WO2002000613A2 - Inhibiteurs d'infections par le virus de l'hepatite b (vhb) - Google Patents

Inhibiteurs d'infections par le virus de l'hepatite b (vhb) Download PDF

Info

Publication number
WO2002000613A2
WO2002000613A2 PCT/EP2001/007260 EP0107260W WO0200613A2 WO 2002000613 A2 WO2002000613 A2 WO 2002000613A2 EP 0107260 W EP0107260 W EP 0107260W WO 0200613 A2 WO0200613 A2 WO 0200613A2
Authority
WO
WIPO (PCT)
Prior art keywords
hbv
compound
independently
ghy
hepatitis
Prior art date
Application number
PCT/EP2001/007260
Other languages
English (en)
Other versions
WO2002000613A3 (fr
Inventor
Dorian Bevec
Sabine Obert
Original Assignee
Axxima Pharmaceuticals Ag
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
Application filed by Axxima Pharmaceuticals Ag filed Critical Axxima Pharmaceuticals Ag
Priority to AU2001269092A priority Critical patent/AU2001269092A1/en
Publication of WO2002000613A2 publication Critical patent/WO2002000613A2/fr
Publication of WO2002000613A3 publication Critical patent/WO2002000613A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C281/00Derivatives of carbonic acid containing functional groups covered by groups C07C269/00 - C07C279/00 in which at least one nitrogen atom of these functional groups is further bound to another nitrogen atom not being part of a nitro or nitroso group
    • C07C281/16Compounds containing any of the groups, e.g. aminoguanidine
    • C07C281/18Compounds containing any of the groups, e.g. aminoguanidine the other nitrogen atom being further doubly-bound to a carbon atom, e.g. guanylhydrazones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses

Definitions

  • the present invention relates to a class of guanyl hydrazones discovered to have activity as inhibitors of Hepatitis B Virus (HBV) replication.
  • the inhibitor compounds are suitable as agents for the treatment of HBV infection, including opportunistic infections, in particular chronic HBV infection and diseases caused by HBV infections.
  • the present invention discloses pharmaceutical compositions useful for prophylaxis and/or treatment of HBV infections, including opportunistic infections, and diseases caused by HBV infections.
  • Hepatitis is an inflammation of the liver that is most often caused by infection with one of five known viruses, hepatitis A, B, C, D or E. In many cases of infection, particularly in cases of hepatitis B and hepatitis C infection, "chronic hepatitis" may result. Chronic hepatitis occurs when the body is unable to completely clear the virus even though the symptoms may not persist. Continued presence of the virus over a number of years can lead to cirrhosis (scarring of the liver) or hepatocellular carcinoma (liver cancer).
  • Hepatitis B virus is transmitted through sexual contact, vertical transmission (mother to child at birth) or by coming into contact with contaminated blood. It is estimated that over 2 billion people worldwide have been infected with hepatitis B virus. Of these 2 billion people, approximately 350 million people have developed chronic HBV infection, putting them at high risk of developing cirrhosis and liver cancer (World Health Organization, fact sheet on Hepatitis B, November, 1998). In the United States alone, 140,000 to 320,000 HBV infections are reported every year, and an estimated 1 to 1.25 million Americans are chronically infected with HBV. The estimated costs of medical treatment and work loss is over 700 million dollars per year. Chronic carriers of the HBV have been defined as those who are HBV surface antigen positive for greater than 6 months.
  • the incubation period for HBV usually lasts from 2 to 4 months, although it may be very short (10 days) or extremly long (9 months).
  • hepatitis B surface antigen HBs-Ag
  • HBe- Ag HBe- Ag
  • anti-HBc-Ag Abs antibodies to hepatitis B core antigen
  • IgM class antibodies to hepatitis B core antigen
  • anti-HBc antibodies will be detectable in the patient's serum for the rest of his life, no matter whether there is an acute hepatitis B, a form of persisting viral infection, or some naturally acquired immunity to HBV.
  • the most significant event indicating a chronic course of hepatitis B is the absence of the HBsAg/anti-HBs seroconversion. If this phenomenon has not occurred within 6 months after the onset of the disease, persistence of the HBV infection and the related clinical pictures (asymptomatic HBsAg carrier, chronic hepatitis, cirrhosis, or hepatoma) have to be reckoned with.
  • a first aspect of the present invention relates to compounds having the general formula (II):
  • Xi represents -CHGhy or -C(CH 3 )Ghy
  • X 2 represents -H, -OCH 3 , -CHGhy or -C(CH 3 )Ghy;
  • R', R" are independently of each other -OH, -SH, -NH 2 , methyl, ethyl or propyl;
  • a and A are independently of each other -NH(CO)-, -NH-, -(CO)NH-, -NH(CO)NH- or -O-;
  • X'1 is independently of X1 -CHGhy or -C(CH 3 )Ghy;
  • X' 2 is independently of X 2 -H, -OCH 3 , -CHGhy or -C(CH 3 )Ghy;
  • n and p are independently of each other an integer of 0 to 10; under the proviso that A ⁇ A'; and pharmaceutically acceptable salts thereof.
  • a second aspect of the present invention relates to the use of compounds of the general formula (II):
  • Xi represents -CHGhy or -C(CH 3 )Ghy
  • X 2 and X' 2 represents -H, -OCH 3 , -CHGhy or -C(CH 3 )Ghy;
  • R', R" are independently of each other -OH, -SH, -NH 2 , methyl, ethyl
  • a and A are independently of each other -NH(CO)-, -NH- -(CO)NH-, -NH(CO)NH- or -O-;
  • X' ⁇ is independently of Xi -CHGhy or -C(CH 3 )Ghy;
  • X' 2 is independently of X 2 -H, -OCH 3 , -CHGhy or -C(CH 3 )Ghy;
  • n and p are independently of each other an integer of 0 to 10; under the proviso that A ⁇ A'; and pharmaceutically acceptable salts thereof as pharmaceutically active agents.
  • a third aspect of the present invention relates to the use of compounds of the general formula (II):
  • Xi represents -CHGhy or -C(CH 3 )Ghy
  • X 2 represents -H, -OCH 3 , -CHGhy or -C(CH 3 )Ghy;
  • X' ⁇ is independently of Xi -CHGhy or -C(CH 3 )Ghy;
  • X' 2 is independently of X 2 -H, -OCH 3 , -CHGhy or -C(CH 3 )Ghy;
  • n and p are independently of each other integer of 0 to 10; and pharmaceutically acceptable salts thereof as pharmaceutically active agents for prophylaxis and/or treatment of Hepatitis B virus infections and associated diseases, including opportunistic infections.
  • the guanylhydrazone compounds of the invention are basic and form pharmaceutically acceptable salts with organic and inorganic acids.
  • suitable acids for such acid addition salt formation are hydrochloric acid, hydrobromic acid, sulfuric acid, phosphoric acid, acetic acid, citric acid, oxalic acid, malonic acid, salicylic acid, p-aminosalicylic acid, malic acid, fumaric acid, succinic acid, ascorbic acid, maleic acid, sulfonic acid, phosphonic acid, perchloric acid, nitric acid, formic acid, propionic acid, gluconic acid, lactic acid, tartaric acid, hydroxymaleic acid, pyruvic acid, phenylacetic acid, benzoic acid, p-aminobenzoic acid, p-hydroxybenzoic acid, methanesulfonic acid, ethanesulfonic acid, nitrous acid, hydroxyethanesulfonic acid, ethylenesulfonic acid,
  • the free base forms may be regenerated by treating the salt with a suitable dilute aqueous base solution such as dilute aqueous sodium hydroxide, potassium carbonate, ammonia and sodium bicarbonate.
  • a suitable dilute aqueous base solution such as dilute aqueous sodium hydroxide, potassium carbonate, ammonia and sodium bicarbonate.
  • the free base forms differ from their corresponding salt forms somewhat in certain physical properties, such as solubility in polar solvents, but the salts are otherwise equivalent to their corresponding free base forms for purposes of this invention.
  • X 2 (X' 2 ) is not hydrogen the meta position of Xi and X 2 (X'1 and X' 2 ) to Z is most preferred, because of steric reasons.
  • A, A', and B represent the residues as mentioned above.
  • inventive guanylhydrazone compounds wherein Z is -NH(COHCH 2 ) n -(CO)NHB and n is an integer of 3 to 10 and these compounds wherein Z stands for -NH(COHCH 2 ) n -C 6 H 4 -(CH 2 ) p -(CO)NHB,
  • B represents another benzene substituted with one or two guanylhydrazone residues as shown above.
  • inventive compounds selected from the group comprising: N-(4-acetylphenyl)-N'-(3,5-diacetylphenyl)urea tris (amidinohydrazone),
  • N,N'-bis (3,5-diacetylphenyl) hexane diamide tetrakis (amidinohydrazone)
  • N,N'-bis (3,5-diacetylphenyl) heptane diamide tetrakis (amidinohydrazone)
  • the compounds of the general formula (I), as defined below, and/or (II) and/or pharmaceutically acceptable salts thereof can be used to manufacture pharmaceutical formulations useful for prophylaxis and/or treatment of Hepatitis B virus infections, including opportunistic infections, and associated diseases.
  • inventive compounds are especially useful for prophylaxis and/or treatment of acute and/or chronic Hepatitis B virus infections.
  • inventive guanylhydrazone compounds can be used to treat diseases induced by HBV infection or diseases associates with HBV infection. Said diseases comprise chronic liver disease, cirrhosis or hepatocellular carcinoma induced by HBV infection.
  • the compounds of the general formula (I), as defined below, and (II) and their pharmaceutical active salts are especially useful for treating drug resistant Hepatitis B virus strains.
  • Said drug resistant hepatitis B virus strains are especially Lamivudine resistant strains.
  • the compounds of the general formula (I) and/or (II) and/or pharmaceutically acceptable salts are also useful for regulating the production and/or replication of Hepatitis B viruses in an individual and/or in cells.
  • mammals As used herein the term "individual” refers to mammals, especially humans.
  • the compounds of the general formula (I) and/or (II) and/or pharmaceutically acceptable salts are administered in a dosage corresponding to an effective concentration in the range of 0.01 - 100 ⁇ M, preferably in the range of 0.1 - 100 ⁇ M, more preferably in the range of 1 - 10 ⁇ M, and most preferably in the range of 1 - 5 ⁇ M.
  • a "pharmaceutical effective concentration" of an inhibitor is an amount effective to achieve the desired physiological result, either in cells treated in vitro or in a subject treated in vivo.
  • a pharmaceutically effective amount is an amount sufficient to inhibit, for some period of time, one or more of the clinically defined pathological processes associated with the viral infection.
  • the effective amount may vary depending on the specific inhibitor selected, and is also dependent on a variety of factors and conditions related to the subject to be treated and the severity of the infection. For example, if the inhibitor is to be administered in vivo, factors such as the age, weight and health of the patient as well as dose response curves and toxicity data obtained in pre- clinical animal work would be among those considered. If the inhibitor is to be contacted with the cells in vitro, one would also design a variety of pre-clinical in vitro studies to assess such parameters as uptake, half-life, dose, toxicity, etc. The determination of a pharmaceutically effective concentration for a given agent is well known within the ability of those skilled in the art.
  • Another preferred embodiment of the present invention describes the use of at least one compound of the general formula (I) and/or (II) and/or pharmaceutically active salts thereof in combination with further therapeutic compounds.
  • Said further therapeutic compounds are selected from the group comprising lamivudine, Zeffix ® , Heptovir ® , 3TC ® , Epivir-HBV, Combivir ® , Trizivir ® by GlaxoSmithKline), alpha interferon (e.g., Intron A ® by Schering-Plough), FTC (e.g., Coviracil ® by Triangle Pharmaceuticals), DAPD (DXG, by Triangle), L- FMAU (e.g., Clevudine ® by Triangle), Adefovir dipivoxil (by Gilead Sciences), tenofovir, epavudine, epcitabine, lobucavir, Penciclovir (GlaxoSmithKline), Famvir, Entecavir
  • a further aspect of the present invention is directed to a method for preventing and/or treating Hepatitis B virus infections, including opportunistic infections, and associated diseases in a mammal, including a human, which comprises administering to the mammal an amount of at least one compound of the general formula (II) and/or pharmaceutically acceptable salts thereof effective to prevent and/or treat Hepatitis B virus infections, including opportunistic infections, and associated diseases.
  • a method for regulating the production and/or replication of Hepatitis B viruses in an individual comprises administering to the individual an amount of at least one compound of the general formula (II) and/or pharmaceutically acceptable salts thereof effective to at least partially inhibit the production and/or replication of Hepatitis B virus.
  • a similar aspect of the present invention describes a method for regulating the production and/or replication of Hepatitis B viruses in cells, which comprises administering to the cells an amount of at least one compound of the general formula (II) and/or pharmaceutically acceptable salts thereof effective to at least partially inhibit the production and/or replication of Hepatitis B virus.
  • Another aspect of the present invention relates to the use of a compound having the formula (I):
  • Xi, X' ⁇ and X' 2 are, independently, GhyCH- or GhyCCH 3 -;
  • Z is -NH(CO)NH-, -(C 6 H 4 )-, -(C 5 NH 3 )-, or -A-(CH 2 ) n -A-, where n is an integer from 2-10, and monounsaturated or diunsaturated derivatives thereof, and wherein Z may be unsubstituted or may be methyl-, dimethyl- or trimethyl-substituted at any carbon atom, or may be substituted with a heteroatom (e.g., N, O, S); and A is independently -NH(CO)-, -(CO)NH-, -NH(CO)NH-, -NH- or -0-, and salts thereof, as an inhibitor of Hepatitis B Virus (HBV).
  • HBV Hepatitis B Virus
  • X 2 is GhyCH- or GhyCCH 3 -
  • X 2 is meta or para to Xi
  • X' 2 is meta or para to XV
  • Z is -NH(CO)-Y-(CO)NH- and Y is (CH 2 ) n , wherein n is an integer from 6-10 and wherein up to 3 CH 2 groups may be substituted by heteroatom groups such as NH, O or S.
  • the compound is N,N'-bis(3,5- diacetylphenyl) decanediamide-tetrakis(amidino)hydrazone (Fig. 1) or a salt thereof, particularly a pharmaceutically acceptable salt such as the tetrahydrochloride.
  • a still further aspect of the present invention relates to the preparation of medicines for treatment of HBV infected individuals.
  • Pharmaceutical compositions including a compound according to formula (I) and/or (II) and/or their pharmaceutically active salts in a pharmaceutically acceptable carrier are contemplated.
  • Such pharmaceutical compositions include inserts instructive as to the treatment of HBV infection using the compound of formula (I) and/or (II) and/or pharmaceutically active salts thereof, and may also advantageously include additional active ingredients either separately or in admixture with the compound of formula (I) and/or (II) and/or pharmaceutically active salts thereof, such additional active ingredient can be selected form he group comprising lamivudine, Zeffix ® , Heptovir ® , 3TC ® , Epivir-HBV, Combivir ® , Trizivir ® by GlaxoSmithKline), alpha interferon (e.g., Intron A ® by Schering-Plough), FTC (e.g., Coviracil ® by Triangle Pharmaceuticals), DAPD (DXG, by Triangle), L-FMAU (e.g., Clevudine ® by Triangle), Adefovir dipivoxil (by Gilead Sciences), tenofovir, epa
  • Fig. 1 shows the structures of:
  • the compounds are provided in the form of their corresponding bis-, tris-, or tetrahydrochloride salts.
  • Fig. 2 shows a plot of the effect of various concentrations of a guanyl hydrazone compound (Compound 4, see Fig. 1 ) on hepatitis B virus expressing HepG2-
  • Fig. 3 shows a plot of the effect of various concentrations of a guanyl hydrazone compound (Compound 1 , see Fig. 1) on Hepatitis B virus expressing HepG2-
  • Fig. 4 shows a plot of the effect of various concentrations of a guanyl hydrazone compound (Compound 4, see Fig. 1 ) on 3TC-resistant HBV production in transiently transfected human hepatoma cells.
  • Fig. 5 shows data and synergy plot of combination anti-HBV activity of a guanyl hydrazone compound (Compound 4, see Fig. 1 ) with 3TC in Hepatitis B virus expressing HepG2-2.2.15 human hepatoma cells.
  • Fig. 6 shows the effect of various concentrations of a guanyl hydrazone compound
  • the present invention relates to the use of certain guanyl hydrazone compounds for inhibiting HBV replication.
  • the guanyl hydrazone compounds disclosed herein are suitable for use in treating HBV infection and are particularly useful for treating chronic HBV infection.
  • the compounds of formula (I) and (II), above, are potent inhibitors of HBV replication in the low micromolar range and exhibit low cellular toxicity at effective antiviral dosages.
  • the compounds described herein may be used as the active ingredient in medicines for the treatment of HBV- infection and in methods for treating patients suffering from hepatitis B.
  • HBV belongs to the DNA virus group of hepadnaviruses, which multiply by an entirely different mechanism and show an entirely different multiplication cycle.
  • no viral encoded protein is known that acts on nucleo- cytoplasmic translocation of viral RNA, unlike the Rev-type mechanism of HIV, which has been shown to be the relevant mechanism that is inhibited by the compounds of formula (I) and (II) and/or pharmaceutically active salts thereof with respect to that virus.
  • the compounds of formula (I) and (II) are potent inhibitors of HBV replication in the micromolar range by screening in a relevant HBV cellular assay.
  • the compounds of Fig. 1 were observed to reduce virus production in HepG2-2.2.15 human hepatoma cells to very low levels at concentrations between about 10 and 20 ⁇ M, while having little cytotoxic effect on control cells.
  • inhibitors While the specific mode of action of these inhibitors is not certain, it is believed that they may act as inhibitors of the biological function of certain cellular proteins involved in the translocation of essential viral mRNAs from the nucleus of an infected cell to the cytoplasm, notably elF-5A (Bevec et al., 1996, Science, 271 , p 1858-1860), TAP (human homologue of yeast Mex67p; - Gr ⁇ ter et al., 1998 ,Mol. Cell ,1 , p 649-659; Segref et al., 1997, EMBO J.
  • elF-5A Bevec et al., 1996, Science, 271 , p 1858-1860
  • TAP human homologue of yeast Mex67p; - Gr ⁇ ter et al., 1998 ,Mol. Cell ,1 , p 649-659; Segref et al., 1997, EMBO J.
  • p15 human homologue of Mtr2p;- Katahira et al., 1999, EMBO J. ,18, p 2593-2609; Santos- Rosa et al., 1998, Mol. Cell. Biol. ,18, p 6826-6838), L5 , TFIIIA (Rosorius et al., 2000, J. Biol. Chem., 275, p 12061-12068), and/or Dbp5/Rat8p (DEAD box type of RNA helicases;- Tseng et al., 1998, EMBO J. ,17, p 2651-2662; Snay-Hodge et al., 1998, EMBO J. ,17, p 2663-2676) proteins.
  • the compounds of formula (I) and (II) may be formulated in suitable pharmaceutical carriers and may be administed by any appropriate means, including but not limited to injection (intravenous, intraperitoneal, intramuscular, subcutaneous) by absorption through epithelial or mucocutaneous linings (oral mucosa, rectal and vaginal epithelial linings, nasopharyngial mucosa, intestinal mucosa); orally, rectally, transdermally, topically, intradermally, intragastrally, intracutaneously, intravaginally, intranasally, intrabuccally, percutaneously, sublingually, or any other means available within the pharmaceutical arts.
  • injection intravenous, intraperitoneal, intramuscular, subcutaneous
  • epithelial or mucocutaneous linings oral mucosa, rectal and vaginal epithelial linings, nasopharyngial mucosa, intestinal mucosa
  • transdermally topically, intradermally,
  • the compound may be administered to a subject in need thereof, e.g., a human patient, by itself or in pharmaceutical compositions where they are mixed with suitable carriers or excipients at doses which are sufficient for an at least partial reduction of virus production.
  • Therapeutically effective doses may be administered alone or as adjunctive therapy in combination with further therapeutic compounds, particularly antiviral agents and more particularly antiviral agents suitable for treatment of HBV infection, e.g., alpha interferon and/or reverse transcriptase inhibitors.
  • the compounds of formula (I) and/or (II) may be specifically combined with compounds such as lamivudine (e.g., Epivir-HBV), alpha interferon (e.g., Intron A) , Coviracil/FTC, DAPD, L-FMAU, adefovir dipivoxil, tenofovir, epavudine, epcitabine, lobucavir, Famvir/penciclovir, entecavir/BMS- 200475, racivir, DXG, L-ddA prodrug, HDP-P-acydovir, LM-019c, CS-1091 , PS- 019, PS-018, ara-AMP prodrugs, thymosin, (-)-Carbovir, hammerhead ribozymes, glycosidase inhibitors, and any other antiviral composition in use or in development for use to treat hepatitis B infection; with therapeutic vaccine strategies including live,
  • This invention also relates to the combination of the compounds of formula (I) and/or (II) with at least one of the above mentioned drugs.
  • the compounds described herein can be used as inhibitors of HBV as well as for treating diseases associated with HBV, especially where treatment with other drugs, particularly NRTIs or NNRTIs, has caused viral resistance against conventional antiviral drugs.
  • a suitable composition comprising the compound of the invention may be a solution of the compound in a suitable liquid pharmaceutical carrier or any other formulation such as tablets, pills, dragees, capsules, gels, syrups, slurries, suspensions and the like.
  • a therapeutically effective dosage of the compound of formula (I) or (II) refers to that amount of the compound that results in an at least partial inhibition of virus production in the patient, which may be measured in several ways, e.g., reduction in viral DNA in a patient hepatocyte sample, increase in cell viability, and/or reduction of HBV "enigma" antigen (HBe-Ag) and/or HBs-Ag in a patient's serum, and consequently results in a desired pharmaceutical effect.
  • Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical, pharmacological, and toxicological procedures in cell cultures or experimental animals for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between toxic and therapeutic effect is the therapeutic index and can be expressed as the ratio between LD50 and ED50.
  • the dosage of the compound lies preferably within a range of circulating concentrations that include the ED50 with little or no toxicity. More preferably, the dosage of the compound corresponds to an effective concentration in the range of 0.1 - 100 ⁇ M, most preferably 1 - 10 ⁇ M.
  • the actual amount of the composition administered will be dependent on the subject being treated, on the subject's weight, the severity of the HBV infection, the manner of administration, and the judgement of the prescribing physician.
  • Example 1 Effect of N,N'-bis(3,5-diacetylphenyl) decanediamide-tetrakis- (amidino)hydrazone on HBV production in the HepG2-2.2.15 cell culture system.
  • HepG2-2.2.15 human hepatoma cells obtainable according to the procedure described in Sells et al., 1987, Prnc Natl. Acart. Sci. USA t Vol. 84, pp. 1005- 1009, which is fully incorporated by reference, were plated in 96-well microtiter plates in DMEM medium supplemented with fetal bovine serum. After incubation at 37°C in a humidified, 5% C0 2 environment for 16-24 hours, the monolayer of HepG2-2.2.15 cells was washed and the medium was replaced with complete medium containing various concentrations of test compound. Every three days, the culture medium was replaced with fresh medium containing the appropriately diluted test compound.
  • the cell culture supernatant was collected and clarified by centrifugation. The cell viability was evaluated by a dye uptake procedure (Cell Titer Aqueous One Solution, by Promega).
  • Virion-associated HBV DNA present in the tissue culture supernatant was amplified employing PCR technology using primer pairs derived from the HBV strain ayw. Subsequently, the PCR-amplified HBV DNA was detected in real time by monitoring increases in fluorescence signals that result from exonucleolytic degradation of a quenched fluorescent probe molecule following hybridization of the probe to the amplified HBV DNA (TaqMan; Perkin-Elmer).
  • Example 2 Effect of N-(4-acetylphenyl)-N'-(3,5-diacetylphenyl)urea tris (amidinohydrazone) on HBV production in the HepG2-2.2.15 cell culture system.
  • the assay was performed as described above in example 1. Briefly, the toxicity of the compound was evaluated by CellTiter 96® AQueous One Solution Cell Proliferation Assay (Promega) and the activity of the compound against HBV by quantification of virion-associated DNA in the cell culture supematants with Real Time PCR.
  • Example 3 Effect of Compound 4 on 3TC-resistant (M552V) HBV production.
  • HepG2 cells were plated in 24- well plates and transfected with either wild type or 3TC resistant (M552V) HBV genome containing plasmid DNA. After an incubation of 16 hours, Compound 4 was added at five half-log dilutions in duplicates. The culture medium was replaced one day later with fresh medium containing the appropriately diluted drug. Six days post transfection, the cells were used for testing toxicity with the CellTiter 96® AQueous Cell Proliferation Assay (Promega) and the cell supernatants were harvested. Following toxicity testing, the cells were lysed. The culture supemates or cell lysates were subjected to treatment with DNase after a low speed centrifugation.
  • M552V 3TC resistant
  • Virus DNA present in the tissue culture supernatants or cell lysates was PCR amplified using primers derived from HBV strain ayw. Subsequently, the PCR-ampIified HBV DNA was detected in real-time by monitoring increases in fluorescence signals that result from exonucleolytic degradation of a quenched fluorescent probe molecule following hybridization of the probe to the amplified HBV DNA.
  • the O.D number obtained with CellTiter 96® AQueous Cell Proliferation Assay and the HBV DNA copy number obtained with Real Time PCR were analyzed using a computer program which calculates the percentage of DNA copy number, IC50, the percentage of cell viability, TC 50 and Tl (therapeutic index) and displays the graphic results.
  • the IC50 of 3TC against the 3TC resistant mutant HBV was > 10 ⁇ M, while it is around 0.05 ⁇ M against HBV ayw (data not shown).
  • This in vitro combination assay was designed to define the antiviral interaction of the two compounds and to determine if their interaction is additive, synergistic or antagonistic. In addition, the data are evaluated for indications of enhanced toxicity when the compounds are used in combination with each other.
  • HepG2-2.2.15 cells were plated in 96-well microtiter plates in DMEM medium supplemented with fetal bovine serum. After incubation at 37°C in a humidified, 5% C0 2 environment for 16-24 hours, the monolayer of HepG2-2.2.15 cells was washed and the medium replaced with complete medium containing five concentrations of the test compound in all possible combinations with five concentrations of 3TC. Every three days, the culture medium was replaced with fresh medium containing the appropriately diluted drug. Six days following the initial administration of test compound, the cell culture supernate was collected and clarified by centrifugation.
  • the cell viability was evaluated by CellTiter 96® AQueous One Solution Cell Proliferation Assay.
  • This assay is a colorimetric method for determining the number of viable cells in proliferation or cytotoxicity assays.
  • the reagent contains a novel tetrazolium compound, MTS, and an electron coupling agent, PES, which, when combined, form a stable solution.
  • MTS novel tetrazolium compound
  • PES electron coupling agent
  • the MTS tetrazolium compound is bioreduced into a formazan product by NADPH or NADH produced by dehydrogenase in metabolically active cells. Therefore, the quantity of formazan product measured is directly proportional to the number of living cells in culture.
  • Virion-associated HBV DNA present in the tissue culture supernate was PCR amplified using primers derived from HBV strain ayw. Subsequently, the PCR- amplified HBV DNA was detected in real-time by monitoring increases in fluorescence signals that result from exonucleolytic degradation of a quenched fluorescent probe molecule following hybridization of the probe to the amplified HBV DNA.
  • Combination Anti-HBV Data Analysis and statistical evaluation of drug combination assays was performed according to the method of Prichard and Shipman (Antiviral Research 14:181-206 [1990]).
  • Combination antiviral assays were performed utilizing the HepG2.2.15 cell-line and a quantitative TaqMan HBV assay to measure virus-associated DNA levels at the experimental endpoint.
  • Five concentrations of 3TC were tested in all possible combinations with five concentrations of Compound 4. Effects of the drug combinations were calculated based on the activity of each compound when tested alone.
  • the expected additive antiviral protection was subtracted from the experimentally determined antiviral activity at each combination concentration resulting in a positive value (synergy), a negative value (antagonism), or zero (additivity).
  • synergy is defined as drug combinations yielding synergy volumes greater than 50 ⁇ M 2 %.
  • synergistic activity and highly synergistic activity have been defined as yielding synergy volumes of 50-100 ⁇ M 2 % and >100 ⁇ M 2 %, respectively.
  • Additive drug interactions have synergy volumes in the range of -50 ⁇ M 2 % to 50 ⁇ M 2 %, while synergy volumes less than -50 ⁇ M 2 % are considered slightly antagonistic or antagonistic ( ⁇ -100 ⁇ M 2 %).
  • Example 5 Compound 4 mechanism of action.
  • HepG2-2.2.15 cells were plated in 24-well microtiter plates in DMEM medium supplemented with fetal bovine serum. After incubation at 37°C in a humidified, 5% C0 2 environment for 16-24 hours, the monolayer of HepG2-2.2.15 cells was washed and the medium replaced with complete medium containing five concentrations of Compound 4 or 3TC. Each concentration was tested in duplicates. Every three days the culture medium was replaced with fresh medium containing the appropriately diluted drug. Six days following the initial administration of test compound, cell viability was evaluated by a dye (CellTiter 96® AQueous) uptake procedure. Following the cell viability testing, the cells were lysed and the nuclei removed.
  • CellTiter 96® AQueous a dye uptake procedure. Following the cell viability testing, the cells were lysed and the nuclei removed.
  • the capsids present in the cell lysates were resolved via agarose/sodium phosphate electrophoresis. Following transfer onto the nylon membrane, the viral positive strand nucleic acids associated with the viral capsids were detected with a P -labeled negative strand riboprobe prepared in vitro and visualized by exposing the membrane onto X-ray film.
  • capsid gels The results from the capsid gels are shown in Fig. 6.
  • capsids from untreated cells (0 ⁇ M compound 4 or 0 ⁇ M 3TC)
  • pregenomic HBV RNA as well as plus strand viral DNA are responsible for the hybridization signal seen with the minus strand riboprobe.
  • the signal for plus strand nucleic acid was significantly higher in capsids from cells treated with the reverse transcriptase inhibitor 3TC, since the pregenomic RNA is not transcribed into DNA and cytoplasmic capsids containing RNA presumably accumulate in the cytoplasm in the absence of virus secretion.

Landscapes

  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Virology (AREA)
  • Oncology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Pulmonology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

L'invention concerne des inhibiteurs d'infections par le VHB. On décrit une classe de guanyl hydrazones qui sont des inhibiteurs puissants d'infections par le VHB. Ce type de composé est utile pour le traitement d'une infection par le VHB, en particulier l'infection chronique par le VHB, et pour le traitement d'infections et de maladies opportunistes engendrées par l'infection en question. L'invention concerne également des compositions pharmaceutiques utiles pour la prophylaxie et/ou le traitement des infections par le VHB, y compris les infections opportunistes, et des maladies résultant d'infections par le VHB.
PCT/EP2001/007260 2000-06-27 2001-06-26 Inhibiteurs d'infections par le virus de l'hepatite b (vhb) WO2002000613A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2001269092A AU2001269092A1 (en) 2000-06-27 2001-06-26 Inhibitors of hepatitis b virus infection

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US21455600P 2000-06-27 2000-06-27
US60/214,556 2000-06-27

Publications (2)

Publication Number Publication Date
WO2002000613A2 true WO2002000613A2 (fr) 2002-01-03
WO2002000613A3 WO2002000613A3 (fr) 2002-11-07

Family

ID=22799534

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2001/007260 WO2002000613A2 (fr) 2000-06-27 2001-06-26 Inhibiteurs d'infections par le virus de l'hepatite b (vhb)

Country Status (2)

Country Link
AU (1) AU2001269092A1 (fr)
WO (1) WO2002000613A2 (fr)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003006426A1 (fr) * 2001-07-13 2003-01-23 Axxima Pharmaceuticals Ag Guanylhydrazones aromatiques utilises comme composes efficaces contre les maladies nerveuses
EP1572095A2 (fr) * 2002-09-13 2005-09-14 Idenix (Cayman) Limited S-l-2'-desoxynucleosides pour le traitement de souches resistantes du vhb, et therapies combinees associees
US7001924B2 (en) 2001-09-21 2006-02-21 Message Pharmaceuticals Inhibitors of RNase P proteins as antibacterial compounds
WO2006130697A1 (fr) * 2005-06-01 2006-12-07 Cytokine Pharmasciences, Inc. Expression de hmgb1 et role protecteur de semapimod dans nec
US7863334B2 (en) 2004-08-17 2011-01-04 Cytokine Pharmasciences, Inc. Guanylhydrazone compounds, compositions, methods of making and using
US8034840B2 (en) 2004-06-25 2011-10-11 Cytokine Pharmasciences, Inc. Guanylhydrazone salts, compositions, processes of making, and methods of using
US8642752B2 (en) 2011-04-21 2014-02-04 Isis Pharmaceuticals, Inc. Modulation of Hepatitis B virus (HBV) expression
US8765802B2 (en) 2007-06-12 2014-07-01 Provid Pharmaceuticals, Inc. Kinase inhibitors, compositions thereof, and methods of use therewith
AU2013355220B2 (en) * 2012-12-06 2018-08-02 Baruch S. Blumberg Institute Functionalized benzamide derivatives as antiviral agents against HBV infection
CN113134002A (zh) * 2021-04-26 2021-07-20 北京慧宝源生物技术股份有限公司 克来夫定的应用

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB923398A (en) * 1960-08-09 1963-04-10 Vismara Francesco Spa Diphenyl mono-and bis-formyl guanylhydrazones
US3560557A (en) * 1965-11-19 1971-02-02 Ciba Geigy Corp Diphenyl-ureas,-thioureas,-guanidines and -parabanic acids
US5689563A (en) * 1993-06-29 1997-11-18 Motorola, Inc. Method and apparatus for efficient real-time authentication and encryption in a communication system
WO1998020868A1 (fr) * 1996-11-15 1998-05-22 The Picower Institute For Medical Research Guanylhydrazones utiles dans le traitement de maladies associees a l'activation des cellules t
WO1998030583A1 (fr) * 1997-01-08 1998-07-16 The Picower Institute For Medical Research Complexes et combinaisons de fetuine et d'agents therapeutiques
US5854289A (en) * 1994-01-21 1998-12-29 The Picower Institute For Medical Research Guanylhydrazones and their use to treat inflammatory conditions
WO2001025192A1 (fr) * 1999-10-06 2001-04-12 Melacure Therapeutics Ab Derives de guanidine et leur utilisation dans la production d'un medicament destine a bloquer les xanthine oxydases/deshydrogenases
WO2001056553A2 (fr) * 2000-02-02 2001-08-09 Axxima Pharmaceuticals Ag Guanylhydrazones aromatiques, actives sur le plan pharmacologique
WO2002011715A2 (fr) * 2000-08-07 2002-02-14 Melacure Therapeutics Ab Benzylideneaminoguanidines et hydroxyguanidines utilisees comme ligands des recepteur de la melanocortine

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB923398A (en) * 1960-08-09 1963-04-10 Vismara Francesco Spa Diphenyl mono-and bis-formyl guanylhydrazones
US3560557A (en) * 1965-11-19 1971-02-02 Ciba Geigy Corp Diphenyl-ureas,-thioureas,-guanidines and -parabanic acids
US5689563A (en) * 1993-06-29 1997-11-18 Motorola, Inc. Method and apparatus for efficient real-time authentication and encryption in a communication system
US5854289A (en) * 1994-01-21 1998-12-29 The Picower Institute For Medical Research Guanylhydrazones and their use to treat inflammatory conditions
WO1998020868A1 (fr) * 1996-11-15 1998-05-22 The Picower Institute For Medical Research Guanylhydrazones utiles dans le traitement de maladies associees a l'activation des cellules t
WO1998030583A1 (fr) * 1997-01-08 1998-07-16 The Picower Institute For Medical Research Complexes et combinaisons de fetuine et d'agents therapeutiques
WO2001025192A1 (fr) * 1999-10-06 2001-04-12 Melacure Therapeutics Ab Derives de guanidine et leur utilisation dans la production d'un medicament destine a bloquer les xanthine oxydases/deshydrogenases
WO2001056553A2 (fr) * 2000-02-02 2001-08-09 Axxima Pharmaceuticals Ag Guanylhydrazones aromatiques, actives sur le plan pharmacologique
WO2002011715A2 (fr) * 2000-08-07 2002-02-14 Melacure Therapeutics Ab Benzylideneaminoguanidines et hydroxyguanidines utilisees comme ligands des recepteur de la melanocortine

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CAVALLINI G ET AL: "ANTIBACTERIAL AGENTS. SOME NEW GUANYLHYDRAZONE DERIVATIVES" JOURNAL OF MEDICINAL AND PHARMACEUTICAL CHEMISTRY, AMERICAN CHEMICAL SOCIETY. EASTON, US, vol. 4, no. 1, 1 July 1961 (1961-07-01), pages 177-182, XP001021572 *
CERAMI, CARLA ET AL: "High-performance liquid chromatographic method for guanylhydrazone compounds" J. CHROMATOGR., B: BIOMED. APPL. (1996), 675(1), 71-5 , XP002206329 *
COHEN P S ET AL: "The critical role of p38 MAP kinase in T cell HIV -1 replication." MOLECULAR MEDICINE, vol. 3, no. 5, May 1997 (1997-05), pages 339-346, XP001018447 *
KORYTNYK, W. ET AL: "Guanylhydrazones with potential antileukemic activity. 2. Synthesis and structure-activity relationships of analogs of 4,4'-diacetyl-N,N'-diphenylurea bis(guanylhydrazone)" J. MED. CHEM. (1978), 21(6), 507-13 , XP002206328 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003006426A1 (fr) * 2001-07-13 2003-01-23 Axxima Pharmaceuticals Ag Guanylhydrazones aromatiques utilises comme composes efficaces contre les maladies nerveuses
US7001924B2 (en) 2001-09-21 2006-02-21 Message Pharmaceuticals Inhibitors of RNase P proteins as antibacterial compounds
EP1572095A2 (fr) * 2002-09-13 2005-09-14 Idenix (Cayman) Limited S-l-2'-desoxynucleosides pour le traitement de souches resistantes du vhb, et therapies combinees associees
EP1572095A4 (fr) * 2002-09-13 2008-11-05 Novartis Ag S-l-2'-desoxynucleosides pour le traitement de souches resistantes du vhb, et therapies combinees associees
US7928086B2 (en) 2002-09-13 2011-04-19 Novartis Ag β-L-2′-deoxynucleosides for the treatment of resistant HBV strains and combination therapies
US8158606B2 (en) 2002-09-13 2012-04-17 Novartis, Ag β-L-2′-deoxynucleosides for the treatment of resistant HBV strains and combination therapies
CN1968686B (zh) * 2004-06-25 2013-01-16 费林有限公司 脒腙盐、组合物、制备方法和使用方法
US8034840B2 (en) 2004-06-25 2011-10-11 Cytokine Pharmasciences, Inc. Guanylhydrazone salts, compositions, processes of making, and methods of using
EP2540703A1 (fr) * 2004-06-25 2013-01-02 Cytokine Pharmasciences, Inc. Sels de guanylhydrazone, compositions à base de ceux-ci, procédés et production et d'utilisation de ceux-ci
CN101010094B (zh) * 2004-08-17 2013-05-22 辉凌公司 脒腙化合物、组合物、制备方法及使用方法
US7863334B2 (en) 2004-08-17 2011-01-04 Cytokine Pharmasciences, Inc. Guanylhydrazone compounds, compositions, methods of making and using
US8247455B2 (en) 2004-08-17 2012-08-21 Ferring B.V. Guanylhydrazone compounds, compositions, methods of making and using
US7795314B2 (en) 2005-06-01 2010-09-14 Cytokine Pharmasciences, Inc. Protective role of semapimod in necrotizing enterocolitis
WO2006130697A1 (fr) * 2005-06-01 2006-12-07 Cytokine Pharmasciences, Inc. Expression de hmgb1 et role protecteur de semapimod dans nec
US8765802B2 (en) 2007-06-12 2014-07-01 Provid Pharmaceuticals, Inc. Kinase inhibitors, compositions thereof, and methods of use therewith
US8642752B2 (en) 2011-04-21 2014-02-04 Isis Pharmaceuticals, Inc. Modulation of Hepatitis B virus (HBV) expression
US9677076B2 (en) 2011-04-21 2017-06-13 Ionis Pharmaceuticals, Inc. Modulation of hepatitis B virus (HBV) expression
AU2013355220B2 (en) * 2012-12-06 2018-08-02 Baruch S. Blumberg Institute Functionalized benzamide derivatives as antiviral agents against HBV infection
CN113134002A (zh) * 2021-04-26 2021-07-20 北京慧宝源生物技术股份有限公司 克来夫定的应用
CN113134002B (zh) * 2021-04-26 2023-12-19 北京慧宝源生物技术股份有限公司 克来夫定的应用

Also Published As

Publication number Publication date
WO2002000613A3 (fr) 2002-11-07
AU2001269092A1 (en) 2002-01-08

Similar Documents

Publication Publication Date Title
US11207370B2 (en) Peptidomimetics for the treatment of coronavirus and picornavirus infections
JP4491136B2 (ja) 関節症を治療するためのファルネシルタンパク質トランスフェラーゼ阻害剤
US20110184019A1 (en) Long chain N-Alkyl compounds and oxa-derivatives thereof
EP2283833A2 (fr) Amines et amides pour le traitement de maladies
US20050261251A1 (en) Antiviral agents and methods of treating viral infections
CA2862193C (fr) Composition anti-tuberculose contenant des composes a base d'oxazole
JPH06219945A (ja) 化合物の改良
JP2008538362A (ja) タンパク質の成熟の遮断を介して疾患を処置するための方法、タンパク質ジスルフィドイソメラーゼなどの分子シャペロンの機能を阻害する、またはグリコシル化を妨害する化合物、それらを含む薬学的組成物、および治療剤を同定するためのスクリーニング方法
JPS6197228A (ja) ジヒドロピリジン類とace阻害剤との配合物
WO2002000613A2 (fr) Inhibiteurs d'infections par le virus de l'hepatite b (vhb)
JP2829440B2 (ja) O▲上6▼―アルキルグアニン―dnaアルキルトランスフェラーゼ減少化活性を有するo▲上6▼―ベンジル化グアニン、グアノシンおよび2′―デオキシグアノシン化合物
JP2023516628A (ja) ヌクレオチド系化合物のコロナウイルス感染症の治療での使用
BR112021000938A2 (pt) composto sal farmaceuticamente aceitável, tautômero, isômero ou estereoisômero do mesmo, composição farmacêutica e processo para a síntese do dito composto
JP2017534591A (ja) 心肺障害の処置のためのスフィンゴシン−1−リン酸受容体モジュレーター
JP2001072585A (ja) ウイルス性心筋炎の予防または治療薬剤
KR20060037258A (ko) P2X7-수용체 길항제 및 종양 괴사 인자 α를 포함하는제약 조성물
WO1998033501A1 (fr) Derives de 2-benzoylamino-3-phenylpropenamide et leurs procedes d'utilisation
WO2021248008A1 (fr) Composés arylamides pour le traitement et la prévention d'infections virales
JP2023514794A (ja) ポリadpリボースポリメラーゼ阻害剤の抗コロナウイルス応用
US20070010497A1 (en) Pharmaceutical composition comprising a p2x7 antagonist and sulfasalazine
JP2023510279A (ja) インフラマソーム活性化の抑制
US20070281931A1 (en) Pharmaceutical Composition Containing a P2x7 Receptor Antagonist and Methotrexate
JP2013500957A (ja) 炎症治療用化合物
CA2251646A1 (fr) Procede permettant de prevenir et de traiter une infection pestivirale et les maladies qui y sont associees
JPH0352816A (ja) 腎炎の治療剤

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

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

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CR CU CZ DE DK DM DZ EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

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 in:

Ref country code: JP