, amioaucyi, alkenyl, alkynyl, alicyclic, aryl. and aralkyl, or together with Y farms a cyclic group including aryl, cyclic alkyl and heterocyclic alkyl; X is selected from the group consisting of aflcylamizio, alkyi(hydroxy), alkyl(curboxyl), alkyl(phospbornate), alkyl, alkenyl, alkynyl, alkyl(sulfonaxe), aryl, carbonylalkyl, 1,1 dibaloalicyl, azninoczbonylamnino, alkylamrinoalkyl, alkoxyalkyl, alkylrhioalkyl, ajicyldujo, alkylazninocarbonyl, alkylcarbonylamjno, alicyclic, aralkyl, and alkyl"ry, all optionally substituted; or together with Y forms a cyclic group including aryl, cyclic alkcyl, and heterocyclic; Y is selected from the group consisting of-H, alkyl, alkenyl, allcyniyj, aryl, alicycuic, aralkyl, aryloxyalcyl, aticoxyalkyl, -cONHut -NRn&, and all except -H arc optionally substituted; or together with X forms a cyclic group including mryl cyclic alkyl, and heterocyclic, I 4 is indepentdently selected from the group consisting of lower alkyl, lower alicyolic, lower aralkyl, and lower aryl; is selected from the group consisting of lower alkyl. lower aryl, lower aralkyl, and lower alicyclic; R6is independently selected from the group consisting of and lower alkyl; Rt 7 is independently selected forr the group consisting of -Hi, lower alkyl, lower alicycie, lower aralkyl, lower aryl. and COR R 5 is independently selected from the group consieting of lower alkyl, lower aralkyl, lower aryl, lower alicyclic, -C(O)R' 0 or together they form a bidenrate alkryl; COMS ID No: SMBI-00629627 Received by iP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:19 SPRUSON FERGUSON NO. 7149 P. 23/45 217 R 0 is selected from the group consisting of-H, lower alky], -NH2, lower arfl. and lower perhaloalkyl; R is selected from the group consisting of alkyl, aryl, -OH, -NH2 and and pharmaceutically acceptable prodrugs and salts thereof; with the provisos that; a) when X is alkyl or alkene, then A is -NR 2; b) X is not alkylamnine and alkylaminoalkyl when an alkyl moiety is substituted with phosphonic esters and acids; and c) A, L, E, J, Y, and X together may only form 0-2 cyclic groups. 201 The prodru0 of claim 141 wherein said oral bioavailabiliy is at least
211. The prodrug of claim 210 wherein said oral bioavailability is at least
212. The prodrug of claim 210 wherein said oral bioavailability is enhanced by compared to the parent drug administered orally. 21J. The prodrug of claim 212 wherein said oral bioavailability is enhanced by at least 109%.
214. A prodrug for delivering a biologically active drug to an animal for a sustained period using compounds of formula I: 0 H O w M-P Z 0H wherein: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:19 SPRUSON FERGUSON NO, 7149 P. 24/45- 218 V is selected from the group consisting of alkyl., aralkyl, alicyclic, aryl, substtutca aryl, heteroaryl, substituted heteroaryl, I-alkenyl, 1-alkynyl. and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom. substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylihiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and'W' are independently selected from the group consisting of alkyl, aralkyl. alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and Z is selected from the group consisting of -CHR 2 OH, -CHROC(O)R', -CHROC(S)R, -CHR'OC(S)OR 3 -CHR'OC(O)SR, -CHRzOCO 2 -OR 2 SR -CHRN,. -CHaryl, -CH(aryl)OH, -CH(CH=CRzz)OH -CH(CaCk')OH, -NRz, -OCOR 3 -OCOzR', -SCOR, -SCO 2 R 3 -NHCOR', -NHCOzR 3 -CH2NHaryl, and -(CH 2 )p-SR 2 R 2 is an R' or -H; R is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R9; and M is selected from the group that attached to PO, 2 P206*, or P 3 09'_ is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:20 SPRUSON FERGUSON NO. 7149-P. 25/45 219
215. The prodrug of claim 214 whereby therapeutic levels of said drug are maintained for at least one hour longer than the levels achieved by oral administration of the bispivaloyloxymethyl (bis-POM) ester.
216. The prodrug of claim 214 whereby therapeutic levels of said FBPase inhibitors are maintained for at least one hour longer after systemic administration relative to an equivalent molar amount of the parent compound administered by the same route.
217. The prodrut of claim 214 wherein MPO 3 2 is an FBPase inhibitor.
218. The prodrug of claim 214 wherein MH or MPOQ- is an antiviral or anticancer agent.
219. The prodrug of claim 218 wherein MH is selected from the group consisting of araA, AZT, d4T, 3TC. l-0-D-ribofuranosyl-1,2,4-triazole-3-carboxamide, PMEA, ACV, 9-(4- hydroxy-3-hydroxymethylbut- I -yl)guanine, 5-yl-carbocyclic 2'-deoxyguanosine, dFdC, araC, F- ara-A, and CdA.
220. A prodrug for delivcring a biologically active drug to an animal with greater selectivity for the liver using compounds of formula I: I wherein: V is selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:20 SPRUSON FERGUSON NO. 7149" P. 26/45-- 220 azyloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to formn a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydrqxy, acyloxy, alkoxycarbonyloxy, alkylthiacarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independemly selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1 -alkenyl, 1-alkynyl, and Z is selected from the group consisting of-CHR2OH, -CHRzOC(O)R -CHR 2 0C(S)R', -CHROC(S)OR, -CHROC(0)SR3. -CWROCOk', -OR 2 .SR., -CHR 2 N, -CH2aryl. -CH(aryl)OH, -CH(CH-CR 2 )OH, -CH(CCRW)OH, -R 2 -NR 2 -OCOR', -OCCOzR, -SCO R -SCO0R, -NHCOR 2 -NHCO2R', -CH2NHaryl, -(CH 2 and -(CH2),.SR R 2 is an R or -H; R' is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and. R' is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' aire not all and b) when Z is -R 2 then at leasE one of V and W is not or and MN is selected from the group that attached to P0 3 2 P206", or P 3 0 9 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and phannaceutically acceptable prodrugs and salts thereof.
221. The prodrug of claim 220 whereby the ratio of a parent drug or a drug metabolite concenration in the liver over a parent drug or a drug metabolite concentration in the plasma is two times greater compared to administration of a parent drug. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 23. FEB. 2004 18:23 SPRUSON FERGUSON NO. 7149 P. 27/45 221
222. The prodrug of claim 221 wherein the liver specificity hlas increqged relativa to administration of M-POJ-. 2-3. The prodrug of claim 220 wherein said biologically active drug is a triphosphate generated in the liver.
224. The prodrug of claiwn 223 wherein MH is selected fom the group consisting of aeA, AZT, d4T, 3TC, I -A-D-ribg tbranosy I- l, 2 4 -triazole-3-carboxamide, ACV, 9 -(4-hydroxy: 3-hydroxymethylbut-. -yl)guanine, S-yl-carbocycrjc Z'-deoxyguanosine, dFdC, amC, F-ana-A, and CdA.
225. The prodrug of claimn 220 wherein the active drug is MPQ 3
226. The prodrug of claim.225 wherein the drug is selected from the group consisting of FdUMP, S-Bromo-( l.P-D-ribotranosyl)imidazole-4ca,,boxamid 1 -mnohopae 6- Amn--epny--Zphshnfrnlprze 6-Chioro. I -isobutyI-2-(2.phosph 0 furanyl)benzimidazole, and 4 -Amnino-7-erhy..5.41uor.. 1 -isobuwyl-2-{2.phosphonos.. fizrnyl)bcnzinaidazole.
227. The prodrug of claimn 220 wherein the active drug is MP,0 9 6 and M is attached via C.
228. The prodrug of clairn 227 wherein the drug is selected from the group consisting of PMEA, PMADAP, H'MEPS, HPMZPA, FPMPA, PMPA, phosphonoformpic acid, and phosphonoacede acid. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:20 SPRUSON FERGUSON NO, 7149 28/45- 222
229. A prodrug for increasing the therapeutic index of a drug by adnirtisterng to an animal compounds of formula I: V 00 H M-PI H W W I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonylaxy, or aryloxycarbonyloxy anached to a carbon atom that is three atoms fom an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an op ti onally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting ofhydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyJ, aralyl, alicyclic, aryl substituted aryl, heteroaryl, substituted heteroaryl, I alkenyl, 1-alkynyl, and -RP; Z is selected from the group consisting of-CHR OH, -CHROC(0)R' -CHROC(s)R', -CHR'OC(S)O -CHO{oC(O)s R, -CHR'OCO2R, -OR 2 -sR2, -CHRN 3 -CHlaryl, -CH(aryl)OH, -CH(CH-CR' 2 )OH, -CH(C=CR )OH, -NR' 2 26 -OCOR', -OCO 2 R' 3 SCOR', -SCOzR, -NHCOR, -NHCO R, -CHzNHatyl, and -(CHz)-SR; R is an R or -H; COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:21 SPRUSON FERGUSON NO. 7149 P. 29/45- 223 R j is selected from the group consisting of alkyl, aryl, aralkyl, and alicyyric; and R 9 is selected from the group consisting of alkyl. aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, 2, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or and 2- M is selected from the group that attached to PO) P20' or P309' is biologically active n vivo. and that is atrachpd to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
230. The prodrug of claim 229 wherein extrahepatic toxicity is reduced.
231. The prodrug of claim 230 wherein M-PO3 'is excreted by the kidney.
232. The prodrug ofclaim 230 wherein the M is selected from the group consisting of PMEA, PMEADAP, HPMPS, HPMPA, FPMPA, and PMPA.
233. The prodrug of claim 230 wherein the gastrointestinal toxicity is reduced.
234. The prodrug of claim 230 wherein central or peripheral nervous system toxicity is reduced.
235. A prodrug for bypassing drug resistance by administering to an animal. compounds of formula I: V M-P Z 0\H W W I wherein: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:21 SPRUSON FERGUSON NO. 7149 30/45- 224 V is selected from the group consisting of -H alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl. and or together V and Z are connected via 3-.5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, anached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted hcteroaryl, 1-alkenyl, I -alkynyl, and Z is selected from the group consisting of -CHROH. -CHROC(O)R', -CHR 2 OC(S)R', -CHR 2 OC(S)OR', -CHR 2 OC(O)SR -CHR 2 OCO 2 R, -SR', -CHR 2 N3. -CHaryl, -CH(aryl)OH, -CHCHCR 2 2 )OIH -CH(CCR')OH, -R 2 -NR 2 -OCOR', -OCOR 3 -SCOR', -SCO2R', -NHCOR', -NHCO 2 R 3 -CHzNHaryl, -(CH2)pOR 2 and 2 R 2 is an R' or -H; R3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alky], aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -RK; and M is selected from the group that attached to PO. P20 3 or p0,"4 is biologically active in vivo, and that is attached to the phosphorus in fonnula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:21 SPRUSON FERGUSON NO. 7149-P. 31/45-
236. The prodrug of claim 235 wherein said resistance arises from decreased cellulae production of M-POa 3
237. The prodrug of claim 238 wherein said compound is an amicancer or antiviral agent.
238. The prodrup of claim 237 wherein M is 5-fluoro-2'-dcoxyuridine and said agent is (5-fluoro-2'-deoxyuridine), or
239. The prodrug of claim 237 wherein said resistance is to an antiviral agent selected from the group consisting of araA, AZT, d4T, 3TC, -b-D-ribofuranosyl-l, 2 ,4-triazole-3. carboxamide, ACV, 9 4 -hydroxy-3-hydroxymethylbut-1-yl)guanine, and 5-yl-carbocyclic 2'- deoxyguanosine.
240. The prodrug of claim 237 wherein the resistance or lack of antihepatitis activity is due to a deficiency in thymidine kinase and said antiviral agent is selected from the group consisting of AZT, d4T. ACV.
241. The prodrug of claim 237 wherein said anticancer agent is selected from the group consisting of dFdC, araC, F-araA, and CdA.
242. formula I: A prodrug for treating cancer by administering to an animal a compound of wherein: COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:21 SPRUSON FERGUSON NO. 7149 P. 32/45 226 V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, hetcroaryl, substituted heteroaryl, 1-alkenyl, l-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxyearbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, Pat is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1 -alkenyl, 1-alkynyl, and Z is selected from the group consisting of-Ci 2 OH, -CHR2OC(O)j, -CHR'OC(S)R', -CHR 2 OC(S)OR'. -CHR'OC(O)SR, -CHRI OCOR 3 -OR z -SR 2 -CHRN3, -CH2ryl, -CH(aryl)OH, -CH(CH=CRl2)OH, -CH(CCR 2 )OH, -R2, -NR 2 -OCOR', OCO W -SCOR', -SCO 2 -NHCOR 2 -NHCO 2 -CH2NHaryl, and -(CH2)-SR; R 2 is an R 3 or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all H; and b) when Z is -R 2 then at least one of V and W is not or and 2- M is selected from the group that attached to P03 P 2 0 6 3 or PO is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:22 SPRUSON FERGUSON NO. 7149-P. 33/45- 227
243. The prodrug of claim 242 wherein the active drug is the triphosphate-ofM-H.
244. The prodrug of claim 242 wherein the active drug is the monophosphare of M-H.
245. The prodrug of claim 242 wherein said prodrug is administered to patients resistant to the parent drug.
246. The prodrug of claim 242 wherein MH is selected from the group consisting of dFdC, araC, P-araA, and CdA.
247. A prodrug for use in treating viral infections by administering towan animal a compound of formula I: V 0 H w i w M-P Z I wherein: V is selected from the group consisting of-H, alkyl, aralkcyl, alicyclic. aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, I-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:22 SPRUSON FERGUSON NO. 7149 P. 34/45 228 aryloxycarbonyloxy, artacned to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, ailyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, l-alkynyl, and -R 9 Z is selected from the group consisting of-CHR'OH, -CHR 2 OC(0)R', -CHR 2 OC(S)R3, -CHR'OC(S)OR', -CHR'OC(O)5R -CHRI OCOR', -OR 2 -SW, -CHR N 3 -CHiaryl, -CH(aryl)OH, -CH(CH-CR')OH, -CH(CCR 2 )OH, -R 2 -NR'2, -OCOR, -OCOzR', -SCOR', -SCOR', -NHCOR', -NHCO 2 R 3 -CH2NHaryl, (CH2)'-OR, and -(CHIz),p-SR; R 2 is an R or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl. and alicyclic; and R' is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, 2, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or and M is selected from the group that attached to PO, P206 or PaO4' is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof.
248. The prodrug of claim, 247 wherein said viral infection is hepatitis.
249. The prodrug of claim 248 wherein MH is selected from the group consisting of AZT, d4T, and ACV.
250. The prodrug of claim 248 wherein said prodrug is administered to patients resistant to the parent drug,
251. The prodrug of claim 248 wherein said hepatitis is hepatitis B.
252. The prodrug of claim 247 wherein viral kinases produce M-PO3". COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:22 SPRUSON FERGUSON NO, 7149-- P. 35/45 229
253. The prodrug of claim 252 wherein said viral infection is hepatitis and said viral kinases are kinases from viruses other than the hepatitis viruses.
254. The prodrug of claim 247 wherein the active drug is the triphosphate of M-H.
255. A prodrug for use in treating liver fibrosis by administering to an animal a compound of formula I; V M-P Z2 H W W. 1 wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, mayl, substituted aryl, hceeroaryl, substituted heteroaryl, 1-alkenyl, 1 -alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy. acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attahed to the phosphorus; or together V and Z arc connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, hereroaryl,-substimuted heteroaryl, 1-alkenyl. 1-alkynyl, and -R 9 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:23 SPRUSON FERGUSON NO. 7149 P. 36/45' 230 Z is selected from the group consisting of-CHR2OH -CIH OC(O)R' -CHROC(S)R, -CHR OC(S)OR' -CHR'OC(O)S, -CHR'OCO 2 -SR 2 -CHR 2 -CH 2 aryl. -CH(aryl)OH, -CH(CH=CR 2 2 )OH, .CH(C=CR')OH, -R 2 -NR 2 2 -OCOR, -OCO 2 -SCOR', -SCO 2 -NHCOR 2 -NHCOR', -CH 2 NHa ry l -(CH 2 ),OR 2 and R' is an R 3 or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkcyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p i& an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to P20 or Pj 3 0 9 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof
256. A prodrug for use in treating hyperlipidemia by administering to an animal a compound of formula I: V 0O H M-P Z H I wherein: V is selected from the group consisting of alky], aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, I-alkynyl, and -RK; or together V and Z arc connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:23 SPRUSON FERGUSON NO. 7149 P. 37/45 231 aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I hctcroatom, that is fUsed to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substitucnt selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkcyl, aralkyl, alicyclic, aryj, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, I-alkynyl, and -R9; Z is selected from the group consisting of-CHROH -CHR 2 OC(O)R', -CHROC(S)R, -CHR 2 OC(S)OR', -CHROC(O)SR, -CHR 2 OCO 2 R -OR 3 -SR 2 -CHR 2 N3, -CH2aryl, -CH(aryl)OH, -CH(CH=CRZ)OH, -CH(C=CRZ)OH, -R2, -NR2, -OCOR', -OCO 2 -SCOR', -SCO 2 R3, -NHCOR2, -NHCO 2 -CHMNHaryl, -(CH),P-OR 2 and -(CHz,)pSR 2 R' is an R' or-H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R' is selected from the group consisting ofalkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -eR, then at least one of V and W is not or -R 9 and M is selected from the group that attached to P0 3 PzO6 or P, is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaceutically acceptable prodrugs and salts thereof
257. The prodrug of claim 256 wherein the byperlipidemia agent is a squalene synthase inhibitor. COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:23 SPRUSON FERGUSON NO. 7149 P, 38/45 232
258. The prodrug of claim 257 wherein said squalene synthase inhibitor is selected from the group consisting of 1-hydroxy-.3.(ethylpentylamino)-propylidene ,1bisphosphonic acid, BMS 187745 and analogs thereof.
259. A prodrug for use in treating parasitic infections by administering to an animal a compound of formula I: 0/0 H M-P HZ 0 w w I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, l-alkenyl, I-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 artoms to form a cyclic group, optionally containing I heteroatorm, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted hcteroaryl, I-alkcenyl, 1-alkynyl, and -R9; Z is selected from the group consisting of-CHR 2 OH, -CHR2OC(O)R, -CHR 2 OC(S)R 3 -CIR 2 OC(S)OR', -CHR'OC(O)SR', -CHROCOzR', -ORt -SR 2 COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:23 SPRUSON FERGUSON NO. 7149 P. 39/45 233 -CHR'N3, -CH 2 aryl, -CH(aryl)OH. -CH(CH-CR22)OH, -CH(CCR)OH, -NR2, -OCOR 3 -OCOR, S CO -SCOzR', -NHCOR 2 -NHCO 2 -CH2NHaryl, -(CH 2 aad -(CHz)p-SRz; R 2 is an R 3 or -H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl. and alicyclic; and R 9 is selected from the group consisting ofalkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: 4 a) V, Z, W, and W' are not all and b) when Z is -R2, then at least one of V and W is not or -R 9 and M is selected from the group that attached to PO P 2 06, or P 3 0,' is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atoim; and pharmnnaceutically acceptable prodrugs and salts thereof.
260. A prodrug for use in delivering diagnostic imaging agents to the liver comprising administration to an animal of compound of formula r; V 0H M-P Z I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted ary], heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB,2004 18:24 SPRUSON FERGUSON NO. 7149 P. 40/45 234 together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryi group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W arc connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H. alkyl, aralkyl, alicyclic, aryl, substituted aryl, heter-aryl, substituted heteroaryl, 1 -alkenyl, I -alkynyl, and Z is selected from the group consisting of -CHR 2 OH, -CHRzOC(O)R 3 .CHR 2 OC(S)R', -CHR 2 OC(S)OR, -CHR 2 OC(0)SR, -CHR'OCO 2 -OR 2 -SR'. -CHR2N 3 -CHzaryl, -CH(aryl)OH. -CH(CH-CR2)OH, -CH(CaCR 2 )OH, -R3, -NR 2 -OCOR', -OCO 2 -SCOR, -SCO 2 R, -NHCOR 2 -NHCOzR', -CH2NHaryl, -(CH 2 )-OR 2 and -(CH2),-SR 2 R is an R 3 or-H; R 3 is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is then at least one of V and W is not or and M is selected from the group that attached to PO 2 P 2 06 or P094 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and phamaceutically acceptable prodrugs and salts thereof.
261. A prodrug for use in making a prodrug of a compound drug having a -PO0 2 moiety comprising, COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:24 SPRUSON FERGUSON NO. 7149 P. 41/45 235 a) transforming said phosph(on)ace into a compound of formula I: V M-P Z wr W-- wherein: V is selected from the group consisting of-H, alkyl. aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkyny), and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroaiom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and aryloxycarbonyloxy, attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; W and W' are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, 1-alkynyl, and -R9; Z is selected from the group consisting of-CHR 2 OH, -CHR 2 OC(0)R 3 -CIHROC(S)R, -CHROC(S)OR', -CHR'OC(O)SR 3 -CHR 2 OCO 2 R 3 -OR 2 -SR 2 -CHR 2 N 3 -CHIaryl, -CH(aryl)OH, -CH(CH-CR 2 )OH, -CH(CaCR')OH, .R -NR2, -OCOR', -OCO 2 R 3 -SCOR 3 -SCO 2 -NHCOR 2 -NHCO2R'. -CH 2 NHaryl, .(CH 2 )p-OR z and *(CH)p-SR 2 R2 is an R3 or *H; R 3 is selected from the group consisting of alkyl, aryl. aralkyl, and alicyclic; and COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:24 SPRUSON FERGUSON NO. 7149 P. 42/45 236 R 9 is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is -R 2 then at least one of V and W is not or -R 9 and M is selected from the group that attached to PO, 2 P 2 0 6 3 or P309 4 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; a and pharmaceutically acceptable prodrugs and salts thereof.
262. A prodrug for use in enhancing the phannacodynamic half-life of a parent drug comprising a prodrug of formula I' V M-P WA W I wherein: V is selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkanyl, 1-alkynyl, and -R 9 or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing I heteroatom, substituted with hydroxy, acyloxy, alkoxycarbonyloxy, or aryloxycarbonyloxy attached to a carbon atom that is three atoms from an oxygen attached to the phosphorus; or together V and Z are connected via 3-5 atoms to form a cyclic group, optionally containing 1 heteroatom, that is fused to an aryl group at the beta and gamma position to the oxygen attached to the phosphorus; or together V and W are connected via 3 carbon atoms to form an optionally substituted cyclic group containing 6 carbon atoms and substituted with one substituent selected from the group consisting of hydroxy, acyloxy, alkoxycarbonyloxy, alkylthiocarbonyloxy, and COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20 FEB. 2004 18:25 SPRUSON FERGUSON NO. 7149 P. 43/45"- 237 aryloxycarbonyloxy. attached to a carbon atom that is three aioms from an oxygen attached to the phosphorus; W and WVP are independently selected from the group consisting of-H, alkyl, aralkyl, alicyclic, aryl, substituted aryl, heteroaryl, substituted heteroaryl, 1-alkenyl, l-alkynyl, and Z is selected from the group consisting of -CHR3OH, 4CHR 2 OC(O)R', -CHROC(S)R, -CHRzOC(S)OR, -CHR 2 OC(0)SR, -CHR'OCOR', -OR 2 -SR', *CHR 2 N 3 -CH 2 aryl, -CH(aryl)OH, -CH(CH-CR22)OH, -CH(C=CR')OH, -NR2, -OCOR', *0CO 2 R 3 -SCOR', -SCO 2 R, .NHCOR, -NHC0 2 R, -CH2NHaryl, <-CHz)p-OR, and R 2 is an R' or -H; R' is selected from the group consisting of alkyl, aryl, aralkyl, and alicyclic; and R! is selected from the group consisting of alkyl, aralkyl, and alicyclic; p is an integer from 2 to 3; with the provisos that: a) V, Z, W, and W' are not all and b) when Z is then at least one of V and W is not-H, or -R7; and M is selected from the group that attached to P 2 06 3 or P 3 0 4 is biologically active in vivo, and that is attached to the phosphorus in formula I via a carbon, oxygen, or nitrogen atom; and pharmaccutically acceptable prodrugs and salts thereof.
263. A prodrug of claim 1, where alpha, hydrogen, and the phosphoryl oxygen are in as configuration. Dated 20 February, 2004 Metabasis Therapeutics, Inc. Patent Attorneys for the Applicaut/Nominated Person SPRUSON FERGUSON COMS ID No: SMBI-00629627 Received by IP Australia: Time 18:45 Date 2004-02-20
AU2004200670A 1998-03-06 2004-02-20 Novel Prodrugs for Phosphorous-containing Compounds Expired AU2004200670B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2004200670A AU2004200670B2 (en) 1998-03-06 2004-02-20 Novel Prodrugs for Phosphorous-containing Compounds

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
US7716598P 1998-03-06 1998-03-06
US7716498P 1998-03-06 1998-03-06
US60/077165 1998-03-06
US60/077164 1998-03-06
AU30699/99A AU767599B2 (en) 1998-03-06 1999-03-05 Novel prodrugs for phosphorus-containing compounds
PCT/US1999/004908 WO1999045016A2 (en) 1998-03-06 1999-03-05 Novel prodrugs for phosphorus-containing compounds
AU2004200670A AU2004200670B2 (en) 1998-03-06 2004-02-20 Novel Prodrugs for Phosphorous-containing Compounds

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU30699/99A Division AU767599B2 (en) 1998-03-06 1999-03-05 Novel prodrugs for phosphorus-containing compounds

Publications (2)

Publication Number Publication Date
AU2004200670A1 true AU2004200670A1 (en) 2004-03-18
AU2004200670B2 AU2004200670B2 (en) 2008-08-28

Family

ID=34279881

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2004200670A Expired AU2004200670B2 (en) 1998-03-06 2004-02-20 Novel Prodrugs for Phosphorous-containing Compounds

Country Status (1)

Country Link
AU (1) AU2004200670B2 (en)

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0338372A3 (en) * 1988-04-22 1991-10-09 American Cyanamid Company Solubilized pro-drugs

Also Published As

Publication number Publication date
AU2004200670B2 (en) 2008-08-28

Similar Documents

Publication Publication Date Title
AU767599B2 (en) Novel prodrugs for phosphorus-containing compounds
US6312662B1 (en) Prodrugs phosphorus-containing compounds
AU775332B2 (en) Novel phosphorus-containing prodrugs
US6284748B1 (en) Purine inhibitors of fructose 1,6-bisphosphatase
US8664195B2 (en) Phosphorus-containing prodrugs
WO1998039344A9 (en) Novel purine inhibitors of fructose-1,6-bisphosphatase
WO2008019309A1 (en) Novel inhibitors of fructose 1,6-bisphosphatase
HUE029022T2 (en) Phosphoramidate derivatives of 5-fluoro-2&#39;-deoxyuridine for use in the treatment of cancer
AU2004200670B2 (en) Novel Prodrugs for Phosphorous-containing Compounds
EP1634886A2 (en) Novel phosphorus-containing prodrug

Legal Events

Date Code Title Description
FGA Letters patent sealed or granted (standard patent)
MK14 Patent ceased section 143(a) (annual fees not paid) or expired