WO1996033201A1 - Acyclovir derivatives as antiviral agents - Google Patents

Acyclovir derivatives as antiviral agents Download PDF

Info

Publication number
WO1996033201A1
WO1996033201A1 PCT/IB1996/000454 IB9600454W WO9633201A1 WO 1996033201 A1 WO1996033201 A1 WO 1996033201A1 IB 9600454 W IB9600454 W IB 9600454W WO 9633201 A1 WO9633201 A1 WO 9633201A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
virus
bis
acyclovir
pharmaceutical composition
Prior art date
Application number
PCT/IB1996/000454
Other languages
French (fr)
Inventor
Gilles Gosselin
Jean-Louis Imbach
Original Assignee
Centre National De La Recherche Scientifique (Cnrs)
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 FR9504797A external-priority patent/FR2733234B1/en
Priority claimed from US08/465,450 external-priority patent/US5627185A/en
Application filed by Centre National De La Recherche Scientifique (Cnrs) filed Critical Centre National De La Recherche Scientifique (Cnrs)
Priority to AU55111/96A priority Critical patent/AU5511196A/en
Publication of WO1996033201A1 publication Critical patent/WO1996033201A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07FACYCLIC, CARBOCYCLIC OR HETEROCYCLIC COMPOUNDS CONTAINING ELEMENTS OTHER THAN CARBON, HYDROGEN, HALOGEN, OXYGEN, NITROGEN, SULFUR, SELENIUM OR TELLURIUM
    • C07F9/00Compounds containing elements of Groups 5 or 15 of the Periodic Table
    • C07F9/02Phosphorus compounds
    • C07F9/547Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom
    • C07F9/6561Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings
    • C07F9/65616Heterocyclic compounds, e.g. containing phosphorus as a ring hetero atom containing systems of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring or ring system, with or without other non-condensed hetero rings containing the ring system having three or more than three double bonds between ring members or between ring members and non-ring members, e.g. purine or analogs
    • 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
    • Y02PCLIMATE CHANGE MITIGATION TECHNOLOGIES IN THE PRODUCTION OR PROCESSING OF GOODS
    • Y02P20/00Technologies relating to chemical industry
    • Y02P20/50Improvements relating to the production of bulk chemicals
    • Y02P20/55Design of synthesis routes, e.g. reducing the use of auxiliary or protecting groups

Definitions

  • This application is directed to phosphotriester derivatives of the drug molecule known as acyclovir (ACV) , and their use as antiviral agents.
  • ACV acyclovir
  • nucleoside analogs which, in order to exert their biological activity, need to be phosphorylated to the corresponding triphosphates.
  • Intracellular metabolism, such as phosphorylation, of nucleosides to active nucleotides reguires the successive action of three kinases, the first one being highly selective and strongly regulated.
  • a nucleosidic analog is not phosphorylated by the first kinase, it cannot exert its inhibitory activity.
  • viruses such as herpes simplex virus (HSV)
  • HSV herpes simplex virus
  • Some viruses encode for a viral kinase which is capable of phosphorylating some nucleoside analogs that are not phosphorylated by cellular kinases.
  • some acyclonucleosides which are not substrates for the first cellular kinase mentioned above do not show any activity against viruses which do not provide their own kinase.
  • acyclovir ACV
  • acyclovir has no activity against many other viruses, such as human immunodeficiency virus (HIV) or hepatitis B virus (HBV) , which do not provide the necessary viral kinases.
  • HIV human immunodeficiency virus
  • HBV hepatitis B virus
  • ACV is poorly active against those herpetic viruses in which thymidine kinase activity is either decreased or absent, such as Epstein-Barr virus (EBV) or Cyto perennialovirus (CMV) .
  • EBV Epstein-Barr virus
  • CMV Cyto egalovirus
  • acyclovir-resistant herpes simplex viruses can appear both in vitro and in vivo, especially in highly immunodepressed patients. This resistance usually arises from a modification of the viral thymidine kinase, which results in a decrease or disappearance of the phosphorylation of ACV.
  • AAVMP acyclovir monophosphate
  • phosphotriesters of ACV can be considered to be new therapeutic species since their spectrum of activity is much broader than that of ACV.
  • Figure 1 is an exemplary synthetic scheme for certain preferred compounds of the invention.
  • Figure 2 is a graph showing the effect of acyclovir on vaginal lesion scores in mice infected with HSV-2. Intraperitoneal treatments were given twice daily for 5 days starting 24 hours after virus challenge.
  • Figure 3 is a graph showing the effect of bis(SPTE)ACVMP on vaginal lesion scores in mice infected with HSV-2. Intraperitoneal treatments were given twice daily for 5 days starting 24 hours after virus challenge.
  • Phosphotriester compounds are provided corresponding to the general formula I:
  • R is (CH 2 ) n -S-X
  • Z is O or S
  • Y and U independently, are alkyl, aryl or a sugar moiety, optionally substituted with OH, SH or NH; n is 1 to 4, preferably 1 or 2; and
  • ACV is an acyclovir moiety having the formula:
  • Y and U are preferably C to C-, alkyl, phenyl, benzyl, glucose, mannose, rhamnose, or cyclofuranose.
  • X represents S-U
  • U preferably represents the radical (CH J I -X 1 where X 1 represents H, OH, SH or NH 2 and n 1 is equal to 1 to 4, preferably 1 or 2.
  • compound (I) be such that R represents (CH 2 ) 2 -S-S-(CH 2 ) 2 -OH.
  • Y is CH 3 or (CH 3 ) 3 C.
  • R is alkyl, especially CH 3 or (CH 3 ) 3 C.
  • the compounds of the present invention are prepared by processes known to those skilled in the art. Such processes are described in patent application PCT/FR93/00498, published as WO 93/24510.
  • functional groups such as R and, optionally, ACV, can be protected by suitable protecting groups, followed by deprotection of said functional groups to give compounds of formula I.
  • a compound of formula II For example, a compound of formula II:
  • Y and U are alkyl, aryl or saccharide moieties that may be optionally substituted, in particular with an OH, SH or NH 2 group, and suitably protected, in order to obtain the compound of formula I in a protected form that can then be deprotected.
  • the compounds of the present invention can be included in antiviral pharmaceutical compositions that include a phosphotriester compound of the invention as an active substance together with a pharmaceutically acceptable diluent, excipient or carrier.
  • the present invention is further directed to antiviral compositions of increased therapeutic activity, for example, anti-HBV, anti-HIV, anti-CMV and anti-VZV compositions, together with other antiviral compositions directed against other viruses in which thymidine kinase activity is either decreased or absent.
  • antiviral compositions of increased therapeutic activity for example, anti-HBV, anti-HIV, anti-CMV and anti-VZV compositions, together with other antiviral compositions directed against other viruses in which thymidine kinase activity is either decreased or absent.
  • Compounds of formula la may be prepared in which the bioreversible protecting group, a substrate for cellular carboxyesterases, is an S-acylthioalkyl group.
  • Acyclovir (preferably suitably protected at position 2) is coupled with an appropriate phosphitylating reagent. Deprotection yields compounds of formula la in which the R group can be, independently, alkyl, aryl, heterocyclyl, sugar, or another similar group.
  • R group can be, independently, alkyl, aryl, heterocyclyl, sugar, or another similar group.
  • Thin layer chro atography was performed on Merck 6OF 254 silica plates (Art. 5554) .
  • Column chromatography on silica gel were carried out with Merck 60 H silica (Art. 7719) or with RP2 Merck silanized silica (Art. 7719) .
  • UV spectra were recorded on a UVIKON 810 spectrophotometer.
  • Mass spectra were obtained using a JEOL JMS DX 300 apparatus by the FAB ionization method, in positive or negative mode, in a matrix of glycerol (G) , glycerol/thioglycerol (GT) or 3-nitrobenzyl alcohol (NBA) .
  • Proton NMR spectra were recorded on a Varian EM 360 apparatus or on a Bruker AC 250 apparatus. The chemical shifts are expressed in ppm relative to the tetra ethylsilane (TMS) signal.
  • the multiplicity and the appearance of the signals observed by NMR are indicated by one or more letter(s) : s(singlet), d(doublet), t(triplet), m(multiplet) , orb (broad).
  • Phosphorus NMR spectra were recorded on a Bruker WP 200 SY apparatus with proton decoupling. The chemical shifts are expressed in ppm relative to the H 3 P0 4 signal, which is taken as the external reference.
  • Figure I depicts an exemplary synthetic scheme for preparing certain compositions in accordance with the invention.
  • Bis-S-acetylthioethyl, BIS(SATE), and bis-S- pivaloylthioethyl, BIS(SPTE), phosphotriester derivatives of acyclovir are shown. These compounds correspond to formula la wherein R is CH 3 and (CH 3 ) 3 C.
  • Tetrazole (210 mg, 3.0 mmol) was added to a stirred solution of N 2 -(p-anisyldiphenylmethyl)-9-[ (-2-hydroxy- ethoxy)methyl] guanine (500 mg, 1 mmol) , prepared in accordance with Martin et al . t J. Med . Chem . , 1986, 29 , 1384-1386, and compound 5 (445 mg, 1.2 mmol) in THF (3.0 mL) . After 35 minutes at room temperature, the reaction mixture was cooled to -40 ⁇ c and a solution of 3-chloroperbenzoic acid (407 mg, 1.3 mmol) in methylene chloride (5 mL) was added.
  • Mass spectra (matrix GT) FAB positive: 782 (M+M) + ; 273 (trityl) + ; 103 (CH 3 C0SCH 2 CH 2 ) + ; FAB negative: 780 (M-H)'; 678 (M-CH 3 C0SCH 2 CH 2 ) ' .
  • Tetrazole (126 mg, 1.8 mmol) was added to a stirred solution of N 2 -(p-anisyldiphenylmethyl)-9-[ (-2-hydroxy- ethoxy) ethyl] guanine (300 mg, 0.60 mmol), prepared in accordance with Martin et al . , J. Med. chem . , 1986, 29 , 1384- 1386, and compound £ (408 mg, 0.90 mmol) in THF (1.8 mL) . After 35 minutes at room temperature, the reaction mixture was cooled to -40°C and a solution of 3-chloroperbenzoic acid (342 mg, 0.99 mmol) in methylene chloride (4 mL) was added.
  • Mass spectra (matrix GT) , FAB positive: 866 (M+M) + ; 273 (trityl) + ; 145 ( (CH 3 ) 3 CCOSHC 2 CH 2 ) + ; FAB negative: 864 (M-H)"; 720 (M-(CH 3 ) 3 CCOSCH 2 CH 2 )-.
  • HIV-1 replication LAI isolate
  • RT reverse transcriptase
  • the toxic effect in non-infected CEM cells was assessed by a calorimetric reaction based on the capacity of living cells to reduce 3-(4,5 dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide into formazan after incubation for five days in the presence of various concentrations of the compounds. The results were expressed as the lowest concentration of compound which resulted in at least 50% inhibition of the formation of formazan (CD*,) .
  • the experimental procedure used for evaluation of activities against herpes simplex virus type 1 (HSV-1) was as described by Genu-Dellac et al . in N cleosides and Nucleotides, 1991, 10 , 1345.
  • HSV-2 herpes simplex virus type 2
  • mice weighing approximately 20 g each at the start of the experiment, were intravaginally infected with HSV-2 (E194 strain) . This was accomplished in a 3-step process. First, the vagina of each mouse was swabbed for 5 seconds with a cotton-tipped applicator dipped in 0.1 N NaOH. Approximately 30-60 minutes later each vagina was dry swabbed for 5 seconds. Then an applicator dipped in virus medium (containing approximately 10 7 plaque forming units of virus/mL) was used to swab each mouse for 20 seconds. Twenty-four hours after infection with virus, compounds were administered by i.p. injection. Treatments were given twice daily for 5 consecutive days.
  • virus medium containing approximately 10 7 plaque forming units of virus/mL
  • Vaginal virus titers were made by titration of virus obtained from vaginal swabs (placed in 1 mL of cell culture medium) 3 days after virus inoculation. These titrations were conducted in MA-104 African green monkey kidney cells in 96-well plates. Calculation of virus titer was made by the 50% endpoint dilution method of Reed and Muench (Am. J. Hyg. , 1938, 27, 493).
  • MHV-68 murine gammaherpes virus type 68
  • Acyclovir was initially prepared as a 4 mM solution in cell culture medium before diluting to micromolar concentrations.
  • African green monkey kidney (Vero) cells were seeded into 12- well plates, then allowed to grow to confluency. The cells were infected with approximately 100 plaque forming units of MHV-68. After 1.5 hours, the medium was changed to include antiviral drug or drug-free medium (MEM, 2% FBS and 0.5% SeaPlaque agarose obtained from FMC Corp. , Rockland, MD) . At 7 days the plates were fixed for 15 minutes with 10% buffered formalin prior to removing the agar overlay.
  • MEM antiviral drug or drug-free medium
  • the cells were then stained with 0.2% crystal violet in 20% methanol.
  • the plaques were counted at 17x magnification using a Plaque Viewer (Bellco Glass Co. , Vineland, NJ) .
  • ECy-, values were determined by plotting inhibition versus drug concentration.
  • the compounds of the present invention were tested for their antiviral activity against human CMV using a plaque reduction assay.
  • the procedure was as follows:
  • the antiviral activity of the compounds against human CMV was determined in MRC-5 cells (a continuous line of diploid, human (male) embryonic lung cells) .
  • the compounds were diluted in MEM, 2% FBS, 0.1% NaHC0 3 and 50 ⁇ g/mL of gentamicin.
  • the growth medium was decanted from established onolayers of MRC-5 cells in 24-well tissue culture plates.
  • Human CMV (1 mL) diluted in test medium, was placed in each well except those to be used for cell controls, which received 1 mL of sterile test medium. The virus was allowed to adsorb to the cells while the plates were centrifuged (2200 rpm, 30 minutes) at room temperature.
  • the medium was aspirated from the plates and 0.8 mL of the compound (varying concentrations) was placed in each well.
  • Test medium 0.8 mL without any compound was added to each cell control and virus control well.
  • the plates were placed in an incubator at 37°C in a moist atmosphere of 5% C0 2 , 95% air until plaques could be distinguished in the virus control wells.
  • the cells were observed microscopically for morphological changes due to compound cytotoxicity before the medium was aspirated from the wells.
  • the cells were stained with crystal violet for 15 minutes, after which the staining solution was aspirated, the plates rinsed with water, inverted and dried at room temperature. Plaques were counted by the use of a dissecting microscope.
  • ED J Q and CD M values were determined by regression analysis of the viral plaque data and the visual cell toxicity data, respectively.
  • the antiviral activity of the compounds of the present invention against murine CMV (MCMV) was determined by a CPE (cytopathic effect) inhibition assay, which was performed as follows:
  • Growth medium was decanted from 96-well tissue culture plates containing monolayers of 3T3 cells (a continuous line of contact-inhibited mouse fibroblasts) . Varying concentrations of compound were added to the wells (4 wells/dilution, 0.1 mL/well of each compound).
  • Compound diluent medium (MEM, without serum, 0.18% NaHC0 3 , 50 ⁇ g of gentamicin/mL) was added to the cell and virus control wells
  • Virus diluted in the test medium, was added to all the compound test wells of the plate (3 wells/dilution) and to virus control wells at 0.1 mL/well.
  • Test medium without virus was added to all toxicity control wells ( 1 well/dilution) and to cell control wells at 0.1 mL/well.
  • the plates were incubated at 37°C in a humidified incubator with 5% C0 2 , 95% air until virus control wells had adequate CPE readings.
  • Cells in test and virus control wells were then examined microscopically and virus CPE was graded on a scale of 0-4, with 0 being no CPE and 4 being 100% CPE.
  • the cells in toxicity control wells were observed microscopically and graded for morphological changes due to cytotoxicity. ED M and CD JO values were determined by regression analysis of the viral CPE data and the visual toxicity data, respectively.
  • the compounds of the present invention were also assayed for their activity against varicella zoster virus (VZV) using a plaque reduction assay.
  • VZV varicella zoster virus
  • the procedure was as described for the assay against human CMV, except that 0.5 mL of VZV was added to each well, and the virus was allowed to adsorb to the cells during a 1 hour stationary incubation period.
  • the compounds of the present invention were designed to be stable in biological fluids and various organs, thus allowing them to reach targeted cells.
  • the significant stability of bis(SPTE)ACVMP in human serum was demonstrated by its half-life of 14 hours.
  • the ex vivo procedure for half-life determination was as follows: Decomposition of the compound was studied at 37°C in human serum. During incubation, aliquots (80 ⁇ L) were removed and injected into a cleaning precolumn (Guard-Pak, Waters) which was equilibrated with triethylammonium acetate buffer (20 mM, pH 6.6). Protein and other undesired components were allowed to elute, while compounds to be analyzed were trapped.
  • Anti-HBV activity in transfected HepG2 (2.2.15) cells, of bis(S-acyl-2-thioethyl)phosphotriester derivatives of acyclovir, (compound 9_, Figure I) , as compared with those of the parent nucleoside and of 2',3'-dideoxyguanosine (ddG) and 2',3'-dideoxycytidine (ddC) are shown in Table 1.
  • EC ⁇ and EC W represent the molar concentrations which provide 90% inhibition and 50% inhibition, respectively, of HBV replication.
  • CC ⁇ represents the molar concentration which reduces the viability of non-infected cells by 50%. The data show that both acyclovir derivatives significantly inhibit HBV activity.
  • COMBOSTAT combination analysis program indicated that there was synergy between bis(SATE)ACVMP and 3TC at EC 50 and EC,,-,.
  • bis(SATE)ACVMP was mixed with 3TC at a 10:1 molar ratio.
  • Anti-HIV activity in CEM TK- cells, of bis(S-acyl-2- thioethyl) phosphotriesters derivatives of acyclovir, as compared with those of the parent nucleoside and of AZT are shown in Table 3.
  • Composition EC 5 o( ⁇ M) CCjo( ⁇ M)
  • EC* represents the molar concentration which produces a 50% inhibition of HIV replication. Significant inhibition of HIV replication was observed.
  • Anti-HSV-1 activity, in MRC-5 cells, of bis(S-acyl-2- thioethyl)phosphotriesters derivatives of acyclovir is compared to that of acyclovir in Table 4.
  • EC W represents the molar concentration which produces a 50% inhibition of HSV-1 replication. Significant activity against HSV was observed.
  • Table 5 shows the survival and vaginal virus titer data in mice infected with HSV-2. Compared to acyclovir, bis(SPTE)ACVMP elicited a superior response with 60% survival in the 100 and 50 mg/kg/day groups of bis(SPTE)ACVMP. Slightly less virus was recovered from lesions of bis(SPTE)ACVMP-treated mice compared to acyclovir-treated mice, however the differences were not significant.
  • Table 6 shows average daily lesion scores.
  • Bis(SPTE)ACVMP at 100 mg/kg/day produced a good response in lesion score reduction over the entire time frame.
  • the data in Table 6 is also depicted graphically in Figures 2 and 3. It is clear from these figures that bis(SPTE)ACVMP is more effective in inhibiting lesion formation than acyclovir.
  • the results discussed above indicate that bis(SPTE)ACVMP was effective against HSV-2 vaginal infection in mice. Acyclovir was effective at the outset, but its effect waned after the discontinuation of treatment.
  • Antiviral activity against MHV-68 is exhibited by the data shown in Table 7.
  • the EC W for bis(SPTE)ACVMP was 8 ⁇ M and the EC M for acyclovir was 5 ⁇ M.
  • Antiviral activity of the compounds of the present invention against human CMV are shown in Table 8.
  • Bis(SPTE)ACVMP was approximately 4-fold more effective in reducing plaques than was acyclovir.
  • Antiviral activity of the compounds of the present invention against murine CMV as determined by a CPE (cytopathic effect) inhibition assay is shown in Table 9.
  • the data indicates that both bis(SATE)ACVMP and bis(SPTE) CVMP were active against murine CMV, with the latter compound being slightly more active.
  • Antiviral activity of the compounds of the present invention against varicella zoster virus (VZV) is shown in Table 10.
  • the data indicates that both bis(SATE)ACVMP and bis(SPTE)ACVMP were as active as acyclovir in the plaque reduction assay.
  • CD50 b > 100 ⁇ M >100 ⁇ M > 100 ⁇ M >100 ⁇ g/ml
  • Not Determinable UA514 is Bis(SPTE)ACVMP.
  • UA491 is Bis(SATE)ACVMP.
  • UA486 is Acyclovir.
  • DHPG is Ganciclovir.
  • Tic > 16 >8.3 >36 a concentration at which the average viral CPE is reduced to 50% of that seen in the virus controls
  • CPE Cytopathic Effect UA514 is Bis(SPTE)ACVMP.
  • UA491 is Bis(SATE)ACVMP.
  • UA486 is Acyclovir.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Biochemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Phosphotriester derivatives of acyclovir having potent antiviral inhibitory activity are provided. Compounds of the invention bear bioreversible protecting groups that can be cleaved intracellularly.

Description

ACYCLOVIR DERIVATIVES AS ANTIVIRAL AGENTS
CROSS REFERENCE TO PRIOR APPLICATIONS
This application is related to and claims priority of French patent application Serial Number 95-04797, filed April 21, 1995. This application is also related to United
States application Serial No. 465,450 filed June 5, 1995. Each of the foregoing is incorporated herein by reference.
BACKGROUND OF THE INVENTION
This application is directed to phosphotriester derivatives of the drug molecule known as acyclovir (ACV) , and their use as antiviral agents.
In a prior patent application, PCT/FR93/00498, which published as WO 93/24510, a very general approach that allows for intracellular delivery of ononucleotides using nucleosidic phosphotriesters bearing two bioreversible protecting groups was described.
It has now been found that this approach can be used for those nucleoside analogs which, in order to exert their biological activity, need to be phosphorylated to the corresponding triphosphates. Intracellular metabolism, such as phosphorylation, of nucleosides to active nucleotides reguires the successive action of three kinases, the first one being highly selective and strongly regulated. As a consequence, if a nucleosidic analog is not phosphorylated by the first kinase, it cannot exert its inhibitory activity.
Some viruses, such as herpes simplex virus (HSV) , encode for a viral kinase which is capable of phosphorylating some nucleoside analogs that are not phosphorylated by cellular kinases. In particular, some acyclonucleosides which are not substrates for the first cellular kinase mentioned above do not show any activity against viruses which do not provide their own kinase. This is the case for the known antiviral drug, acyclovir (ACV) , which has anti-herpetic activity due to its selective monophosphorylation by thy idine kinase of herpes simplex virus. However, acyclovir has no activity against many other viruses, such as human immunodeficiency virus (HIV) or hepatitis B virus (HBV) , which do not provide the necessary viral kinases. Moreover, ACV is poorly active against those herpetic viruses in which thymidine kinase activity is either decreased or absent, such as Epstein-Barr virus (EBV) or Cyto egalovirus (CMV) . Further, it has been well established that acyclovir-resistant herpes simplex viruses can appear both in vitro and in vivo, especially in highly immunodepressed patients. This resistance usually arises from a modification of the viral thymidine kinase, which results in a decrease or disappearance of the phosphorylation of ACV.
The intracellular delivery of acyclovir monophosphate (ACVMP) from various kinds of bioreversible phosphotriesters, as described in the patent application PCT/FR93/00498, is thus capable of enlarging the antiviral activity spectrum of this molecule.
Compared to ACV, phosphotriesters of ACV can be considered to be new therapeutic species since their spectrum of activity is much broader than that of ACV.
It is, therefore, an object of this invention to provide phosphotriester derivatives of acyclovir together with antiviral pharmaceutical formulations including such compounds.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is an exemplary synthetic scheme for certain preferred compounds of the invention.
Figure 2 is a graph showing the effect of acyclovir on vaginal lesion scores in mice infected with HSV-2. Intraperitoneal treatments were given twice daily for 5 days starting 24 hours after virus challenge. Figure 3 is a graph showing the effect of bis(SPTE)ACVMP on vaginal lesion scores in mice infected with HSV-2. Intraperitoneal treatments were given twice daily for 5 days starting 24 hours after virus challenge.
SUMMARY OF THE INVENTION
Phosphotriester compounds are provided corresponding to the general formula I:
(RO)2-P(=0)-0-ACV I Wherein:
R is (CH2)n-S-X;
X is C(=Z)-Y or S-U;
Z is O or S;
Y and U, independently, are alkyl, aryl or a sugar moiety, optionally substituted with OH, SH or NH; n is 1 to 4, preferably 1 or 2; and
ACV is an acyclovir moiety having the formula:
Figure imgf000005_0001
In accordance with the present invention, Y and U are preferably C to C-, alkyl, phenyl, benzyl, glucose, mannose, rhamnose, or cyclofuranose. In one embodiment, when X represents S-U, U preferably represents the radical (CHJ I-X1 where X1 represents H, OH, SH or NH2and n1 is equal to 1 to 4, preferably 1 or 2.
It is preferred for some embodiments that compound (I) be such that R represents (CH2)2-S-S-(CH2)2-OH. In another preferred embodiment, X is C(=Z)-Y, and Y is CH3 or (CH3)3C.
In other preferred embodiments, R represents (CH2)B-S- C(«0)-CH3or (CH2)B-S-C(=0)-tBu, with n- l or 2.
Also preferred are compounds of formula la:
Figure imgf000006_0001
la
wherein R is alkyl, especially CH3 or (CH3)3C.
In general, the compounds of the present invention are prepared by processes known to those skilled in the art. Such processes are described in patent application PCT/FR93/00498, published as WO 93/24510. In preparing compounds of the invention, functional groups such as R and, optionally, ACV, can be protected by suitable protecting groups, followed by deprotection of said functional groups to give compounds of formula I. For example, a compound of formula II:
O—P—0— - iACV
I
0"
II in which ACV, which can be derivatized or protected, is reacted with a compound of formula III, X-S-(CH2)n-0H, wherein X represents the radical C(=Z)-Y or S-U, wherein Z is O or S and
Y and U are alkyl, aryl or saccharide moieties that may be optionally substituted, in particular with an OH, SH or NH2 group, and suitably protected, in order to obtain the compound of formula I in a protected form that can then be deprotected.
In another embodiment, the ACV moiety of compound II
< is reacted with a phosphitylating reagent, then subjected to an oxidation reaction, and finally deprotected, in order to obtain a compound of formula I. An exemplary process for the preparation of the compounds of the present invention is illustrated in the following detailed description, in which other characteristics and advantages of the present invention are also discussed.
The compounds of the present invention can be included in antiviral pharmaceutical compositions that include a phosphotriester compound of the invention as an active substance together with a pharmaceutically acceptable diluent, excipient or carrier.
The present invention is further directed to antiviral compositions of increased therapeutic activity, for example, anti-HBV, anti-HIV, anti-CMV and anti-VZV compositions, together with other antiviral compositions directed against other viruses in which thymidine kinase activity is either decreased or absent. Other characteristics and advantages of the invention will be evident from a review of the detailed examples and descriptions that follow.
Compounds of formula la may be prepared in which the bioreversible protecting group, a substrate for cellular carboxyesterases, is an S-acylthioalkyl group. Acyclovir (preferably suitably protected at position 2) is coupled with an appropriate phosphitylating reagent. Deprotection yields compounds of formula la in which the R group can be, independently, alkyl, aryl, heterocyclyl, sugar, or another similar group. The following examples and procedures illustrate the present invention and are not intended to limit the same. I . SYNTHESIS
A. General Condition*
Thin layer chro atography was performed on Merck 6OF 254 silica plates (Art. 5554) . Column chromatography on silica gel were carried out with Merck 60 H silica (Art. 7719) or with RP2 Merck silanized silica (Art. 7719) .
Before analysis or lyophilization, solutions were filtered on Millex HV-4 filter (Millipore) .
UV spectra were recorded on a UVIKON 810 spectrophotometer.
Mass spectra were obtained using a JEOL JMS DX 300 apparatus by the FAB ionization method, in positive or negative mode, in a matrix of glycerol (G) , glycerol/thioglycerol (GT) or 3-nitrobenzyl alcohol (NBA) . Proton NMR spectra were recorded on a Varian EM 360 apparatus or on a Bruker AC 250 apparatus. The chemical shifts are expressed in ppm relative to the tetra ethylsilane (TMS) signal. The multiplicity and the appearance of the signals observed by NMR are indicated by one or more letter(s) : s(singlet), d(doublet), t(triplet), m(multiplet) , orb (broad). Phosphorus NMR spectra were recorded on a Bruker WP 200 SY apparatus with proton decoupling. The chemical shifts are expressed in ppm relative to the H3P04 signal, which is taken as the external reference.
B. Synthetic Scheme
Figure I depicts an exemplary synthetic scheme for preparing certain compositions in accordance with the invention. Bis-S-acetylthioethyl, BIS(SATE), and bis-S- pivaloylthioethyl, BIS(SPTE), phosphotriester derivatives of acyclovir are shown. These compounds correspond to formula la wherein R is CH3 and (CH3)3C.
EXAMPLE 1
Synthesis of 2,2-Dimethylthiopropanoic acid, _.
Hydrogen sulfide was bubbled, with stirring, for 1.5 hours into anhydrous pyridine (200 mL) maintained at -30 °C. Pivaloyl chloride (62 mL, 0.5 mol) was added dropwise, over 1 hour, to the resulting solution while maintaining the reaction mixture at -30°C. Sulfuric acid (5 N, 425 mL) was slowly added in order to obtain a solution Ph of 5. The two layers were separated and the organic layer was diluted with ether, dried over sodium sulfate, filtered and concentrated under reduced pressure. The residue was then evaporated under reduced pressure to afford compound _. (47 g, 80%) .
Η NMR (CDC13) , δ: 3.69 (s, 1H, SH) ; 1.27 (s, 9H, (CH3)3).
Mass spectra (matrix = NBA), FAB negative: 117 (M-H) . Boiling point: 64°C (92 mm Hg) .
EXAMPLE 2
Synthesis of S-Acetylthioβthanol or S-2- hydroxyethylthioacetatβ, l,8-Diazabicyclo-(5,4,0)undec-7-ene (DBU) (33.7 mL, 0.23 ol) and iodoethanol (15.6 L, 0.20 mol) were consecutively added at 0°C to a stirred solution of commercial thioacetic acid (16.5 L, 0.23 mol) in toluene (80 mL) . The reaction mixture was allowed to react for two hours at room temperature, then diluted with methylene chloride and washed with water. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude residue 3_ (15.3 g, 64%) was used in the following condensation step without further purification.
*H NMR (DMSO-d , δ: 4.95 (t, 1H, CH2Ofl, J - 5.5 Hz); 3.45 (m, 2H, CH-jCifcOH) ; 2.90 (t, 2H, SC&CH-j, J - 6.6 Hz); 2.31 (S, 3H, CHj) .
EXAMPLE 3 Synthesis of s-pivaloylthioethanol or 8- (2,2- dimethylpropanoyl) thioethanol or 8-2-hydroxyβthylthio(2,2- dimethyDpropionate, ± l,8-Diazabicyclo-(5,4,0)undec-7-ene (DBU) (4.2 mL, 29 mmol) and iodoethanol (1.95 Ml, 25 mmol) were added consecutively, at 0βC, to a stirred solution of 2,2- - 8 - di ethylthiopropanoic acid, 2. (3.43 g, 29 mmol) in toluene (12 Ml) . The reaction mixture was allowed to stand for two hours at room temperature, then diluted with methylene chloride and washed with water. The organic layer was dried over sodium sulfate, filtered and concentrated under reduced pressure. The crude residue was dissolved in a small amount of methylene chloride and chromatographed on a silica gel column using a stepwise gradient of methanol (0-2%) in methylene chloride to give the title compound 4. (3.6 g, 89%). »H NMR (DMSO-dβ) , δ : 4.94 (t, 1H, CH20H, J - 5.5 Hz);
3.45 (m, 2H, CH2Cfl2θH) ; 2.88 (t, 2H, SC&CH-j, J - 6.6 Hz) ; 1.16 (s, 9H, (CH3)3)C).
EXAMPLE 4
Synthesis of 0,0'-Bis(8-acetylthioethyl)N,N-diisopropyl- phosphoramidite, £
A solution of S-acetylthioethanol, 3_ (4.81 g, 40 mmol) and triethylamine (6.1 mL, 44 mmol) in THF (100 mL) , cooled at -78βC, was added over a period of 45 minutes to a solution of N,N-diisopropylphosphorodichloridite (4.04 g, 20 mmol) in THF (150 mL) . The reaction mixture was allowed to stand for two hours at room temperature, then filtered and evaporated under reduced pressure. The residue was dissolved in cyclohexane, filtered and evaporated under reduced pressure. The resulting pale yellow oil was dissolved in a small amount of cyclohexane containing 1% of triethylamine and chromatographed on a silica gel column that contained cyclohexane having 5% of triethylamine. The column was eluted with ethylacetate (0-2%) in cyclohexane containing 1% of triethylamine to yield the title compound, 5 (5.3 g, 72%). *H NMR (DMSO-d , δ : 3.59 (m, 6H, CH20H. or CH) ,* 3.04
(t, 4H, SC H2CH2, J - 6.4 HZ); 2.32 (s, 6H, CH3C0) ; 1.10 (d, 12H, (CHj)2CH, J = 6.8 Hz).
31P NMR (DMSO-d , δ : 147.9.
Mass spectra (matrix GT) , FAB positive: 370 (M+H)+; 103 (CH3C0SCH2CH2)+. EXAMPLE 5 synthesis of θ,θ-Bis(β-(2,2-dimethylpropanoyl)thioethyl)N,N- diisopropylphosphora idite, £
A solution of S-(2 ,2-dimethylpropanoyl)thioethanol, 4. (5 g, 30.6 mmol) and triethylamine (4.7 mL, 33.7 mmol) in THF
(60 mL) , cooled at -78°C, was added, over a period of 15 minutes, to a solution of N,N-diisopropylphosphorodichloridite
(3.09 g, 15.3 mmol) in THF (130 mL) . The reaction mixture was allowed to stand for two hours at room temperature, then filtered and evaporated under reduced pressure. The residue was dissolved in cyclohexane, filtered and evaporated under reduced pressure. The resulting pale yellow oil was dissolved in a small amount of cyclohexane containing 1% of triethylamine, and chromatographed on a silica gel column containing cyclohexane having 5% of triethylamine. The column was eluted with ethylacetate (0-2%) in cyclohexane containing
1% of triethylamine to yield the title compound (3.2 g, 46%) . lH NMR (DMSO-d , δ : 3.59 (m, 6H, CH20H. or CH) ; 3.01 (t, 4H, SC IfcCHj, J =■= 6.3 Hz); 1.16 (s, 18H, (CH3)3C) ; 1.10 (d, 12H, (CH jCH, J = 6.8 Hz) .
31P NMR (DMSO-d , δ : 148.0.
Mass spectra (matrix GT) , FAB positive: 454 (M+H)+: 145 ((CH3)3CCOSCH2CH2)+; 85 ((CH3)3CCO)+; 57 ( (CH3)3C)+.
EXAMPLE 6 Synthesisof0-[l-(Methyl-(9-<N2-p-anisyldiphenylmethyDguanin- yl))l-hydroxyβthyl-2-yl]-0', O',-bis(8-acβtyl-2-thioβthyl)- phosphate, 2
Tetrazole (210 mg, 3.0 mmol) was added to a stirred solution of N2-(p-anisyldiphenylmethyl)-9-[ (-2-hydroxy- ethoxy)methyl] guanine (500 mg, 1 mmol) , prepared in accordance with Martin et al . t J. Med . Chem . , 1986, 29 , 1384-1386, and compound 5 (445 mg, 1.2 mmol) in THF (3.0 mL) . After 35 minutes at room temperature, the reaction mixture was cooled to -40βc and a solution of 3-chloroperbenzoic acid (407 mg, 1.3 mmol) in methylene chloride (5 mL) was added. The solution was then allowed to warm up to room temperature over one hour. Sodium hydrogen sulfite (10% solution, 3 mL) was added to reduce excess peracid. The organic layer was separated, diluted with methylene chloride (10 mL) , washed with a saturated solution of aqueous sodium bicarbonate (3 mL) , then with water (3x3 mL) , dried over Na2S04 and evaporated to dryness. The residue was dissolved in a small amount of methylene chloride and chromatographed on a silica gel column (methanol (0-4%) in methylene chloride) . Compound 2 was obtained as a white foam (531 mg, 68%) . fH NMR (DMSO-d , δ : 10.63 (s, 1H, NH) ; 7.71 (s, 1H,
NH) ; 7.67 (s, 1H, H-8) ; 7.16-7.31 (m, 12H, aromatic); 6.87 (d, 2H, aromatic, J = 8.9 Hz); 4.86 (s, 2H, NCH20) ; 3.97 (m, 4H, OC CHjS) ; 3.71 (s, 5H, 0CH3 or POC CH ) ; 3.10 (t, 4H, OCH-jCHjS, J = 6.3 Hz); 3.03 (m, 2H, POCH2,CH20); 2.35 (s, 6H,CH3COS). 3lP NMR (DMSO-d , δ : -0.83.
Mass spectra (matrix GT) , FAB positive: 782 (M+M)+; 273 (trityl)+; 103 (CH3C0SCH2CH2)+; FAB negative: 780 (M-H)'; 678 (M-CH3C0SCH2CH2)'.
EXAMPLE 7 Synthesisof0-[l-(Methyl-[9-<N2-p-anisyldiphenylmethyl)guanin- yl])l-hydroxyethyl-2-yl]-0', 0"-bis (S-pivaloyl-2-thioβthyl)- phosphate, a.
Tetrazole (126 mg, 1.8 mmol) was added to a stirred solution of N2-(p-anisyldiphenylmethyl)-9-[ (-2-hydroxy- ethoxy) ethyl] guanine (300 mg, 0.60 mmol), prepared in accordance with Martin et al . , J. Med. chem . , 1986, 29 , 1384- 1386, and compound £ (408 mg, 0.90 mmol) in THF (1.8 mL) . After 35 minutes at room temperature, the reaction mixture was cooled to -40°C and a solution of 3-chloroperbenzoic acid (342 mg, 0.99 mmol) in methylene chloride (4 mL) was added. The solution was then allowed to warm up to room temperature over one hour. Sodium hydrogen sulfite (10% solution, 3 mL) was added in order to reduce excess peracid. The organic layer was separated, diluted with methylene chloride (10 mL) , washed with a saturated solution of aqueous sodium bicarbonate (3 mL) , then with water (3x3 mL) , dried over Na2S04 and evaporated to dryness. The residue was dissolved in a small amount of methylene chloride and chromatographed on a silica gel column (methanol (0-4%) in methylene chloride) to give compound 8_ as a white foam (372 mg, 71%) . *H NMR (DMSO-d , δ : 10.63 (s, 1H, NH) ; 7.71 (s, 1H,
NH) ; 7.68 (s, 1H, H-8); 7.17-7.30 (m, 12H, aromatic); 6.86 (d, 2H, aromatic, J - 8.9 Hz); 4.87 (s, 2H, NCH20) ; 3.97 (m, 4H, OCHjCHjS) ; 3.71 (Si, 5H, OCH3 or POCH2CH20) ; 3.09 (t, 4H, 0CH2CH2S, J = 6.4 Hz); 3.06 (m, 2H,
Figure imgf000013_0001
1.17 (s, 18H, (CH3)3C).
31P NMR (DMSO-dβ) , δ : -0.88.
Mass spectra (matrix GT) , FAB positive: 866 (M+M)+; 273 (trityl)+; 145 ( (CH3)3CCOSHC2CH2) +; FAB negative: 864 (M-H)"; 720 (M-(CH3)3CCOSCH2CH2)-.
EXAMPLE 8
Synthesis of0-[l-(Methyl-[9-guanin-yl])-l-hydroxyethyl-2-yl]- O', 0"-bis(S-acetyl-2-thioethyl) phosphate, 2 [Bis(SATE)ACVMP]
A solution of compound 2 (450 mg, 0.57 mmol), in a mixture of acetic acid (32 mL) , methanol (4 mL) and water (4 mL) , was heated at 50°c for 18 hours, then evaporated to dryness. The residue was coevaporated three times with ethanol, twice with methylene chloride, then diluted in a small amount of methylene chloride and chromatographed on a silica gel column (methanol (0-10%) in methylene chloride) . The desired product, compound 9_, was obtained as a white foam (270 mg, 92%) .
UV: λ max (EtOH 95) 253 nm (e 14100). Η NMR (DMSO-dβ) , δ : 10.65 (s, 1H, NH) ; 7.81 (s, 1H, H-8); 6.52 (S, 2H, NH2) ; 5.35 (s, 2H, NCH20) ; 4.05 (m, 2H, POCHCHjO) ; 3.99 (m, 4H, OCH^C^S) ; 3.65 (m, 2H, POCH^C&O); 3.09 (t, 4H, OCH-jC S, J = 6.3 Hz) ; 2.34 (s, 6H, CH3C0S) . 31P NMR (DMSO-d , δ : -0.74.
Mass spectra (matrix GT) , FAB positive: 510 (M+M)+; 152 (BH2)+; 103 (CH3COSCH2CH2)+; FAB negative: 508 (M-H)"; 406 (M- (CH3COSCH2CH2)-; 150 (B)\ - 12 -
EXAMPLE 9
Synthesisof0-[l-(Methyl-[ -guanin-yl])-l-hydroxyethyl-2-yl]- O', 0"-bis(S-pivaloyl-2-thioethyl) phosphate, 10 [Bis(SPTE)ACVMP] A solution of compound 8. (360 mg, 0.42 mmol) , in a mixture of acetic acid (24 mL) , methanol (3 mL) and water (3 mL) , was heated at 50°C for 18 hours, then evaporated to dryness. The residue was coevaporated three times with ethanol, twice with methylene chloride, then diluted in a small amount of methylene chloride and chromatographed on a silica gel column (methanol (0-9%) in methylene chloride) . The desired product, compound _J_, was obtained as a white foam (210 mg, 85%) .
UV: λ max (EtOH 95) 253 nm (e 13400). lH NMR (DMSO-d , δ : 10.59 (s, 1H, NH) ; 7.79 (s, 1H,
H-8); 6.46 (s, 2H, NH2) ; 5.36 (s, 2H, NCH20) ; 3.95-4.09 (m, 6H,
Figure imgf000014_0001
or OCHjCHjS) ; 3.67 (m, 2H, POCH2CH2θ) ; 3.08 (t, 4H, OCH2,CH2S, J - 6.3 HZ); 1.17 (s, 18H, (CH3)3C) . 3,P NMR (DMSO-d , δ : -0.80. Mass spectra (matrix GT) , FAB positive: 594 (M+H)+;
152 (BH2)+; 145 ((CH3)3CCOSCH2CH2)+; FAB negative: 592 (M-H)"; 448 (M-(CH3)3CCOSCH2CH2)-; 150 (B)\
II. BIOLOGICAL EVALUATION
The experimental procedure used for evaluation of activity against hepatitis B virus was as previously described by Korba and Milman in j ntiviral Res . , 1991, 217 , 217.
The experimental procedure used for evaluation of activity against human immunodeficiency virus (HIV) was as follows: HIV-1 replication (LAI isolate) in CEM cells was measured by assaying for reverse transcriptase (RT) in the culture supernatant after infection for five days. This activity reflects the presence of the virus released by the cells. After absorption of the virus, the test compounds were added at various concentrations in the culture medium. Antiviral activity was expressed as the lowest concentration of compound which reduced the viral production by at least 50% (EDjo) . The toxic effect in non-infected CEM cells was assessed by a calorimetric reaction based on the capacity of living cells to reduce 3-(4,5 dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide into formazan after incubation for five days in the presence of various concentrations of the compounds. The results were expressed as the lowest concentration of compound which resulted in at least 50% inhibition of the formation of formazan (CD*,) . The experimental procedure used for evaluation of activities against herpes simplex virus type 1 (HSV-1) was as described by Genu-Dellac et al . in N cleosides and Nucleotides, 1991, 10 , 1345.
The experimental procedure used for the evaluation of compounds of the present invention against herpes simplex virus type 2 (HSV-2) involved the treatment of HSV-2 vaginitis in BALB/c mice, and is as follows:
BALB/c female mice, weighing approximately 20 g each at the start of the experiment, were intravaginally infected with HSV-2 (E194 strain) . This was accomplished in a 3-step process. First, the vagina of each mouse was swabbed for 5 seconds with a cotton-tipped applicator dipped in 0.1 N NaOH. Approximately 30-60 minutes later each vagina was dry swabbed for 5 seconds. Then an applicator dipped in virus medium (containing approximately 107 plaque forming units of virus/mL) was used to swab each mouse for 20 seconds. Twenty-four hours after infection with virus, compounds were administered by i.p. injection. Treatments were given twice daily for 5 consecutive days. Lesion scores in infected mice were determined daily on days 3-10 of the infection. A score of 1+ to 4+ was given depending on the progression of lesion formation. Deaths were recorded daily for 21 days. Vaginal virus titers were made by titration of virus obtained from vaginal swabs (placed in 1 mL of cell culture medium) 3 days after virus inoculation. These titrations were conducted in MA-104 African green monkey kidney cells in 96-well plates. Calculation of virus titer was made by the 50% endpoint dilution method of Reed and Muench (Am. J. Hyg. , 1938, 27, 493). Statistical interpretation of survival (Fisher exact test) , mean day to death (Mann-Whitney U-test) and virus titer (Mann-Whitney U-test) were made by two-tailed analyses using the Instat™ computer program (GraphPad, Inc., San Diego, CA) .
The compounds of the present invention were also assayed for antiviral activity against murine gammaherpes virus type 68 (MHV-68) using a plaque reduction assay. MHV-68 is a mouse counterpart virus to EBV which infects mice, and may be used as a rodent model for studying anti-EBV compounds. The procedure for this assay was as follows:
Compounds were prepared as a 10 mM solution in DMSO, which was then diluted below 100 μM in cell culture medium. Acyclovir was initially prepared as a 4 mM solution in cell culture medium before diluting to micromolar concentrations. African green monkey kidney (Vero) cells were seeded into 12- well plates, then allowed to grow to confluency. The cells were infected with approximately 100 plaque forming units of MHV-68. After 1.5 hours, the medium was changed to include antiviral drug or drug-free medium (MEM, 2% FBS and 0.5% SeaPlaque agarose obtained from FMC Corp. , Rockland, MD) . At 7 days the plates were fixed for 15 minutes with 10% buffered formalin prior to removing the agar overlay. The cells were then stained with 0.2% crystal violet in 20% methanol. The plaques were counted at 17x magnification using a Plaque Viewer (Bellco Glass Co. , Vineland, NJ) . ECy-, values were determined by plotting inhibition versus drug concentration.
The compounds of the present invention were tested for their antiviral activity against human CMV using a plaque reduction assay. The procedure was as follows:
The antiviral activity of the compounds against human CMV was determined in MRC-5 cells (a continuous line of diploid, human (male) embryonic lung cells) . The compounds were diluted in MEM, 2% FBS, 0.1% NaHC03 and 50 μg/mL of gentamicin. The growth medium was decanted from established onolayers of MRC-5 cells in 24-well tissue culture plates. Human CMV (1 mL) , diluted in test medium, was placed in each well except those to be used for cell controls, which received 1 mL of sterile test medium. The virus was allowed to adsorb to the cells while the plates were centrifuged (2200 rpm, 30 minutes) at room temperature. The medium was aspirated from the plates and 0.8 mL of the compound (varying concentrations) was placed in each well. Test medium (0.8 mL) without any compound was added to each cell control and virus control well. The plates were placed in an incubator at 37°C in a moist atmosphere of 5% C02, 95% air until plaques could be distinguished in the virus control wells. The cells were observed microscopically for morphological changes due to compound cytotoxicity before the medium was aspirated from the wells. The cells were stained with crystal violet for 15 minutes, after which the staining solution was aspirated, the plates rinsed with water, inverted and dried at room temperature. Plaques were counted by the use of a dissecting microscope. EDJQ and CDM values were determined by regression analysis of the viral plaque data and the visual cell toxicity data, respectively. The antiviral activity of the compounds of the present invention against murine CMV (MCMV) was determined by a CPE (cytopathic effect) inhibition assay, which was performed as follows:
Growth medium was decanted from 96-well tissue culture plates containing monolayers of 3T3 cells (a continuous line of contact-inhibited mouse fibroblasts) . Varying concentrations of compound were added to the wells (4 wells/dilution, 0.1 mL/well of each compound). Compound diluent medium (MEM, without serum, 0.18% NaHC03, 50 μg of gentamicin/mL) was added to the cell and virus control wells
(0.1 mL/well). Virus, diluted in the test medium, was added to all the compound test wells of the plate (3 wells/dilution) and to virus control wells at 0.1 mL/well. Test medium without virus was added to all toxicity control wells ( 1 well/dilution) and to cell control wells at 0.1 mL/well. The plates were incubated at 37°C in a humidified incubator with 5% C02, 95% air until virus control wells had adequate CPE readings. Cells in test and virus control wells were then examined microscopically and virus CPE was graded on a scale of 0-4, with 0 being no CPE and 4 being 100% CPE. The cells in toxicity control wells were observed microscopically and graded for morphological changes due to cytotoxicity. EDM and CDJO values were determined by regression analysis of the viral CPE data and the visual toxicity data, respectively.
The compounds of the present invention were also assayed for their activity against varicella zoster virus (VZV) using a plaque reduction assay. The procedure was as described for the assay against human CMV, except that 0.5 mL of VZV was added to each well, and the virus was allowed to adsorb to the cells during a 1 hour stationary incubation period.
The compounds of the present invention were designed to be stable in biological fluids and various organs, thus allowing them to reach targeted cells. The significant stability of bis(SPTE)ACVMP in human serum was demonstrated by its half-life of 14 hours. The ex vivo procedure for half-life determination was as follows: Decomposition of the compound was studied at 37°C in human serum. During incubation, aliquots (80 μL) were removed and injected into a cleaning precolumn (Guard-Pak, Waters) which was equilibrated with triethylammonium acetate buffer (20 mM, pH 6.6). Protein and other undesired components were allowed to elute, while compounds to be analyzed were trapped. After 5 minutes, the precolumn was connected to an analytical column (Hypersil ODS, 100x4.6 mm, 3 μM particle size, Shandon) . The trapped analytes were transferred from the precolumn to the analytical column and eluted with 40 mM triethylammonium acetate:acetonitrile (1:1, v/v) . The structure of eluted analytes was confirmed by HPLC-UV-MS (a Waters HPLC module and a UV-diode array detector 996 were connected by two automated 7010 Rheodyne valves and controlled by a Millenium software program (Waters) . The mass spectrometer (SSQ 7000, Finnigan Mat) was directly coupled to the UV detector outlet) . RESULTS:
Anti-HBV activity, in transfected HepG2 (2.2.15) cells, of bis(S-acyl-2-thioethyl)phosphotriester derivatives of acyclovir, (compound 9_, Figure I) , as compared with those of the parent nucleoside and of 2',3'-dideoxyguanosine (ddG) and 2',3'-dideoxycytidine (ddC) are shown in Table 1.
TABLE 1
HBV virion
Composition
EC50(μM) ECw(μM) CCjo(μM) SI
ddC 1.3 ± 0.2 1.1 ± 1.2 219 ± 19 20
1 ddC 2.2 ± 0.3 8.3 ± 0.8 218 ± 19 30
Bis(SATE)ACVMP 0.7 ± 0.1 5.1 + 1 987 ± 99 194 2.
Bis(SPTE)ACVMP 0.2±0.04 7.1 ± 0.8 1593 ± 131 224 1G
Acyclovir 111 ± 15 >100 631 ± 40 ND
SI = CC^/EC,,,; ND = Not Determinable
In Table 1, EC^ and ECW represent the molar concentrations which provide 90% inhibition and 50% inhibition, respectively, of HBV replication. CC^ represents the molar concentration which reduces the viability of non-infected cells by 50%. The data show that both acyclovir derivatives significantly inhibit HBV activity.
Synergistic anti-HBV activity was observed upon treatment with Bis(SATE)ACVMP (9_) and 3TC in combination. The results are shown in Table 2 (SI - CCso/EC^) . TABLE 2
I Composition ECjo (μM) EC*, (μM) CCJO (μM) SI
Bis(SATE)ACVMP + 0.03 0.19 913 4805 3TC
Acyclovir + 3TC 6.0 20 644 32
Analysis of the results shown in Table 2 by the
COMBOSTAT combination analysis program indicated that there was synergy between bis(SATE)ACVMP and 3TC at EC50 and EC,,-,. In this combination treatment, bis(SATE)ACVMP was mixed with 3TC at a 10:1 molar ratio.
Anti-HIV activity, in CEM TK- cells, of bis(S-acyl-2- thioethyl) phosphotriesters derivatives of acyclovir, as compared with those of the parent nucleoside and of AZT are shown in Table 3.
TABLE 3
Composition EC5o(μM) CCjo(μM)
AZT >100(20%) >100(5%)
Bis(SATE)ACVMP 77 >100(0%)
Bis(SPTE)ACVMP 3.6 >10(26%)
I Acyclovir >100(0%) >100(0%)
In Table 3, EC*, represents the molar concentration which produces a 50% inhibition of HIV replication. Significant inhibition of HIV replication was observed.
Anti-HSV-1 activity, in MRC-5 cells, of bis(S-acyl-2- thioethyl)phosphotriesters derivatives of acyclovir is compared to that of acyclovir in Table 4. TABLE 4
Composition HSV-1 HSV-1 TK-
Figure imgf000021_0001
Bis(SATE)ACVMP 1.25 100
Bis(SPTE)ACVMP 2.5 10
Acyclovir 0.63 >100
In Table 4, ECW represents the molar concentration which produces a 50% inhibition of HSV-1 replication. Significant activity against HSV was observed.
Anti-HSV-2 activity of bis(SATE)ACVMP in BALB/c mice is demonstrated by the data shown in Tables 5 and 6, and Figures 2 and 3.
Table 5 shows the survival and vaginal virus titer data in mice infected with HSV-2. Compared to acyclovir, bis(SPTE)ACVMP elicited a superior response with 60% survival in the 100 and 50 mg/kg/day groups of bis(SPTE)ACVMP. Slightly less virus was recovered from lesions of bis(SPTE)ACVMP-treated mice compared to acyclovir-treated mice, however the differences were not significant.
Table 6 shows average daily lesion scores. Bis(SPTE)ACVMP, at 100 mg/kg/day produced a good response in lesion score reduction over the entire time frame. The data in Table 6 is also depicted graphically in Figures 2 and 3. It is clear from these figures that bis(SPTE)ACVMP is more effective in inhibiting lesion formation than acyclovir. The results discussed above indicate that bis(SPTE)ACVMP was effective against HSV-2 vaginal infection in mice. Acyclovir was effective at the outset, but its effect waned after the discontinuation of treatment. Bis(SPTE)ACVMP, at 100 and 50 mg/kg/day, reduced mortality, vaginal virus titers and vaginal lesion scores in HSV-2 vaginally infected mice when treatments were given twice daily for 5 days starting 1 day after virus inoculation. TABLE 5
Effects of Acyclovir and Bis(SPTE) CVMP on Survival and Vaginal virus Titers in Hβv-lnfected Mice
Figure imgf000022_0001
Compound Dose1 Survivors/ Mean Day to Vaginal Virus (mg/Jcg/day) Total (%) Dβathb Titerse
Bis(SPTE)ACVMP 100 6/10 (60)* 12.5 ± 2.4** 2.5_ 0.7***
50 6/10 (60)* 11.7 ± 2.1* 2.8 ± 1.2***
25 4/10 (40) 10.7 ± 0.5** 2.3 + 0.8***
Acyclovir 100 3/8 (38) 16.2 ± 2.4d*** 3.2 + 1.04***
50 5/8 (63)** 12.0 ± 2.0* 2.3 ± 0.8***
25 1/10 (10) 11.0 ± 1.9** 3.0 + 0.7***
Placebo 0/10 (0) 7.8 + 1.6 5.3 ± 0.4
Intraperitoneal treatments were given twice daily for 5 days starting day 1 after virus infection. 10 b Of the mice that died (survivors lived through 21 days) . c Logio CCID50/lesion, determined 3 days after virus challenge. d Standard deviation.
* P<0.05 ** P<0.01 *** P<0.001
Figure imgf000023_0001
- 22 -
Antiviral activity against MHV-68 is exhibited by the data shown in Table 7. In the plaque reduction assay, the ECW for bis(SPTE)ACVMP was 8 μM and the ECM for acyclovir was 5 μM.
TABLE 7
Compound Cone. (μM) % Plaques*
Bis(SPTE)ACVMP 40 1
20 15
10 34
5 76
2.5 100
1.25 100
0 100
Acyclovir 40 0
20 6
10 22
5 54
2.5 82
1.25 100
0 100
Relative to the wells devoid of antiviral compound.
Antiviral activity of the compounds of the present invention against human CMV are shown in Table 8. Bis(SPTE)ACVMP was approximately 4-fold more effective in reducing plaques than was acyclovir.
Antiviral activity of the compounds of the present invention against murine CMV as determined by a CPE (cytopathic effect) inhibition assay is shown in Table 9. The data indicates that both bis(SATE)ACVMP and bis(SPTE) CVMP were active against murine CMV, with the latter compound being slightly more active. Antiviral activity of the compounds of the present invention against varicella zoster virus (VZV) is shown in Table 10. The data indicates that both bis(SATE)ACVMP and bis(SPTE)ACVMP were as active as acyclovir in the plaque reduction assay.
TABLE 8
Figure imgf000026_0001
Antiviral Activity (Plaque Reduction) of UA514, UA491 , UA486, or DHPG vs Human Cytomegalovirus, Strain AD-
169, In MRC-5 Cells
.UA514 UA491 UA486 DHPG
UA514. 491. & 486 Number Number Numbe ir Number DHPG
Conc'n. of % of % of % of % Concentration
(μM) Plaques Reduction Plaques Reduction Plaques Reduction Plaques Reduction (ug/mll
100 0.0 100 28.5 31 12.0 71 0.0 100 100
32 3.0 93 42.5 0 50.5 0 0.0 100 32
10 32.5 0 49.0 0 43.0 0 8.5 80 10
3.2 42.5 0 40.0 0 62.0 0 30.5 27 3.2
1.0 32.5 0 32.0 0 36.5 0 37.0 0 1.0
0.32 35.5 0 33.0 0 37.0 0 37.0 0 0.32
0.1 40.5 0 40.0 0 28.5 31 44.0 0 0.1
ED503: 18 μM >100 μM 71 μM 5.2 μg/ml
CD50b: > 100 μM >100 μM > 100 μM >100 μg/ml
Tlc: >5.6 N/D* >1.4 > 19 a The concentration at which the average number of plaques is reduced to 50% of that seen in the virus controls (Effective
Dose, 50% endpoint). b The concentration halfway between those at which 100% and 0% cytotoxicity are seen. c Therapeutic Index (CD50+ED50).
* Not Determinable UA514 is Bis(SPTE)ACVMP.
UA491 is Bis(SATE)ACVMP. UA486 is Acyclovir. DHPG is Ganciclovir.
TABLE 9
Antiviral Activity (CPE Inhibition) of UA514, UA491 , UA486, or Acyclovir vs. Murine
Cytomegalovirus, Strain Smith MSGV, In 3T3 Cells
UA514, 491, & 486 UA5 14 UA491 UA486
Conc'n. Visible Avg. Visible Avg. Visible Avg.
(μM) Cyotox, GEE CytQtox, CPE Cvtotox. CPE
100 0 0.5 0 0.5 0 0.0
32 0 0.3 0 0.5 0 0.0
10 0 0.5 0 2.5 0 0.5
3.2 0 4.0 0 4.0 0 1.8
1.0 0 4.0 0 4.0 0 4.0
0.32 0 4.0 0 4.0 0 4.0
0.1 0 4.0 0 4.0 0 4.0
4.0 (virus controls)
ED50a: 6.1 μM 12 μM 2.8 μM
CD50&: >100 μM >100 μM >100 μM
Tic; > 16 >8.3 >36 a The concentration at which the average viral CPE is reduced to 50% of that seen in the virus controls
(Effective Dose, 50% endpoint). b The concentration halfway between those at which 100% and 0% cytotoxicity are seen. c Therapeutic Index (CD50+ED50).
CPE = Cytopathic Effect UA514 is Bis(SPTE)ACVMP.
UA491 is Bis(SATE)ACVMP. UA486 is Acyclovir.
Figure imgf000028_0001

Claims

What is claimed is:
1. A phosphotriester compound of the structure: (RO)2-P(=0)-0-ACV wherein: R is a radical (CH2)β-S-X, X is C(=Z)-Y or S-U; Z is O or S;
Y and U are alkyl, aryl or a saccharide which is optionally substituted with OH, SH or NH2; n is equal to 1 to 4, and ACV is an acyclovir moiety.
2. The compound of claim 1 wherein the acyclovir moiety has the structure:
Figure imgf000029_0001
3. The compound of claim 1 wherein: X is S-U and U represents the radical (CH2)nl-X1;
X1 is H, OH, SH or NH2; and n1 is 1 to 4.
4. The compound of claim l wherein:
X is C(=Z)-Y; and Y is CH3 or (CH3)3-C.
5. The compound of claim 4 wherein:
R is (CH2)n-S-C(=0)-CH3or (CH2)n-S-C(=0)-tBu; and n is 1 or 2.
6. The compound of claim 3 wherein R is (CH2)2-S-S-(CH2)2-OH.
7. A compound having the structure:
Figure imgf000030_0001
wherein R is CH3 or (CH3)3C.
8. A pharmaceutical composition comprising the compound of claim 1 as an active ingredient and a pharmaceutically acceptable diluent, carrier or excipient.
9. The pharmaceutical composition of claim 8 exhibiting inhibitory activity against a virus whose thymidine kinase activity is either decreased or absent.
10. The pharmaceutical composition of claim 8 exhibiting inhibitory activity against human immunodeficiency virus, hepatitis B virus, herpes simplex virus type 1, herpes simplex virus type 2, or cytomegalovirus.
11. The pharmaceutical composition of claim 8 exhibiting inhibitory activity against varicella zoster virus.
12. An pharmaceutical composition comprising a compound of claim 7 as an active ingredient and a pharmaceutically acceptable excipient, diluent or carrier.
13. The pharmaceutical composition of claim 12 exhibiting inhibitory activity against a virus whose thymidine kinase activity is either decreased or absent.
14. The pharmaceutical composition of claim 12 exhibiting inhibitory activity against human immunodeficiency virus, hepatitis B virus, herpes simplex virus type 1, herpes simplex virus type 2, or cytomegalovirus.
15. The pharmaceutical composition of claim 12 exhibiting inhibitory activity against varicella zoster virus.
16. The compound of claim 1 wherein n is 1 or 2.
PCT/IB1996/000454 1995-04-21 1996-04-18 Acyclovir derivatives as antiviral agents WO1996033201A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU55111/96A AU5511196A (en) 1995-04-21 1996-04-18 Acyclovir derivatives as antiviral agents

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
FR95/04797 1995-04-21
FR9504797A FR2733234B1 (en) 1995-04-21 1995-04-21 DERIVATIVES OF ACYCLOVIR AS ANTIVIRAL AGENTS
US08/465,450 1995-06-05
US08/465,450 US5627185A (en) 1994-11-23 1995-06-05 Acyclovir derivatives as antiviral agents

Publications (1)

Publication Number Publication Date
WO1996033201A1 true WO1996033201A1 (en) 1996-10-24

Family

ID=26231907

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB1996/000454 WO1996033201A1 (en) 1995-04-21 1996-04-18 Acyclovir derivatives as antiviral agents

Country Status (2)

Country Link
AU (1) AU5511196A (en)
WO (1) WO1996033201A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997040056A1 (en) * 1996-04-22 1997-10-30 Drug Innovation & Design, Inc. Prodrugs of acyclovir
WO1999051613A1 (en) * 1998-04-03 1999-10-14 Medivir Ab Prodrugs of phosphorous-containing pharmaceuticals
WO1999054336A1 (en) * 1998-04-22 1999-10-28 Institut National De La Sante Et De La Recherche Medicale (Inserm) (alpha-aminophosphino) peptidesderivative and compositions containing same
US6458772B1 (en) 1909-10-07 2002-10-01 Medivir Ab Prodrugs
US8299293B2 (en) 2007-07-09 2012-10-30 Evonik Degussa Gmbh Process for preparing α-keto acids and derivatives thereof
US9887104B2 (en) 2014-07-03 2018-02-06 Intel Corporation Electronic package and method of connecting a first die to a second die to form an electronic package

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2381781A1 (en) * 1977-02-24 1978-09-22 Wellcome Found GUANINE DERIVATIVES AND MEDICINAL PRODUCTS CONTAINING THESE SUBSTANCES
WO1993024510A1 (en) * 1992-05-25 1993-12-09 Centre National De La Recherche Scientifique (Cnrs) Phosphotriester-type biologically active compounds

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2381781A1 (en) * 1977-02-24 1978-09-22 Wellcome Found GUANINE DERIVATIVES AND MEDICINAL PRODUCTS CONTAINING THESE SUBSTANCES
WO1993024510A1 (en) * 1992-05-25 1993-12-09 Centre National De La Recherche Scientifique (Cnrs) Phosphotriester-type biologically active compounds

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PERIGAUD C ET AL: "Bis(S-acyl-2-thioethyl)esters of 2',3'-dideoxyadenosine 5'-monophosphate are potent anti-HIV agents in cell culture", NUCLEOSIDES NUCLEOTIDES (NUNUD5,07328311);95; VOL.14 (3-5); PP.789-91, UNIV. MONTPELLIER II;LABORATOIRE CHEMIE BIO-ORGANIQUE; MONTPELLIER; 34095; FR. (FR), XP000573956 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6458772B1 (en) 1909-10-07 2002-10-01 Medivir Ab Prodrugs
WO1997040056A1 (en) * 1996-04-22 1997-10-30 Drug Innovation & Design, Inc. Prodrugs of acyclovir
US6974802B2 (en) 1998-02-13 2005-12-13 Medivir Ab Treatment of viral infections using prodrugs of 2′,3-dideoxy,3′-fluoroguanosine
US7071173B2 (en) 1998-02-13 2006-07-04 Medivir Ab Antiviral methods employing double esters of 2′, 3′-dideoxy-3′-fluoroguanosine
WO1999051613A1 (en) * 1998-04-03 1999-10-14 Medivir Ab Prodrugs of phosphorous-containing pharmaceuticals
WO1999054336A1 (en) * 1998-04-22 1999-10-28 Institut National De La Sante Et De La Recherche Medicale (Inserm) (alpha-aminophosphino) peptidesderivative and compositions containing same
FR2777780A1 (en) * 1998-04-22 1999-10-29 Inst Nat Sante Rech Med Alpha amino phosphino peptide derivatives having morphine-like analgesic properties
US6391866B1 (en) 1998-04-22 2002-05-21 Institut National De La Sante Et De La Recherche Medicale (Inserm) (α-aminophosphino) peptides derivative and compositions containing same
US8299293B2 (en) 2007-07-09 2012-10-30 Evonik Degussa Gmbh Process for preparing α-keto acids and derivatives thereof
CN101687753B (en) * 2007-07-09 2013-06-19 赢创德固赛有限责任公司 Method for the production of ketoacids and their derivatives
US9887104B2 (en) 2014-07-03 2018-02-06 Intel Corporation Electronic package and method of connecting a first die to a second die to form an electronic package

Also Published As

Publication number Publication date
AU5511196A (en) 1996-11-07

Similar Documents

Publication Publication Date Title
AU701574B2 (en) Acyclovir derivatives for topical use
Lefebvre et al. Mononucleoside phosphotriester derivatives with S-acyl-2-thioethyl bioreversible phosphate-protecting groups: intracellular delivery of 3'-azido-2', 3'-dideoxythymidine 5'-monophosphate
US5756116A (en) Nucleoside phosphate therapy for viral infection
McGuigan et al. Intracellular delivery of bioactive AZT nucleotides by aryl phosphate derivatives of AZT
Puech et al. Intracellular delivery of nucleoside monophosphates through a reductase-mediated activation process
JP4782365B2 (en) Compositions and methods for dual targeting of viral infections and cancer cells
EP0820461B1 (en) Aryl-ester phosphoramidate derivatives of 2&#39;,3&#39;-didehydronucleosides
US5962684A (en) Method of preparation of dideoxycarbocyclic nucleosides
US5627185A (en) Acyclovir derivatives as antiviral agents
EP0074306B1 (en) Anti-viral guanine derivatives
NO154751B (en) ANALOGUE PROCEDURE FOR THE PREPARATION OF THERAPEUTICALLY EFFECTIVE HYDROXYCARBONYL PHOSPHONIC ACID ESTERS.
Farrow et al. Synthesis and biological properties of novel phosphotriesters: A new approach to the introduction of biologically active nucleotides into cells
EP0642528A1 (en) Phosphotriester-type biologically active compounds.
US5879700A (en) Nucleoside analogue phosphates for topical use
KR20010071673A (en) ARYL PHOSPHATE DERIVATIVES OF d4T HAVING ANTI-HIV ACTIVITY
WO1998038202A1 (en) Antiviral phosphonate prodrugs of nucleosides and nucleoside analogues
WO1996033201A1 (en) Acyclovir derivatives as antiviral agents
Mao et al. Structure-activity studies on phosphonoacetate
Qiu et al. Synthesis and antiviral activity of phosphoralaninate derivatives of methylenecyclopropane analogues of nucleosides
EP0309491A1 (en) Antiviral agents
JPH013197A (en) Antiviral compounds and antiviral agents containing them
CA2040854C (en) Chiral 2-(phosphonomethoxy)propyl guanines as antiviral agents
Burns et al. Novel 6-alkoxypurine 2', 3'-dideoxynucleosides as inhibitors of the cytopathic effect of the human immunodeficiency virus
McGuigan et al. Novel nucleoside phosphoramidates as inhibitors of HIV: studies on the stereochemical requirements of the phosphoramidate amino acid
EP0145739B1 (en) Novel antiviral agents

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BB BG BR BY CA CH CN CZ DE DK EE ES FI GB GE HU IS JP KE KG KP KR KZ LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK TJ TM TR TT UA UG US UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: CA