WO2004112687A2 - Antiviral acylguanidine compounds and methods - Google Patents

Antiviral acylguanidine compounds and methods Download PDF

Info

Publication number
WO2004112687A2
WO2004112687A2 PCT/AU2004/000866 AU2004000866W WO2004112687A2 WO 2004112687 A2 WO2004112687 A2 WO 2004112687A2 AU 2004000866 W AU2004000866 W AU 2004000866W WO 2004112687 A2 WO2004112687 A2 WO 2004112687A2
Authority
WO
WIPO (PCT)
Prior art keywords
guanidine
cinnamoylguanidine
amiloride
trans
hydrochloride
Prior art date
Application number
PCT/AU2004/000866
Other languages
French (fr)
Other versions
WO2004112687A3 (en
Inventor
Peter William Gage
Gary Dinneen Ewart
Lauren Elizabeth Wilson
Wayne Best
Anita Premkumar
Original Assignee
Biotron Limited
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 AU2003903251A external-priority patent/AU2003903251A0/en
Priority claimed from AU2003903850A external-priority patent/AU2003903850A0/en
Priority claimed from AU2003904692A external-priority patent/AU2003904692A0/en
Priority to US10/562,296 priority Critical patent/US20070099968A1/en
Priority to EP04737487A priority patent/EP1646371A4/en
Priority to EP13164504.6A priority patent/EP2617709B8/en
Priority to JP2006515560A priority patent/JP5030587B2/en
Priority to BRPI0411900A priority patent/BRPI0411900B8/en
Priority to KR1020057024860A priority patent/KR101153254B1/en
Application filed by Biotron Limited filed Critical Biotron Limited
Priority to AU2004248859A priority patent/AU2004248859C1/en
Priority to NZ544671A priority patent/NZ544671A/en
Priority to CN2004800240974A priority patent/CN101111475B/en
Priority to CA2529949A priority patent/CA2529949C/en
Publication of WO2004112687A2 publication Critical patent/WO2004112687A2/en
Publication of WO2004112687A3 publication Critical patent/WO2004112687A3/en
Priority to US13/553,239 priority patent/US20130035328A1/en
Priority to US14/615,616 priority patent/US20150313909A1/en
Priority to US15/602,526 priority patent/US10472332B2/en
Priority to US16/554,990 priority patent/US11192863B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D241/00Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings
    • C07D241/02Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings
    • C07D241/10Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D241/14Heterocyclic compounds containing 1,4-diazine or hydrogenated 1,4-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D241/24Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D241/26Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with nitrogen atoms directly attached to ring carbon atoms
    • C07D241/28Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals with nitrogen atoms directly attached to ring carbon atoms in which said hetero-bound carbon atoms have double bonds to oxygen, sulfur or nitrogen atoms
    • C07D241/34(Amino-pyrazine carbonamido) guanidines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D277/00Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings
    • C07D277/02Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings
    • C07D277/20Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D277/32Heterocyclic compounds containing 1,3-thiazole or hydrogenated 1,3-thiazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D277/38Nitrogen atoms
    • C07D277/44Acylated amino or imino radicals
    • C07D277/46Acylated amino or imino radicals by carboxylic acids, or sulfur or nitrogen analogues thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/15Oximes (>C=N—O—); Hydrazines (>N—N<); Hydrazones (>N—N=) ; Imines (C—N=C)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/38Heterocyclic compounds having sulfur as a ring hetero atom
    • A61K31/381Heterocyclic compounds having sulfur as a ring hetero atom having five-membered rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4406Non condensed pyridines; Hydrogenated derivatives thereof only substituted in position 3, e.g. zimeldine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • 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
    • 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
    • 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
    • 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
    • A61P31/18Antivirals for RNA viruses for HIV
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C279/00Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups
    • C07C279/20Derivatives of guanidine, i.e. compounds containing the group, the singly-bound nitrogen atoms not being part of nitro or nitroso groups containing any of the groups, X being a hetero atom, Y being any atom, e.g. acylguanidines
    • C07C279/22Y being a hydrogen or a carbon atom, e.g. benzoylguanidines
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the present invention relates to methods for retarding, reducing or otherwise inhibiting viral growth and/or functional activity.
  • the invention also relates to compounds and compositions suitable for use in the methods.
  • PCT/AU99/00872 describes the use of compounds 5-(N,N- hexamethylene)-amil ⁇ ide and 5-(N,N-dime1hyl)-amiloride in the treatment of HIV infection.
  • HCV Hepatitis C virus
  • Coronaviruses (Order Nid ⁇ vir les. family Coronaviridae, Genus Coronavirus) are enveloped positive-stranded RNA viruses that bud from the endoplasmic reticulum- Golgi intermediate compartment or the er ⁇ -Golgi network (Fischer, Stegen et al. 1998; Maeda, Maeda et al. 1999i Corse and Machamer 2000; Maeda, Repass et al. 2001 ; Ruo and Masters 2003)
  • Animal coronaviruses can cause respiratory, gastrointestinal, neurological, or hepatic diseases in their host (Peiris, Lai et al. 2003).
  • Several animal coronavirus are significant veterinary pathogens (Rota, Oberste et al. 2003).
  • Severe acute respiratory syndrome is caused by a newly identified virus.
  • SARS is a respiratory illness that has recently been reported in Asia, North America, and Europe (Peiris, Lai et al.2003).
  • the causative agent of SARS was identified as a coronavirus. (Drosten, Gunther et al.2003; Ksiazek, Erd an et al. 2003; Peiris, Lai et al.2003).
  • the World Health Organization reports that the cumulative number of reported probable cases of SARS from 1 November 2002 to the 11 th July 2003 is 8,437 with 813 deaths, nearly a 10% death rate. It is believed that SARS will not be eradicated, but will cause seasonal epidemics like the.cold or influenza viruses (Vogel 2003).
  • the inventors have surprisingly found that certain compounds that fall under the classification of substituted acylguanidines have antiviral activity against viruses from a range of different virus families, Without intending to be bound by any $ particular theory or mechanism of action, and despite current dogma, it appears possible that viral replication can be retarded by inhibiting or otherwise down- regulating the activity of ion channels expressed in the host cell.
  • the negative impact of the compounds of the present invention on viral replication may be mediated by the inhibition or otherwise down-regulation of a membrane ion channel 0 relied upon by the virus for repHcation.
  • This membrane ion channel may be a viral membrane ion channel (exogenous to the host cell) or a host cell ion channel induced as a result of viral infection (endogenous to the host cell).
  • the compounds f the present invention may inhibit Vpu or p7 function and thereby inhibit the continuation of the respective HIV or HCV life cycle.
  • the SARS virus encodes an E protein which is shown for the first time, by the present inventors, to act as an ion channel.
  • E proteins are present in other coronaviruses, the compounds, compositions and methods of the present invention would have utility in the inhibition and/or treatment of infections by other coronaviruses.
  • the present invention is concerned with novel antiviral compounds that fall under the classification of substituted acylguanidines.
  • a first aspect of the present invention provides an acylguanidine with antiviral activity.
  • the present invention provides an antiviral compound of Formula I
  • R4 7 wherein t -1 ⁇ are independently aromatic groups, heteroaromatic groups, alkylaromatic groups, alkylheteroaromatic groups, alkenylaromatic groups, alkenylheteroaromatic groups, cycloalkylaromatic groups, cycloalkylheteroaromatic groups, aryloxyalkyl groups, heteroaryloxyalkyl groups, said groups are mono or polycyclic, and are optionally substituted with one or more substitutents independently selected from hydrogen, hydroxy, nitro, halo, amino, substituted amino, alkyl-substituted ammo, cycloalkyl-substituted amino, aryl-substituted amino, Ci.
  • the present invention provides an antiviral compound of Formula I
  • R 2 , 3 and R are independently hydrogen
  • X hydrogen, hydroxy, nitro, halo, Ci-ealkyl, halo-substituted Ci. ⁇ alkyloxy, phenyl, C t -ealkeneyl, C 3 . 6 cycloalkeneyl, C ⁇ alkeneoxy, or benzo; ⁇ Be, R ⁇ ,,Rfl f I , e, Rf.
  • the compounds of the invention include the following: 5-(N,N-hcxamethylene)a iloride comprising the structure
  • EIPA 5-(N-ethyl-N-isopropyl)amiloride
  • N-Benzyl -N'rtS.S-diamino- ⁇ -chioro-pyizme ⁇ -carbony ⁇ -guamdine comprising the structure
  • N-amidino-3,5-diamino-6-phemyl-2-pyrazinecarboxamide comprising the structure
  • N-amidino-3-am o-5-phenyl-6-chloro-2-pyrazinecarboxamide comprising the structure
  • Bodipy-FL Amiloride comprising the structure
  • N-(2-napthoyl)-N'-phenylguanidine comprising the structure
  • N,N'-bis(l-napthoyl)guanidme comprising the structure
  • N-Cinnamoyl-N' N'-dimethylguanidine comprising the structure
  • N,N'-Bis(amidino)napthalene-2,6-dica boxamide comprising the structure
  • N,N'-Bis(3-phenyl ⁇ ro ⁇ aaoyl)guamdine comprising the structure
  • the compounds of the invention are capable of reducing, retarding or otherwise inhibiting viral growth and/or replication.
  • the antiviral activity of the compounds of the invention is against viruses such as those belonging to the Lentivirus family, and the Coronovirus family family of viruses.
  • viruses such as Human Immunodeficiency Virus (HIV), Severe Acute Respiratory Syndrome virus (SARS), Mouse Hepatitis virus (), and Hepatitis C virus (HCV).
  • HIV Human Immunodeficiency Virus
  • SARS Severe Acute Respiratory Syndrome virus
  • HCV Hepatitis C virus
  • a pharmaceutical composition comprising an antiviral compound according to any one of the first, second or third aspects, and optionally one or more pharmaceutical acceptable carriers or derivatives, wherein said compound is capable of reducing, retarding or otherwise inhibiting viral growth and/or replication.
  • the antiviral activity of the compounds of the invention is against viruses such as those belonging to the Lentivirus family, and the Coronovirus family of viruses.
  • viruses such as Human Immunodeficiency Virus (HIV), Severe Acute Respiratory Syndrome virus (SARS), Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV) and Equine Arteritis Virus (EAV).
  • viruses such as Human Immunodeficiency Virus (HIV), Severe Acute Respiratory Syndrome virus (SARS), Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV) and Equine Arteritis Virus (EAV).
  • Coronaviruses hich can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1.
  • compositions of the invention may further comprise one or more known antiviral compounds or molecules.
  • a method for reducing, retarding or otherwise inhibiting growth and/or replication of a virus comprising contacting a cell infected with said virus or exposed to said virus with a compound according to any one of the first, second or third aspects.
  • the virus is from the Lentivirus family, or the Coronavirus family.
  • the virus is Human Immunodeficiency Virus (HIV), Severe Respiratory Syndrome virus (SARS), Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Mouse Hepatitis virus (MHV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV).
  • HIV Human Immunodeficiency Virus
  • SARS Severe Respiratory Syndrome virus
  • Human Coronavirus 229E Human Coronavirus OC43
  • MHV Mouse Hepatitis virus
  • BCV Bovine Coronavirus
  • PRCV Porcine Respiratory Coronavirus
  • MHV Mouse Hepatitis virus
  • HCV Hepatitis C virus
  • EAV Equine Arteritis Virus
  • the virus is HW-1, HIV-2, the SARS virus, Coronaviruse 229E, Coronavirus OC43, PRCV, BCV, HCV, or EAV.
  • Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1 ,
  • a method for preventing the infection of a cell exposed to a virus comprising contacting said cell with a compound according to any one of the first, second or third aspects.
  • the virus is from the Lentivirus family, or the Coronavirus family. More preferably, the virus is Human Immunodeficiency Virus (HIV), Severe Respiratory Syndrome virus (SARS), Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Mouse Hepatitis virus (MHV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, the virus is HJV-1, HIV-2, the SARS virus, Coronaviruse 229E, Coronavirus OC43, PRCV, BCV, HCV, EAV.
  • HCV Human Immunodeficiency Virus
  • SARS Severe Respiratory Syndrome virus
  • MHV Mouse Hepatitis virus
  • BCV Bovine Coronavirus
  • PRCV Porcine Respiratory Coronavirus
  • HCV Hepatitis C virus
  • EAV Equine Arteritis Virus
  • the virus is HJ
  • Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1.
  • a method for the therapeutic or prophylactic treatment of a subject infected with or exposed to a virus comprising the administration of a compound according to any one of the first, second or third aspects, to a subject in need of said treatment.
  • infection with a virus or exposure to a virus occurs with viruses belonging to the Lentivirus family, or the Coronovirus family. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with HlV-1, HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1.
  • the subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer).
  • livestock animal e.g. sheep, cow, horse, donkey, pig
  • companion animal e.g. dog, cat
  • laboratory test animal e.g. mouse, rabbit, rat, guinea pig, hamster
  • captive wild animal e.g. fox, deer.
  • the subject is a primate, or horse.
  • the subject is a human.
  • a method of down regulating a membrane ion channel functional activity in a cell infected with a virus comprismg contacting said cell with a compound according to any one of the first, second or third aspects,
  • the membrane ion channel may be endogenous to the cell or exogenous to the celL
  • the membrane ion channel of which functional activity is down ⁇ egulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral - replication and include, for example, the HIV membrane ion channel Vp , the HCV membrane ion channel P7, the Coronavirus E protein membrane ion channel, and the .
  • infection with a virus or exposure to a virus occurs with viruses belonging to the Lentivirus family, or the Coronovirus family. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with HIV-1, HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV).
  • a method of reducing, retarding or otherwise inhibiting growth and/or replication of a virus that has infected a cell comprising contacting said infected cell with a compound according to any one of the first, second or third aspects, wherein said compound down regulates functional activity of a membrane ion channel derived from said virus and expressed in said infected cell.
  • infection occurs with a virus belonging to tiie Lentivirus family, or the Coronovirus family.
  • infection or exposure occurs withHTV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV).
  • HTV Human Coronavirus 229E
  • Human Coronavirus OC43 Mouse Hepatitis virus (MHV)
  • BCV Bovine Coronavirus
  • PRCV Porcine Respiratory Coronavirus
  • HCV Hepatitis C virus
  • EAV Equine Arteritis Virus
  • the membrane ion channel of which functional activity is down regulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral replication and include, for example, the HIV membrane ion channel Vpu , the HCV membrane ion channel VI, and the Coronavirus E protein membrane ion channel.
  • the present invention provides a method of reducing, retarding or otherwise inhibiting growth and or replication of a virus that has infected a cell in a mammal, said method comprising administering to said mammal a compound according to any one of the first, second or third aspects, or a pharmaceutical composition according to the fourth aspect, wherein said compound or said composition down regulates functional activity of a membrane ion channel expressed in said infected cell
  • infection occurs with a virus belonging to the Lentivirus family, or the Coronovirus family. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with HIV-1, HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV).
  • HIV-1 HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV).
  • the membrane ion channel of which functional activity is down regulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral replication and include, for example, the HIV membrane ion channel Vpu , the HCV membrane ion channel P7, and the Coronavirus E protein membrane ion channel.
  • the subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer).
  • livestock animal e.g. sheep, cow, horse, donkey, pig
  • companion animal e.g. dog, cat
  • laboratory test animal e.g. mouse, rabbit, rat, guinea pig, hamster
  • captive wild animal e.g. fox, deer
  • the subjec is a primate, or horse.
  • the subject is a human.
  • the present invention provides a method for the therapeutic or prophylactic treatment of a subject infected with or exposed to a virus comprismg administering to said subject a compound according to any one of the first, second or third aspects, or a pharmaceutical composition according to the fourth aspect, wherein said compound or said composition down-regulates functional activity of a membrane ion channel derived from said virus.
  • infection occurs with a virus belonging to the Lentivirus family, or the Coronovirus family of viruses. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with EtfV-1, HTV-2, SARS " , Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1.
  • the membrane ion channel of which functional activity is down regulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral replication and include, for example, the HTV membrane ion channel Vpu , the HCV membrane ion channel P7, and the Coronavirus E protein membrane ion channel,.
  • the subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer).
  • livestock animal e.g. sheep, cow, horse, donkey, pig
  • companion animal e.g. dog, cat
  • laboratory test animal e.g. mouse, rabbit, rat, guinea pig, hamster
  • captive wild animal e.g. fox, deer.
  • the subject is a primate, or horse.
  • the subject is a human.
  • the invention provides an antiviral compound selected from the group consisting of:
  • Bodipy-FL amiloride 3-hydroxy-5-hexamefhyleneimino-amiloride
  • the present invention provides a pharmaceutical composition comprising a compound according to the twelfth aspect, and optionally one or more pharmaceutical acceptable carriers or derivatives.
  • the pharmaceutical composition may further comprise one or more known antiviral compounds or molecules.
  • Figure 1 is a schematic representation of plasmids used for expression of Vpu in E. coli.
  • A The amino acid sequence ( ⁇ 400 > 1) encoded by the vpu open reading frame (ORF) generated by PCR from an HIV- 1 strain HXB2 cDNA clone.
  • the vpu ORF was cloned in-frame at the 3' end of the GST gene in p2GEX to generate p2GEXVpu (B). It was subsequently cloned into pPL451 to produce the plasmid pPL + Vpu (Q.
  • FIG. 2 is a photographic representation of the expression and purification of Vpu in E. coli.
  • A Western blotting after SDS-PAGE was used to detect expressed Vpu in E. coli extracts. Lanes 1-4 contain samples, at various stages of purity, of Vpu expressed from p2GEXVpu: lane 1, GST-Vpu fusion protein isolated by glutathione-agarose affinity chromatography; lane 2, Vpu liberated from the fusion protein by treatment with thrombin; lane 3, Vpu purified by HPLC anion exchange chromatography; lane 4, Vpu after passage through the im unoaffinity column. Lanes 5 and 6, membrane vesicles prepared from 42'C induced cells containing pPL+Vpu or pPL451 , respectively. B. Silver stained SDS-PAGE gelt lane 1, Vpu purified by HPLC anion exchange chromatography; lane 2, Vpu after passage through the immunoaffinity column.
  • Figure 3 is a graphical representation of ion channel activity observed after exposure of lipid bilayers to aliquots containing purified Vpu.
  • the CIS chamber contained 500mM NaCl and the TRANS chamber contained 50mM NaCl; both solutions were buffered at pH 6.0 with 10 mM MES.
  • B shows a current versus voltage curve generated from data similar to that shown in A.
  • Figure 4 is a photographic representation of bacterial cross-feeding assays.
  • Met * , Pro auxotrophic strain was used to seed a soft agar overlay.
  • Plates A and B contain minimal drop-out medium minus proline; in plate C the medium was minus methionine.
  • the discs labelled P and M contained added proline or methionine, respectively.
  • the discs labelled C and V were inoculated with Met + , Pro + E, coli cells containing the plasmids ⁇ PL451 orpPL+Vpu, respectively.
  • FIG. 5 is a graphical representation of the screening of drugs for potential Vpu channel blockers. The photograph shows a section of a minimal medium-lacking adenine - agarose plate onto which a lawn of XLrl-blue E. coli cells containing the Vpu expression plasmid pPLVpu has been seeded.
  • Numbers 6-11 are located at the sites of application of various drugs being tested, which were applied in 3 ⁇ l drops and allowed to soak into the agarose. The plate was then incubated at 37°C for 4Shr prior to being photographed. The background grey shade corresponds to areas of no bacterial growth.
  • the bright circular area around " 10 " represents bacterial cell growth as a result of application of adenine at that location (positive control).
  • the smaller halo of bacterial growth around "9” is due to the application of 5-(HN- he ⁇ amethylene)-amiloride at that location.
  • FIG. 6 S RS E protein ion channel activity observed in NaCl solutions after exposure of lipid bi ⁇ ayer to 3-10 ⁇ g of E protein.
  • the CIS chamber contained 50mM NaCl in 5mM HEPES buffer pH 7.2
  • the TRANS chamber contained 500mM NaCl in 5mM HEPES buffer pH 7.2.
  • the CIS chamber was earthed and the TRANS chamber was held at various potentials between -100 to +1 OOmV.
  • FIG. 7 SARS E protein ion channel activity observed in NaCl solutions after exposure of lipid bilayer to 3-10 ⁇ g of E protein.
  • the CIS chamber contained 50n ⁇ M NaCl in 5mM HEPES buffer pH 7.2
  • the TRANS chamber contained 500mM NaCl in 5mM HEPES buffer pH 7.2.
  • the CIS chamber was earthed and the TRANS chamber was held at various potentials between -100 to +1 OOmV.
  • Cinnamoylgua ⁇ idine inhibits SARS E protein ion channel activity in NaCl solution.
  • A Representative currents at holding potential of-40mV, Scale bar is 30QmS and 5pA.
  • B All points histogram at holding potential of - 4QmV.
  • C Average current (pA), before formation of E protein ion channel, E protein ion channel activity and after addition of 1 OO ⁇ M Bit036.
  • Figure 10 Part A shows raw currents generated by the 229E-E protein ion channel in a planar lipid bilayer. The top trace shows current activity prior to drug addition and the lower trace shows the effect of addition of lOO ⁇ M cinnamoylguanidine on channel activity.
  • Part B is a graphical representation of the average current flowing across the bilayer (in arbitrary units), before and after addition of cinnamoylguanidine.
  • Figure 11 MHV E protein Ion channel activity in Hpid bilayers NaCl solutions.
  • Part A shows raw currents generated by the MHV-E protein ion channel in a planar lipid bilayer.
  • the top trace shows current activity prior to drug addition and the lower trace shows the effect of addition of lOO ⁇ M cinnamoylguanidine on channel activity
  • Part B is a graphical representation of the average current flowing across the bilayer (in arbitrary units), before and after addition of cinnamoylguanidine.
  • the present invention is based, in part, on the surprising determination that certain compounds that fall under the classification of substituted acylguanidines have antiviral activity against viruses from a range of different virus families.
  • the negative impact of the compounds of the present invention on viral replication may be mediated by the inhibition or otherwise down-regulation of a membrane ion channel rehed upon by the virus for replication.
  • This membrane ion channel may be a viral membrane ion channel (exogenous to the host cell) or a host cell ion channel induced as a result of viral infection (endogenous to the host cell).
  • the compounds of the present invention may inhibit Vpu or p7 function and thereby inhibit the continuation of the respective HIV or HCV life cycle.
  • the SARS virus encodes an E protein which is shown for the first time, by the present inventors, to act as an ion channel.
  • E proteins are present in other coronaviruses, the compounds, compositions and methods of the present invention would have utility in the inhibition and/or treatment of infections by other coronaviruses.
  • While the present invention is concerned with novel antiviral compounds falling under the classification of substituted acylguanidines, it does not include in its scope the use of compounds 5-(N,N-hexamethylene)amiloride and 5-(N,N-dimethyl)- amiloride for retarding, reducing or otherwise inhibiting viral growth and/or functional activity of HIV. It will be understood by those skilled in the art that the compounds of the invention may be administered in the form of a composition or formulation comprising pharmaceutically acceptable carriers and excipients.
  • compositions of the invention may further comprise one or more known antiviral compounds or molecules.
  • the known antiviral compounds are selected from the group consisting of Vidarabine, Acyclovir, Ganciclovir, Valgan clovir, Valacyclovir, Cidofovir, Fa ciclovir, Ribavirin, Amantadine, Rimantadine, Interferon, Oseltamivir, Palivizumab, Rimantadine, Zanamivir, nucleoside-analog reverse transcriptase inhibitors (NRTI) such as Zidovudine, Didanosine, Zalcitabine, Stavudine, Lamivudine and Abacavir, non- nucleoside reverse transcriptase inhibitors (NNRIT) such as Nevirapine, Delavirdine and Efav ⁇ renz, protease inhibitors such as Saquinavir, Ritonavir, Indinavir, Nelfinavir, Amprenavir, and
  • Canine enteric coronavirus (strain INSAVC-l)
  • Canine enteric coronavirus (strain K378)
  • Feline enteric coronavirus (strain 79-1683)
  • Bovine coronavirus (strain LSU-94LSS-0S1)
  • Bovine coronavirus (STRAIN LY-138)
  • Bovine coronavirus (strain O .-0514-3)
  • Bovine coronavirus (STRAIN QUEBEC)
  • Bovine enteric coronavirus (strain 98TXSF-110-ENT)
  • Murine coronavirus strain DVBVt
  • Murine hepatitis virus (strain A59)
  • Murine hepatitis virus (strain S)
  • Murine hepatitis virus strain l Murine hepatitis virus strain l
  • Murine hepatitis virus strain 2 Murine hepatitis virus strain 2
  • Murine hepatitis virus strain 3 Murine hepatitis virus strain 3
  • Murine hepatitis virus strain 4 Murine hepatitis virus strain 4.
  • Porcme hemagglutinating encephalomyelitis virus (strain IAF-404) Puffinosis virus
  • Rat coronavirus (strain 681)
  • Rat coronavirus (strain NJ)
  • Bovine respiratory coronavirus (strain 98TXSF-110-LUN)
  • the present invention extends to ion channels which may function by means such as passive, osmotic, active or exchange transport.
  • the ion channel may be formed by intracellular or extracellular means.
  • the ion channel maybe an ion channel which is naturally formed by a cell to facilitate its normal functioning.
  • the ion channel ma be formed by extracellular means.
  • Extracellular means would include, for example, the formation of ion channels due to introduced chemicals, drugs or other agents such as ionophores or due to the functional activity of viral proteins encoded by a virus which has entered a cell.
  • the ion channels which are the subject of certain embodiments of the present invention facilitate the transport of ions across membranes.
  • Said membrane may be any membrane and is not limited to Ihe outer cell wall plasma membrane.
  • membrane encompasses the membrane surrounding any cellular organelle, such as the Golgi apparatus and endoplasmic reticulum, the outer cell membrane, the membrane surrounding any foreign antigen which is located within the cell (for example, a viral envelope) or the membrane of a foreign organism which is located extracellularly.
  • the membrane is typically, but not necessarily, composed of a fluid lipid bilayer.
  • the subject ion channel may be of any structure.
  • the Vpu ion channel is foiflied by Vpu which is an integral membrane protein encoded by HTV-1 which associates with, for example, the Golgi and endoplasmic reticulum membranes of infected cells.
  • Vpu ion channels is a reference to all related ion channels for example P7 HCV and M2 cf influenza and the like.
  • HAV HTV
  • SARS SARS
  • Coronavirus coronavirus
  • HCV HCV
  • references to the "functional activity" of an ion channel should be understood as / a reference to any one or more of the functions which an ion channel performs or is involved in.
  • the Vpu protein encoded ion channel in addition to facilitating the transportation of Na + , K + , CI * and P0 3" , also plays a role in the degradation of the CD4 molecule in the endoplasmic reticulum.
  • the Vpu protein encoded ion channel is also thought to play a role in mediating the HIV life cycle.
  • the present invention is not limited to treating HIV infection via the mechanism of inhibiting the HIV life cycle and, in particular, HIV replication. Rather, the present invention should be understood to encompass any mechanism by which the compounds of the present invention exert fheir anti-viral activity and may include inhibition of HIV viability or functional activity. This also applies to HCV, Coronaviruses, and to other viruses.
  • Ion channel mediation of viral replication may be by direct or indirect means. Said ion channel mediation is by direct means if the ion channel interacts directly with the virion at any one or more of its life cycle stages. Said ion channel mediation is indirect if it interacts with a molecule other than those of the virion, which other molecule either directly or indirectly modulates any one or more aspects or stages of the viral life cycle. Accordingly, the method of the present invention encompasses the mediation of viral replication via the induction of a cascade of steps which lead to the mediation of any one or more aspects or stages of the viral life cycle.
  • references to "down-regulating' ion channel functional activity should be understood as a reference to the partial or complete inhibition of any one or more aspects of said activity by both direct and indirect mechanisms.
  • a suitable agent may interact directly with an ion channel to prevent replication of a virus or, alternatively, may act indirectly to prevent said replication by, for example, interacting with a molecule other than an ion channel.
  • a further alternative is that said other molecule interacts with and inhibits the activity of the ion channel.
  • a "cell” infected with a virus should be understood as a reference to any cell, prokaryotic or eukaryotic, which has been infected with a virus. This includes, for example, immortal or primary cell lines, bacterial cultures and cells in situ.
  • the preferred infected cells would be macrophages monocytes or hepatocytes/lymphoid cells infected with either HIV or HCV respectively.
  • the compounds of the present invention are thought to inhibit viral replication or virion release from cells by causing ion channels, namely VPU of HIV, the E protein of SARS and other Coronaviruses, or P7 of HCV to become blocked.
  • the present invention encompasses antiviral compounds that are substituted acylguanidines.
  • the present invention also includes the use of compounds 5-(N,N- hexamethylene)amiloride and 5-(N,N-dimethyl)-amiloride in the control of viral replication and/or growth other than HTV.
  • the subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer).
  • livestock animal e.g. sheep, cow, horse, donkey, pig
  • companion animal e.g. dog, cat
  • laboratory test animal e.g. mouse, rabbit, rat, guinea pig, hamster
  • captive wild animal e.g. fox, deer
  • the method of the present invention is useful in the treatment and prophylaxis of viral infection such as, for example, but not limited to HTV infection, HCV infection and other viral infections.
  • the antiviral activity may be effected in subjects known to be infected with HIV in order to prevent replication of HIV thereby preventing the onset of AIDS.
  • the method of the present invention may be used to reduce serum viral load or to alleviate viral infection symptoms.
  • antiviral treatment may be effected in subjects known to be infected with, for example, HCV, in order to prevent replication of HCV, thereby preventing the further hepatocyte involvement and the ultimate degeneration of liver tissue.
  • the method of the present invention may be particularly useful either in the early stages of viral infection to prevent the establishment of a viral reservoir in affected cells or as a prophylactic treatment to be applied immediately prior to or for a period after exposure to a possible source of virus.
  • terin "prophylaxis” may be considered as reducing the severity of onset of a particular condition. Therapy may also reduce the severity of an existing condition or the frequency of acute attacks.
  • more than one compound or composition may be co-administered with one or more other compounds, such as known anti-viral compounds or molecules.
  • co- administered is meant simultaneous administration in the same formulation or in two different formulations via the same or different routes or sequential administration by the same or different routes.
  • sequential administration is meant a time difference of from seconds, minutes, hours or days between the administration of the two or more separate compounds.
  • the subject antiviral compounds may be administered in any order.
  • Routes of administration include but are not limited to intravenously, intraperitionealy, subcutaneously, intracranialy, intradermally, intramuscularly, intraocularly, intrathecaly, intracerebrally, intranasally, transmucosally, by infusion, orally, rectally, via iv drip, patch and implant. Intravenous routes are particularly preferred.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) and sterile powders for the extemporaneous preparation of sterile injectable solutions.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), suitable mixtures thereof and vegetable oils.
  • the prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thi ⁇ nerosal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by, for example, filter sterilization or sterilization by other appropriate means.
  • Dispersions are also contemplated and these may be prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • a preferred method of preparation includes vacuum drying and the fieeze-drying technique which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution.
  • the active ingredients may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets.
  • the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • compositions and preparations should contain at least 0.01 % by weight, more preferably 0.1 % by weight, even more preferably 1% by weight of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 1 to about 99%, more preferably about 2 to about 90 %, even more preferably about 5 to about 80% of the weight of the unit.
  • the amount of active compound in such therapeutically useful compositions in such that a suitable dosage will be obtained.
  • Preferred compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains between about 0.1 ng and 2000 mg of active compound.
  • the tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: A binder such as gum, acacia, com starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin may be added or a flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring.
  • a binder such as gum, acacia, com starch or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose or saccharin
  • a flavouring agent such as peppermint, oil of wintergreen, or
  • tablets, pills, or capsules may be coated with shellac, sugar or both.
  • a Syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavouring such as cherry or orange flavour. Any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compound(s) may be incorporated into sustained-release preparations and formulations.
  • the present invention also extends to forms suitable for topical application such as creams, lotions and gels.
  • the anti-clotting peptides may need to be modified to permit penetration of the surface barrier.
  • Procedures for the preparation of dosage unit forms and topical preparations are readily available to those skilled in the art from texts such as Pharmaceutical Handbook. A Martind ⁇ le Companion Volume Ed. Ainley Wade Nineteenth Edition The Pharmaceutical Press London, CRC Handbook of Chemistry and Physics Ed. Robert C. Weast Ph D. CRC Press Inc.; Goodman and G ⁇ lm n's; The Pharmacological basis ofTherapeutics. Ninth Ed McGraw Hill; Remington- and The Science and Practice of Pharmacy. Nineteenth Ed. Ed. Alfonso R. Gennaro Mack Publishing Co. Easton Pennsylvania.
  • Pharmaceutically acceptable carriers and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like.
  • the use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, use thereof in the therapeutic compositions is contemplated.
  • Supplementary active ingredients can also be incorporated into the compositions. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active material calculated.to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the specification for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved and (b) the limitations inherent in the art of compounding.
  • Effective amounts contemplated by the present invention will vary depending on the severity of the pain and the health and age of the recipient. In general terms, effective amounts may vary from 0.01 ng/kg body weight to about 100 g kg body weight.
  • Alternative amounts include for about 0. 1 ng kg body weight about 100 mg kg body weight or from 1.0 ng/kg body weight to about 80 mg kg body weight.
  • the subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer).
  • livestock animal e.g. sheep, cow, horse, donkey, pig
  • companion animal e.g. dog, cat
  • laboratory test animal e.g. mouse, rabbit, rat, guinea pig, hamster
  • captive wild animal e.g. fox, deer.
  • the subject is a human or primate. Most preferably, the subject is a human.
  • the methods of the present invention is useful in the treatment and prophylaxis of viral infection such as, for example, but not limited to HTV infection, HCV infection and other viral infections.
  • the antiviral activity may be. effected in subjects known to be infected with HIV in order to prevent replication of HIV thereby preventing the onset of AIDS.
  • the methods of the present invention may be used to reduce serum viral load or to alleviate viral infection symptoms.
  • antiviral treatment may be effected in subjects known to be infected with, for example, HCV, in order to prevent replication of HCV, thereby preventing the further hepatocyte involvement and the ultimate degeneration of liver tissue.
  • the methods of the present invention may be particularly useful either in the early stages of viral infection to prevent the establishment of a viral reservoir in affected cells or as a prophylactic treatment to be applied immediately prior to or for a period after exposure to a possible source of virus.
  • the compounds of the present invention may be made from the corresponding acid chlorides or methyl esters as shown in Scheme 1. Both of these methods are well described in the literature.
  • cDNA fragments for the various viral proteins listed in Table 2 were obtained either by PCR amplification from a parental virus genome clone, or by direct chemical synthesis of the polynucleotide sequence.
  • Vpu open reading frame encoding Vpu (Fig la) was amplified by PCR from a cDNA clone of an Nde I fragment of the HIV-1 genome (isolate HXB2, McFarlane Burnet Centre, Melbourne, Australia) as follows: Native PfU DNA polymerase (Stratagene; 0.035 U/ D) was chosen to catalyse the PCR reaction to minimise possible PCR introduced errors by virtue of the enzyme's proofreading activity.
  • the 5 sense, primer AGTAGGATCCATGCAACCTATACC ( ⁇ 400 > 2) introduces a BamHl site (underlined) for cloning in-frame with the 3' end of the GST gene in p2GEX (41).
  • This primer also repairs the start codon (bold T replaces a Q of the vpu gene which is a threonine codon in the HXB2 isolate.
  • TCTGGAATTLTACAGATCAT CAAC ( ⁇ 400 > 3) introduces an EcoRl site (underlined) to the other end of the PCR product to facilitate cloning.
  • the 268bp fragment was purified, digested with BamHl and EcoRl and ligated to p2GEX prepared by digestion with the same two enzymes.
  • the resultant recombinant plasmid is illustrated in Fig lb.
  • the entireVpu open reading frame and the BamHl and EcoRl ligation sites were sequenced by cycle sequencing, using the Applied Biosystems dye-terminator kit, to confirm the DNA sequence.
  • Other cDNAs were synthesised for us using state of the art methods by GenScript Corporation (New Jersey, USA). Codon sequences were optimised for expression in bacterial, insect or mammalian cells, as appropriate. Restriction endonuclease enzyme recognition sites were incorportated at the 5' and 3' ends of the synthetic cDNAs to facilitate cloning into plasmid expression vectors, ⁇ cDNA3.1, pFastBac and pPL451 for expression of the encoded virus proteins in mammalian, insect or bacterial cells, respectively.
  • p2GEXVpu was first digested with BamHl and the 5' base overhang was filled in the Klenow DNA poly erase in the presence of dNTPs.
  • the , Vpu-encoding fragment was then liberated by digestion with EcoRl, purified from an agarose gel and ligated into pPL451 which had been digested with Hpal and EcoRl.
  • Western blots subsequently confirmed that the pPLVpu construct (Fig lc) expressed Vpu after induction of cultures at 42°C to inactivate the cI857 repressor of the PR and PL promoters.
  • E. coli strain XU-blue cells containing p2GEXVpu were grown at 30°C with vigorous aeration in LB medium supplemented with glucose (6g L) and ampicillin (50mg L) to a density of approximately 250 Klett units, at which time IPTG was added to a final concentration of O.OhnM and growth was continued for a further 4hr.
  • the final culture density was approximately 280 Klett units.
  • the osmotically sensitised cells were pelleted at 12,000g and resuspended to the original volume in water to burst the cells.
  • the suspension was then made up to lxMTEBS/DTT using a lOx buffer stock and the ghosts were isolated by centrifugation and resuspended in MTPBS/DTT to which was then sequentially added glycerol (to 20 % wt vol) and CHAPS (to 2 % wt/vol) to give a final volume of one quarter the original volume. This mixture was stirred on ice for 1 hr and then centrifuged at 400,000g for lhr to remove insoluble material.
  • the GST-Vpu fusion protein was purified from the detergent extract by affinity chromatography on a glutathione agarose resin (Sigma). The resin was thoroughly washed in 50mM Tris pH 7.5 containing glycerol (5 %), DTT (ImM), and CHAPS (0.5 %) (Buffer A) and then the Vpu portion of the fusion protein was liberated and eluted from the resin-bound GST by treatment of a 50% (v v) suspension of the beads with human thrombin (lOOU/ml; 37°C for lhr). PMSF . (0.5mM) was added to the eluant to eliminate any remaining thrombm activity.
  • This Vpu fraction was further purified on a column of MA7Q anion exchange resin attached to a BioRad HPLC and eluted with a linear NaCl gradient (0-2M) in buffer A.
  • the Vpu was purified to homogeneity - as determined on silver stained gels - on an immunoaffinity column as follows: HPLC fractions containing Vpu were desalted on a NAP 25 column (Pharmacia) into buffer A and then mixed with the antibody- agarose beads for lhr at room temperature. The beads were washed thoroughly and Vpu was eluted by increasing the salt concentration to 2M. Protein was quantitated using the BioRad dye binding assay.
  • the plasmid p2GEXVpu (Fig. 1) was constructed to create an in-frame gene fbsion between the GST and Vpu open-reading frames. This system enabled IPTG-inducible expression of the Vpu polypeptide fUsed to the C-te ⁇ ninus of GST and allowed purification of the fusion protein by affinity chromatography on glutathione agarose. Optimal levels of GST-Vpu expression were obtained by growing the cultures at 30°C to a cell density of approximately 250-300 Klett units and inducing with low levels of IPTG (O.Ol M).
  • a combined cellular fraction containing the cell debris and plasma membrane was prepared by lysozyme treatment of the induced cells followed by a low-speed centrifugation. Approximately 50% of the GST-Vpu protein could be solubilised from this fraction using the zwitterionic detergent CHAPS. Affinity chromatography using glutathione-agarose beads was used to enrich the fusion protein and thrombm was used to cleave the fusion protein at the high affinity thrombin site between the fusion partners, liberating Vpu (Fig. 2A). In fractions eluted from the anion exchange column Vpu was the major protein visible on silver stained gels (Fig.2B, lane 1).
  • Vpu was purified to apparent homogeneity on an immunoaffinity column (Fig.2B, lane 2).
  • the N-terminal amino acid sequence of the protein band (excised from SDS-PAGE gels) corresponding to the immunodetected protem confirmed its identity as Vpu.
  • Proteoliposomes containing Vpu were prepared by the detergent dilution method (New, 1990). A mixture of lipids (PEiPC:PS; 5:3:2; lmg total lipid) dissolved in chloroform was dried under a stream of nitrogen gas and resuspended in 0.1 ml of potassium phosphate buffer (50mM pH 7.4) containing DTT (ImM). A 25 ⁇ l aliquot containing purified Vpu was added, followed by octylglucoside to a final concentration of 1.25 % (wt vol).
  • This mixture was subject to three rounds of freezing in liquid nitrogen, thawing and sonication in a bath type sonicator (20-30 sec) and was then rapidly diluted into 200 volumes of the potassium phosphate buffer.
  • Proteoliposomes were collected by centrifugation at 400,000g for lhr and resuspended in approximately 1 0 ⁇ l of phosphate buffer.
  • Vpu was tested for its ability to induce channel activity in planar lipid bilayers using standard techniques as described elsewhere (Miller, 1986; and Piller et al, 1996).
  • the solutions in the CIS and TRANS chambers were separated by a DelrinTM plastic wall containing a small circular hole of approximately lOO ⁇ m diameter across which a lipid bilayer was painted so as to form a high resistance electrical seal.
  • Bilayers were painted from a mixture (8:2) of palmitoyl-oleoly- phosphatidyl-ethanolamine and pahnitoyl-oleolyphosphatidyl-choline (Avanti Polar L ⁇ pids, Alabaster, Alabama) in n-decane.
  • the solutions in the two chambers contained MES buffer (lOmM, pH 6.0) to which various NaCl or KCl concentrations were added. Currents were recorded with an AxopatchTM 200 amplifier. The electrical potential between the two chambers could be manipulated between +/-200mV (TRANS relative to grounded CIS). Aliquots containing Vpu were added to the CIS chamber either as a detergent solution or after incorporation of the protein into phospholipid vesicles. The chamber was stined until currents were observed.
  • Example 10 Vpu Forms Ion Channels in Lipid Bilayers.
  • Channel activity was observed in over 40 individual experiments with Vpu samples prepared from five independent purifications. In different experiments, the amplitude of the currents varied over a large range and, again, seemed to approximately correlate with the amount of protein added. The smallest and largest channels measured had conductances of 14 pS and 280 pS, respectively. The channels were consistently smaller when lipid vesicles containing Vpu were prepared and fused to the bilayer rather than when purified protein in detergent solution was added. This may be because the former method included treatment with high concentrations of detergent and a dilution step that may have favoured the breakdown of large aggregates into monomers.
  • This bio-assay is based on the observation that expression of Vpu in E. coli results in an active Vpu channel located in the plasmalemma that dissipates the transmembrane sodium gradient.
  • a sodium dependent co-transporter for example proline or adenine
  • metabolites whose accumulation within the cells is mediated by a sodium dependent co-transporter (for example proline or adenine) leak out of the cell faster than they can be synfhesised so that the metabolites' intracellular levels become limiting for growth of the cell.
  • a sodium dependent co-transporter for example proline or adenine
  • the vpu open-reading frame was cloned into the plasmid pPL451 to create the recombinant plasmid pPL-Vpu (Fig. lb).
  • the strong P and P lambda promoters are used to drive expression of Vpu under control of the temperature sensitive cl857 represser, such that when grown at 30°C expression is tightly repressed and can be induced by raising the temperature to between 37*C and 42 ⁇ C.
  • the temperature sensitive cl857 represser such that when grown at 30°C expression is tightly repressed and can be induced by raising the temperature to between 37*C and 42 ⁇ C.
  • cells containing pPL-Vpu grew when incubated at 30°C and 37°C but not at 42°C, while control strains grew well at 42°C.
  • the plasma membrane fraction was prepared and western blotting, using an antibody that specifically binds to the C-terminus of Vpu, detected a single band at approximately l ⁇ fcDa, indicating that Vpu was expressed an associated with the membranes (Fig. 2A, lane 5).
  • Example 14 E.Coli Cells Expressing Vpn Require Adenine in the External Medium for Growth. It was observed that, due to an uncharacterised mutation in the adenine synthesis pathway, growth of E. coli cells of the XLI-blue strain expressing Vpu at 37°C was dependant on the presence of adenine in the medium.
  • Vpu N-terminal peptide (residues 1- 32) dissolved in trifluoroethanol was added to the CIS chamber of the bilayer apparatus and the solutions was stirred until ion currents were observed, indicating incorporation of one or more Vpu ion channels into the bilayer. After recording the channel activity for a few minutes, drugs were added to the solutions in the CIS and TRANS chambers - with stirring - to a final concentration of lOO ⁇ M.
  • Channel activity was then recorded for at least a further three minutes and the effect of drug addition on ion current was determined by comparing the channel activity before and after drug addition.
  • drug effect was classified into four categories: “Stong block”, if current was inhibited approximately 90-100%; “weak block”, approx. 50-90% inhibition; “partial block”, ⁇ 50%; and “no effect”.
  • Stong block if current was inhibited approximately 90-100%
  • “weak block” approx. 50-90% inhibition
  • Partial block ⁇ 50%
  • no effect Experiments were disregarded if currents larger than ⁇ 50pA were generated after . addition of Vpu N-peptide because in such cases it is possible that non-native peptide aggregates contribute to bilayer breakdown. Such aggregates, by virtue of their disorganized structure may not be specifically blocked by the drugs at the concentrations tested.
  • Example 16 Compound Screening using the Bacterial Bio-Assav for the Vpu protein.
  • halos of growth around the site of application of particular drugs - as described in example 14— were given a score between zero and six reflecting the size and density of the zone of bacterial cell growth. Scores greater than 3 represent strong inhibition of the Vpu protein; scores between 1.5 and 3 . represent moderate inhibition and scores between 0.01 and 1.5 represent fair inhibition.
  • Table 4 lists the scores for inhibition of Vpu protein in the bacterial bio-assay.
  • Human monocytes were isolated from peripheral blood and cultured either for 24hr (one day old monocytes) or for 7 days to allow differentiation into monocyte derived acrophages (MDM). These cells were then exposed to cell-free preparations of HIV isolates and allowed to absorb for 2hr before complete aspiration of the medium, washing once with virus-free medium and resuspension in fresh medium. The cells were exposed to various concentration of compound either 24 hr prior to infection or after infection. Subsequent HIV replication, at various times after infection, was compared in cells exposed to drugs and in cells not exposed to drugs (controls). The progression and extent of viral replication was assayed using either an HIV DNA PCR method (Fear et al, 1998) or an ELISA method to quantitate ⁇ 24 in culture supernatants (Kelly et al, 1998).
  • Table 5 provides examples of results obtained using this assay and test antiviral compounds.
  • Example IS SARS Coronavirus.
  • a peptide corresponding to the full-length SARS-CoV (isolate Tor2 and Urbani) E protein (MYSFVSEETGTLIVNSVLLFLAFVVi iVTLA ⁇ LTAlRLCA YCCNIVNVSLVKPTVYVYSRVKNLNSSEGVPDLLV) and a second peptide comprising the first 40 amino acids of the full length E protein which correspond to the transmembrane domain (MYSFVSEETGTLIVNSVLLFLAFVVF LLVTLAILTALRLC) were synthesized manually using FMOC chemistry and solid phase peptide synthesis The synthesis was done at the Biomolecular Resource Facility (John Curtin School of Medical Research, ANU, Australia) using a Symphony 11 Peptide Synthesiser from Protein Technologies fcic.(Tucson, AZ, USA) according to the manufacturers instructions.
  • Example 19 Peptide purification Mass spectral analysis of the synthetic peptide revealed that the preparation contained significant amounts of material with lower na/z ratio than expected for the full-length product. The majority of these are presumably truncated peptides generated during the peptide synthesis process. To enrich the full-length E protein, the following procedure was used, which relies on differential solubility of the smaller molecules and full-length peptide. The crude preparation was suspended at 12 mg ml in 70% CH3CN, 0.1 %TFA and droneexed for 10 minutes. This suspension was centrifiiged at 10,000g for 10 minutes at 20°C. The supernatant was discarded and the insoluble fractions was extracted with 70% CH 3 CN, 0.1 % TFA, as above, two .
  • the SARS virus E protein was resuspended at lmg/ l in 2,2,2-trifluoroethanoL
  • the SARS virus E protein's ability to form ion channels was tested on a Warner (Warner instruments, Inc. 1125 Dixwell Avenue, Hamden, CT 06 14) bilayer rig as follows; A lipidmix of 3:1:1, l-Palmitoyl-2-oleolyi phosphatidyl Ethanolamine: l-Pahnitoyl-2- oleolyl phosphatidyl Serine: l-Pahnitoyl-2-oleolyl phosphatidyl choline in CHC1 3 was dried under 2 gas and resuspended to 50mg ml in n-decane, Bilayers were painted across a circular hole of approximately lOO ⁇ m diameter in a DelrinTM cup separating aqueous solution in the CIS and TRANS chambers.
  • the CIS chamber contained a solution of 500mM NaCl or KCl, in a 5mM HEPES buffer pH 7.2
  • the TRANS chamber contained a solution of 50mM NaCl or KCl, in a 5mM HEPES buffer pH 7.2.
  • Silver electrodes coated in chloride with 2% agarose bridges are placed in the CIS and TRANS chamber solutions.
  • the SARS E protein full-length or N- terminal peptides (3-1 Ou ) were added to the CIS chamber, which was st ⁇ ed until channel activity was detected.
  • the CIS chamber was earthed and the TRANS chamber was held at various holding potentials ranging between +100 to -lOOmV.
  • cinnamoylguanidine (Bit036), a compound which was shown in earlier experiments to be antiviral and to inhibit ion channel proteins from other viruses.
  • Example 20 Polvacrylamide gel electrophoresis Purified E protein was dissolved to 1 mg/ml, 5 mg/ml and 10 mg ml in, 6 M
  • the purified synthetic peptide was reconstituted into planar lipid bilayers' (21).
  • 3 ⁇ g of SARS full-length E protein was added to the CIS chamber, while stirring.
  • This CIS chamber contained 500 mM NaCl and the TRANS chamber contained 50 mM NaCl.
  • ion currents due to SARS E protein ion channel activity were observed after about 5 -15 minutes of stirring. Activity was detected more rapidly and reliably with a holding potential of approximately —1 OOmV across the bilayer. Currents recorded at -lOOmV, (A) and at -60mV (B) in one of these experiments are shown in Figure 6.
  • Figure 7a shows typical current traces recorded over a range of potentials in NaCl solutions.
  • the IV curve shows that at the lower voltages the average current flow across the bilayer is small but at higher potentials there is an increase in average current across the bilayer, resulting in a non-linear IV relationship.
  • the average reversal potential was +48.3 + 2.3 mV (mean + 1SEM), indicating that the channels were about 37 times more permeable to Na+ ion than to CI " ions.
  • the reversal potential is close to the Na+ equilibrium potential (+53mV), therefore the channel is selective for Na+ ions.
  • SARS E protein N-terminal peptide also formed ion channels in KCl solution that were similarly selective for K+ ions compared to the full-length E protein.
  • the average channel reversal potential was +39.5 ⁇ 3.6 V, therefore the channel is about 11 times more permeable to + ions than CI " ions.
  • SDS-PAGE of the purified full-length E protein peptide showed bands corresponding to the full-length E protein (Data not shown). Larger bands of varying size up to about 20 kDa were detected, suggesting that SARS E protein may form homo- oligomers.
  • Example 21 SARS E protein ion channel is blocked by cinnamoylguanidine and other compounds
  • the average current across the bilayer was reduced to baseline by 1 OO ⁇ M cinnamoylguanidine.
  • 100 to 200 ⁇ M ci ⁇ namoulguanidine reduced the average current across the bilayer about 4 fold'.
  • 100 to 200 ⁇ M cinnamoylguanidine blocked channels formed by full-length E protein in KCl solutions.
  • E protein Ion channels were about 37 times more selective for Na+ ions over Cl-ions and about 7.2 times more selective for K+ ions over CI- ions. In over 60 experiments the Na+ conductance of the E protein ion channel varied from as low as 26 pS to as high as 164 pS.
  • the SARS virus full length E protem ion channel activity and N-terminal domain E protem ion channel activity on planar lipid bilayers in NaCl and KCl solutions was inhibited by addition of between lOO ⁇ M to 200 ⁇ M cinnamoylguanidine to the CIS chamber. Inhibition or partial, inhibition of the E protein ion channel activity by cinnamoylguanidine has been observed in seven independent experiments in NaCl solution and four independent experiments in KCl solution.
  • coronaviruses encode an E protein with a hydrophobic N-terminus transmembrane domain therefore all coronaviruses E proteins could form ion channels on planar lipid bilayers. This indicates that the E protein could be a suitable target for antiviral drugs and potentially stop the spread of coronavirus from infected host cells. Drugs that block the E protein ion channel could be effective antiviral therapy for the treatment of several significant human and veterinary coronavirus diseases including SARS and the common cold.
  • Example 22 Bacterial Bio-Assav for Screening Potential SARS-CoV E protein Ion Channel-Blocking Drugs. SARS-CoV E protein Ion Channel inhibits Bacterial Cell growth.
  • a bio-assay of SARS-CoV E protem function in bacterial cells was developed.
  • a synthetic cDNA fragment encoding SARS-CoV E protein was cloned into the expression plasmid ⁇ PL451, creating a vector in which E protein expression is . temperature inducible, as described in Example 4.
  • Inhibition of the growth of E.coli cells expressing E protein at 37°C was observed as an indicator of ⁇ 7 ion channel function dissipating the normal Na+ gradient maintained by the bacterial cells.
  • Example 23 Compound Screening using the Bacterial Bio-Assay for SARS coronavirus E protein.
  • Table 7 lists the scores for inhibition of SARS-CoV E protein in the bacterial bio- assay.
  • Example 24 SARS Antiviral Assay for testing compounds against replication of SARS co ⁇ mavir ⁇ s (SARS-CoV).
  • Example 25 Effect of compounds in SARS CoV antiviral assay
  • Table 8 provides Virus titration data presented as % of a control (SARS CoV grown for 48 hours in the absence of compounds).
  • a peptide corresponding to the full-length 229E-E protein (sequence: MFLKLVDDHALVVIWLLWCVVLIVILLVC ⁇ KLIKLCFTCHMFCNRTVYG ⁇
  • KNVYHIYQSYMHIDPFPKRVIDF accession number NP_073554
  • the synthesis was done at the Biomolecular Resource Facility (John Curtin School of Medical Research, ANU, Australia) using a Symphony* 1 Peptide Synthesiser from Protein Technologies Inc.(Woburn, MS, USA) according to the manufacturers instructions to give C-terminal amides, the coupling was done with HBTU and hydroxybenzotriazole in N-methylpyrrolidone. Each of the synthesis cycles used double coupling and a 4-fold excess of the amino acids. Temporary ⁇ -N Fmoc- protecting groups were removed using 20% piperidine in DMF.
  • the crude synthetic peptide was purified using the ProteoPIusTM kit (Qbiogene inc. CA), following manufactures instructions. Briefly, the peptides were diluted in loading buffer (60mM Tris-HCl pH 8.3, 6M urea, 5% SDS, 10% glycerol, 0.2% Bromophenol blue, + 100 mM ⁇ -tnercaptoethanol) and run on 4-20% gradient polyacrylamide gels (Gradipore, NSW, Australia) in tris-glycine electrophoresis buffer (25 mM Tris, 250 M glycine, 0.1% SDS). The peptides were stained with gel code blue (Promega, NSW) and the bands corresponding to the full-length peptide were excised out of the gel.
  • loading buffer 60mM Tris-HCl pH 8.3, 6M urea, 5% SDS, 10% glycerol, 0.2% Bromophenol blue, + 100 mM ⁇ -tnercaptoethanol
  • the gel slice was transferred to the ProteoPLUSTM tube and filled with tris-glycine electrophoresis buffer.
  • the tubes were emerged in tris-glycine electrophoresis buffer and subjected to 100 volts for approximately 1 hour. The polarity of the electric current was reversed for 1 minute to increase the amount of protein recovered.
  • the peptides were harvested and centrifuged at 13, 000 rp for 1 minute. The purified peptides were dried in a Speedvac and the weight of the final product was used to calculate the yield.
  • Examnle 27.229E-E protein forms ion channels in planar lipid bflavers.
  • Lipid bilayer studies were performed as described elsewhere (Sunstrom, 1996; Miller, 1 86).
  • the lipid mixture was painted onto an aperture of 150-200 ⁇ m in the wall of a 1 ml deidn cup, The aperture separates two chambers, cis and trans, both containing salt solutions at different concentrations.
  • the cis chamber was connected to ground and the trans chamber to the input of an Axopatch 200 amplifier. Normally the cis chamber contained either 500mMNaClor500mM Cl an thetrans 50 mMNaCl or50mMKCl.
  • the bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis.
  • the synthetic peptide was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 5000 Hz and steered on magnetic disk.
  • the 229E E synthetic peptide was dissolved in 2,2,2-trifluorethanol (TFE) at 0.05mg ml to 1 mg ml. 10 ⁇ l of this was added to the cis chamber (1ml aqueous volume) of the bilayer apparatus, which was stirred via a magnetic "flea". Ionic currents, indicating channel activity in the bilayer, were typically detected within 15- 30 min. After channels were detected the holding potential across the bilayer was varied between -l ⁇ OmV and +100mV to characterise the size and polarity of current flow and enable the reversal potential to be determined.
  • TFE 2,2,2-trifluorethanol
  • the graph is a representative plot of average bilayer current (pA; y-axis) versus holding potential (mV; x-axis).
  • Example 28 Chemical compounds inhibit the ion channel activity of the 229E E protein synthetic peptide.
  • Compound stock solutions were typically prepared at 500 mM in DMSO. This solution was further diluted to 50 M, or lower concentration in 50% DMSO/50% methanol and 2 ⁇ l of the appropriately diluted compound was added to the cis and/or trans chambers to yield the desired final concentration.
  • Example. 29 Bacterial Bio-Assay for Screening Potential 229E-CoV E protein Ion Channel-Blocking Drugs.
  • 229E-CoV E-protein Ion Channel inhibits Bacterial Cell growth.
  • a bio-assay of 229E-CoV E-protein function in bacterial cells was developed.
  • a synthetic cDNA fragment encoding 229E-CoV E-protein was cloned into the expression plasmid ⁇ PL451, creating a vector in which E protein expression is temperature inducible, as described in Example 4.
  • Inhibition of the growth of Kcoli cells expressing E protein at 37°C was observed as an indicator of p7 ion channel function dissipating the normal Na+ gradient maintained by the bacterial cells.
  • Example 30 Compound Screening using the Bacterial Biq-Assay for 229E-CoV E-protein.
  • Table 9 list the scores for inhibition of 229E-CoV E-protein in the bacterial bio-assay. Table 9
  • Example 31 Antiviral Assay for testing compounds against replication of human coronavirus 229E (229E1
  • an ELISA assay was developed measuring the release of the viral N-protein into culture supernatants from monolayers of OC43- infected MRC-5 cells (human lung fibroblasts ;ATCC CCL-171): First, a virus working stock was prepared by amplification in MRC-5 cells. This was then used to infect confluent monolayers of MRC-5 cells grown in 6-well tissue culture plates by exposure to the virus at an MOI of approx. 0.01 pfu/cell for 1 hour at 35°C in 5%C ⁇ 2 .
  • the infective inoculum was removed and replaced with fresh medium (DMEM supplemented with 10% fetal calf serum) containing various test concentrations of compounds or the appropriate level of solvent used for the compounds (control). Plates were subsequently incubated at 35°C (in 5% CO 2 ) for 5 days post infection, after which time culture supernatant was harvested and cellular debris removed by centrifugation at 5000 x g for 10 minutes. For N-antigen detection, lOO ⁇ l samples of clarified culture supernatant were added to duplicate wells of a 96-well Maxi-Sorb plate; lOO ⁇ l of RIPA buffer was added per well with mixing and the plate was covered and incubated at 4°C overnight to enable protein binding to the plastic wells.
  • fresh medium DMEM supplemented with 10% fetal calf serum
  • Example 34 Mouse Hepatitis Virus (MHV).
  • VLSPSIYLYDRSKQLYKYYNEEMRLPLLEVDDI accession number NP_068673
  • Each of the synthesis cycles used double coupling and a 4-fold excess of the amino acids.
  • Temporary ⁇ -N F oc- protecting groups were removed using 20% piperidine in DMF.
  • the crude synthetic peptide was purified using the ProteoPlusTM kit (Qbiogene inc. CA), following manufactures instructions. Briefly, the peptides were diluted in loading buffer (60mM Tris-HCl H 8.3, 6M urea, 5% SDS, 10% glycerol, 0.2% Bromophenol blue, ⁇ 100 mM ⁇ -mercaptoethanol) and run on 4-20% gradient polyacrylamide gels (Gradipore, NSW, Australia) in tris-glycine electrophoresis buffer (25 mM Tris, 250 M glycine, 0.1 % SDS). The peptides were stained with gel code blue (Promega, NSW) and the bands corresponding to the full-length peptide were excised out of the gel.
  • loading buffer 60mM Tris-HCl H 8.3, 6M urea, 5% SDS, 10% glycerol,
  • the gel slice was transferred to the ProteoPLUSTM tube and filled with tris- glycine electrophoresis buffer.
  • the tubes were emerged in tris-glycine electrophoresis buffer and subjected to 100 volts for approximately 1 hour.
  • the polarity of the electric curreait was reversed for 1 minute to increase the amount of protein recovered.
  • the peptides were harvested and centrifiiged at 13, 000 rp for 1 minute.
  • the purified peptides were dried in a Speedvac and the weight of the final product was used to calculate the yield.
  • Example 35 MHV-E protein forms ion channels in planar lipid bilayers.
  • the cis chamber was connected to ground and (he trans chamber to the input of an Axopatch 200 amplifier. Normally the cis chamber contained either 500 mM NaCl or 500mM KCl and the trans 50 mM NaCl or 50mM KCl. The bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis. The synthetic peptide was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 5000 Hz and stored on magnetic disk.
  • the MHV E synthetic peptide was dissolved in 2,2,2-trifluorethanol (TFE) at 0.05mg ml to 1 mg ml. 10 ⁇ l of this was added to the cis chamber (1ml aqueous volume) of the bilayer apparatus, which was stirred via a magnetic "flea". Ionic currents, indicating channel activity in the bilayer, were typically detected within 15- 30 min. After channels were detected the holding potential across the bilayer was varied between -lOOmV and +100mV to characterise the size and polarity of current flow and enable the reversal potential to be determined.
  • TFE 2,2,2-trifluorethanol
  • Figure 11 shows examples of raw current data for the MHV E ion channel at various holding potentials (cis relative to trans) in asymmetrical NaCl solutions (500/50 M).
  • the graph is a representative plot of average bilayer current (pA; y- axis) versus holding potential (mV; x-axis).
  • Example 36 Chemical compounds inhibit the ion channel actrvity of the MHV F. protein synthetic peptide.
  • Example 37 Bacterial Bio-Assay for Screening Potential MHV E-protein Ion Channel-Blocking Drugs.
  • MHV E-protein Ion Channel inhibits Bacterial Cell growth.
  • a bio-assay of MHV E-protein function in bacterial cells was developed.
  • a synthetic cDNA fragment encoding MHV E-protein was cloned into the expression plasmid pPL451, creating a vector in which E protein expression is temperature inducible, as described in Example 4.
  • Inhibition of the growth of E.coli cells expressing E protein at 37°C was observed as an indicator of p7 ion channel function dissipating the normal Na+ radient maintained by the bacterial cells.
  • Example 38 Compound Screening using the Bacterial Bio-Assav for MHV E protein.
  • Table 12 lists the scores for inhibition of MHV E protein in the bacterial bio-assay.
  • Example 39 MHV Antiviral Assay for testing compounds against replication of mouse hepatitis virus (MHV).
  • the infective inoculum was removed and replaced with fresh medium (DMEM supplemented with 10% horse serum) containing various test concentrations of compounds or the appropriate level of solvent used for the compounds (control). Plates were subsequently incubated at 37°C (in 5% CO2) for 16 - 24 hours post infection, after which time culture supernatant was removed and the cells were stained with 0.1% crystal violet solution in 20% ethanol for 10 minutes. Plaques were counted in all wells and the percentage reduction in plaque number compared to solvent control was calculated. Measurements were performed in duplicate to quadruplicate wells.
  • Example 40 Effect of compounds in MHV antiviral assay.
  • Table 13 provides the results obtained from this study.
  • Example 41 Porcine Respiratory Coronavims (PRCV) Antiviral Assay for testing compounds against replication of porcine respiratory coronavirns fPRCV).
  • PRCV Porcine Respiratory Coronavims
  • luM, luM, lOuM and 20uM Either duplicates or quadruplicates were performed at each concentration. Controls were performed where the same volume of solvent was. added to the overlay. The overlay was allowed to set at room temp for 20 ins. The plates were then incubated at 37°C for 2 days. The monolayers were then fixed and stained overnight at room temperature by adding Iml/well of 0.5% methylene blue, 4% formaldehyde. Overlay agarose and stain was then rinsed off to visualize stained and fixed monolayer
  • the viral supernatant was removed and 2ml well of overlay containing 1% Seaplaque agarose in lx MEM, 5% FCS was added to each well.
  • Compounds to be tested were added to the overlay mixture by diluting the compounds from a 0.5M frozen stock to a concentration so that the same volume of compound/solvent would be added to the overlay for each concentration of compound. The volume of compound/solvent never exceeded 0.07% of the volume of the overlay.
  • the solvent used to dissolve compounds was DMSO and raiethanol mixed in equal proportions.
  • Compounds were tested for anti-plaque forming activity at four concentrations, O.luM, luM, lOuM and 20uM. Quadruplicates were performed at each concentration.
  • Controls were performed where the same volume of solvent was added to the overlay.
  • the overlay was allowed to set at room temp for 20 mins.
  • the plates were then incubated at 37°C for 7 days.
  • the monolayers were then fixed and stained by adding Iml well of 0.5% methylene blue, 4% formaldehyde.
  • a peptide mimicking the protein P7 encoded by the hepatitis C virus (HCV) 5 was synthesised having the following amino acid sequence:
  • phosphatidylserme and palmitoyl-oleoyl-phosphatidylcholine 5:3:2) (Avanti Polar Lipids, Alabaster, Alabama) was used.
  • the lipid mixture was painted onto an aperture of 150-200 m in the wall of a 1 ml dehrin cup.
  • the aperture separates two chambers, s and trans, both containing salt solutions at different concentrations.
  • the cis chamber was connected to ground and the trans chamber to the input of an Axopatch
  • the cis chamber contained 500 mM KCl and the trans 50 mM KCl.
  • the bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis.
  • the protein was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 0 2000 Hz and stored on magnetic disk.
  • the P7 peptide was dissolved in 2,2,2- trifluorethanol (TFE) at lGmg ml. 10 ul of this was added to the cis chamber of the bilayer which was stirred. Channel activity was seen within 15-20 min.
  • TFE 2,2,2- trifluorethanol
  • the channels formed by the P7 peptide were blocked by 5-(N,N- hexamethylene) amiloride (HMA),
  • cDp7.coli Two cDNA fragments, each encoding the same polypeptide corresponding to the amino acid sequence of the HCV-la p7 protein, were synthesised commercially by GeneScript.
  • the two cDNAs differed in nucleotide sequence such that in one cDNA (“cDp7.coli") the codons were optimised for expression of the p7 protein in . E.coli while in the other cDNA ( ⁇ cDp7,mar ⁇ )" codons were biased for expression in mammalian cell lines.
  • cD ⁇ 7.coli was cloned into the plasmid pPL451 as a BamHI/EcoRI fragment for expression in E.coli.
  • cDp7.mam was cloned into vectors (for example, pcDNA3.1 vaccinia virus, pfasfBac-1) for expression of ⁇ 7 in mammalian cell lines.
  • Example 47 Role of p7 in enhancement of Gag VLP Budding.
  • VLP virus-like particles
  • Example 48 Assay of the ability of compounds to inhibit p7 ion channel functional activity.
  • Example 33 The two methods of detecting p7 ion channel functional activity, described in Examples 33-35, were employed to assay the ability of compounds to inhibit the p7 channel.
  • compounds were tested for their abihty to inhibit p7 channel activity in planar lipid bilayers.
  • Example 35 compounds were tested for their ability to reduce the number ofVLPs released from cells expressing both p7 and HTV-l Gag. Example 49.
  • HCV D7 Ion Channel inhibits Bacterial Cell growth.
  • a bio-assay of p7treatment in bacterial cells was developed.
  • the p7-encoding synthetic cDNA fragment cDp7.coli was cloned into the expression plasmid pPL451, creating the vector pPLp7, in which p7 expression is temperature inducible, as described in Example 4.
  • Inhibition of the growth of E.coli cells expressing p7 at 37'C was observed as an indicator of p7 ion channel function dissipating the normal Na+ gradient maintained by the bacterial cells.
  • Example 50 Compound Screening using the Bacterial Bio-Assav for HCV p7 protein.
  • Table 16 lists the scores for inhibition of HCV ⁇ 7 protein in the bacterial bio-assay.
  • Phenamil methanesulfonate salt 0.5 /1
  • EAV equine arteritis virus
  • strain Bucyrus ATCC VR-796
  • ATCC CCL-10 EAV infected BHK-21 cells
  • a virus stock was amplified in RK-13 cells (ATCC CCL-37) and this was then used to infect confluent monolayers of BHK-21 cells grown in 6-well tissue .
  • Example 54 Incorporation of Dengue M virus protein into lipid bilayers. Lipid bilayer studies were performed as described elsewhere (Sunstrom, 1996;
  • the cis chamber contained 500 M KCl and the trans 50 mM KCl.
  • the bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis.
  • the protein was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 5000 Hz and stored on magnetic disk.
  • DMVC dengue virus M protein C-terminal peptide
  • TFE 2,2,2- trifluorethanol
  • E ⁇ am ⁇ le 55 Hexamethylene amiloride (HMA) to inhibits ion channel activity of the dengue virus M protein C-termfnal peptide.
  • HMA Hexamethylene amiloride
  • Solutions of 50 mM HMA were prepared by first making a 500 inM solution in DMSO. This solution was further diluted to 50 mM HMA using 0.1 M HCI.2 ⁇ l of the 50 mM HMA was added to the cis chamber after channel activity was seen. The cis chamber contained 1 ml of solution making the final concentration of HMA 100 ⁇ M.
  • Example 56 Antiviral Assay for testing compounds against Effects of Dengne flavivirns against cvtoproliferation.
  • the cultures were allowed to grow for 7 days and then Alamar Blue, a fluorescent dye that measures the metabolism of the cultures (red/ox), was added to each culture and the fluorescence value for each culture was measured.
  • the negative control without experimental compound or virus was fixed at 100%.
  • the positive controls and the cultures with compound were scored by calculating their average fluorescence as a percentage of the negative control. At least six replicate wells were measured for each experimental condition.
  • Example 58 Positive correlation between Bacterial Assay and Anti-viral Assays
  • the p24-antige ⁇ data for twelve compounds representing various substituted acylguanidines was compared with the activity scores obtained for those compounds in the Vpu bacterial assay.
  • the data from each assay was initially rank ordered for effectiveness.
  • the rank order for the Vpu bacterial assay was determined from all activity scores, the highest score indicating the greatest effectiveness.
  • the rank order for the anti-HIV-1 assay was determined based on the overall average value of p24 antigen measured in culture sup ⁇ matants at all of the drug concentrations tested, with the lowest score indicating the greatest effectiveness.
  • the two rank orders generated ware then compared statistically by generating the Spearman's Rank correlation coefficient.
  • Table 19a Comparison of Rank order of efficacy of 12 substituted acylguanidines in the Vpu bacterial assay and anti-HTV assay.
  • MHV plaque reduction activity data for 96 compounds screened were sorted from greatest to least percent plaque reduction and rank orders were assigned to the list of compounds. This was performed for the data generated by exposure to both lO ⁇ M and 1 ⁇ M concentrations of the compounds, giving rise to two rank order lists. Similarly, a rank order list was generated for the MHVE bacterial bioassay scores for the same 96 compounds. Where one or more compounds had the same score, the rank values for that group were averaged.
  • the rank order comparison of 96 compounds assayed in the bacterial bioassay and the antiviral assay show that MHVE bacterial assay rank order for the compounds tested is significantly positively correlated with the rank orders generated by the MHV plaque reduction assay.
  • the significant correlation between the assays is highly indicative that either assay may be utilised to identity compounds that may be useful.
  • the bacterial assay may thereby be a useful tool in screening for compounds that exhibit anti-viral activity.
  • 229E plaque reduction activity data for 97 compounds screened against 2.5 ⁇ M compound concentration were sorted from greatest to least percent plaque reduction and rank orders were assigned to the list of compounds.
  • a rank order list was generated for the 229E E bacterial bioassay scores for the same 97 compounds. Where one or more compounds had the same score, the rank values for that group were averaged.
  • Wcstcrvclt P., Hcnkcl, T., Trowbridgc, D.B., Orcnstcin, J., Heuser, , Gendclman,

Abstract

The invention relates to acylguanidine compounds having antiviral activity and methods utilising the compounds to treat viral infections.

Description

ANTIVIRAL COMPOUNDS AND METHODS
Έ__U> OF INVENTION
The present invention relates to methods for retarding, reducing or otherwise inhibiting viral growth and/or functional activity. The invention also relates to compounds and compositions suitable for use in the methods.
BACKGROUND OF THE INVENTION
Currewtly, there is a great need for the development of new treatments that are effective against viral infections, particularly against viral infections which are associated with high morbidity and mortality, and which impact on sizable populations. Treatments currently available are inadequate or ineffective in large proportions of infected patients.
For example, in ameliorating AIDS symptoms and prolonging life expectancy, a measure of success has been achieved with drugs targeting the viral reverse transcriptase and protease enzymes (Miller and Sarver, 1997; Mitsuya, 1992; Moore,
1997; and Thomas and Brady, 1997). However no single treatment method is completely effective against HIV infection. (Barry et al, 1998; Deeks, 199S; Miles,
1997; Miles, 199S; Moyle et al, 1998; Rachlis and Zarowny, 1998; Veil et al, 1 97;
Volberding and Deeks, 1998; and Voϊberdin, 1998). PCT application PCT/AU99/00872 describes the use of compounds 5-(N,N- hexamethylene)-amil ϊide and 5-(N,N-dime1hyl)-amiloride in the treatment of HIV infection.
Another virus considered to be a significant human pathogen is the Hepatitis C virus (HCV). This is a significant human pathogen in terms of both cost to human health and associated economic cosfs. HCV causes chronic hepatitis and cirrhosis and is the leading indicator for liver replacement surgery. In 2002 the Centre for
Disease Control and Prevention estimated that more' than 4 million people were infected in the USA alone and that approximately 8,000 to 1 , 000 die as a result of chronic HCV infection yearly. There is no known cure or vaccine. More effective pharmacological agents are urgently required. A fiirther well-known family of pathogenic viruses are the Coronaviruses. Coronaviruses (Order Nidσvir les. family Coronaviridae, Genus Coronavirus) are enveloped positive-stranded RNA viruses that bud from the endoplasmic reticulum- Golgi intermediate compartment or the erø-Golgi network (Fischer, Stegen et al. 1998; Maeda, Maeda et al. 1999i Corse and Machamer 2000; Maeda, Repass et al. 2001 ; Ruo and Masters 2003)
Coronaviruses infect humans and animals and it is thought that there could be a coronavirus that infects every animal. The two human coronaviruses, 229E and OC43, are known to be the major causes of the common cold and can occasionally cause pneumonia in older adults, neonates, or immimocomprømised patients (Peiris, Lai et al.2003). Animal coronaviruses can cause respiratory, gastrointestinal, neurological, or hepatic diseases in their host (Peiris, Lai et al. 2003). Several animal coronavirus are significant veterinary pathogens (Rota, Oberste et al. 2003).
Severe acute respiratory syndrome (SARS) is caused by a newly identified virus. SARS is a respiratory illness that has recently been reported in Asia, North America, and Europe (Peiris, Lai et al.2003). The causative agent of SARS was identified as a coronavirus. (Drosten, Gunther et al.2003; Ksiazek, Erd an et al. 2003; Peiris, Lai et al.2003). The World Health Organization reports that the cumulative number of reported probable cases of SARS from 1 November 2002 to the 11th July 2003 is 8,437 with 813 deaths, nearly a 10% death rate. It is believed that SARS will not be eradicated, but will cause seasonal epidemics like the.cold or influenza viruses (Vogel 2003).
To improve the prospect of treating and preventing viral infections, there is an on-going need to identify molecules capable of inhibiting various aspects of the viral life cycle.
It is an object of the present invention to overcome or ameliorate at least one of the disadvantages of the prior art, or to provide a useful alternative.
Any discussion of the prior art throughout the specification should in no way be considered as an admission that such prior art is widely known or forms part of common general knowledge in the field. SUMMARY OF THE INVENTION
The inventors have surprisingly found that certain compounds that fall under the classification of substituted acylguanidines have antiviral activity against viruses from a range of different virus families, Without intending to be bound by any $ particular theory or mechanism of action, and despite current dogma, it appears possible that viral replication can be retarded by inhibiting or otherwise down- regulating the activity of ion channels expressed in the host cell. Thus, the negative impact of the compounds of the present invention on viral replication may be mediated by the inhibition or otherwise down-regulation of a membrane ion channel 0 relied upon by the virus for repHcation. This membrane ion channel may be a viral membrane ion channel (exogenous to the host cell) or a host cell ion channel induced as a result of viral infection (endogenous to the host cell).
As an example, the compounds f the present invention may inhibit Vpu or p7 function and thereby inhibit the continuation of the respective HIV or HCV life cycle. 5 The SARS virus encodes an E protein which is shown for the first time, by the present inventors, to act as an ion channel. As similar E proteins are present in other coronaviruses, the compounds, compositions and methods of the present invention would have utility in the inhibition and/or treatment of infections by other coronaviruses. The present invention is concerned with novel antiviral compounds that fall under the classification of substituted acylguanidines. It does not include in its soope the use of compounds 5-(N,N-hexamethylene)amiloride and 5-(N, -dimethyl)- amiloride for retarding, reducing or otherwise inhibiting viral growth and/or functional activity of HIV. Accordingly, a first aspect of the present invention provides an acylguanidine with antiviral activity.
According to a second aspect, the present invention provides an antiviral compound of Formula I
.R3 0 N
H I
R4 7 wherein t-1^ are independently aromatic groups, heteroaromatic groups, alkylaromatic groups, alkylheteroaromatic groups, alkenylaromatic groups, alkenylheteroaromatic groups, cycloalkylaromatic groups, cycloalkylheteroaromatic groups, aryloxyalkyl groups, heteroaryloxyalkyl groups, said groups are mono or polycyclic, and are optionally substituted with one or more substitutents independently selected from hydrogen, hydroxy, nitro, halo, amino, substituted amino, alkyl-substituted ammo, cycloalkyl-substituted amino, aryl-substituted amino,
Figure imgf000005_0001
Ci. βalkyloxy, C3-6cycloalkyl, halo-substituted Ct^alkyl, halo-substituted Ci- βalkyloxy.phenyl, Ci-galkeneyl, C3.6cycloalkeneyl, Cι.6alkeιieoxy, benzo,
aryl, substituted aryl, PrS,
Figure imgf000005_0002
According to a third aspect, the present invention provides an antiviral compound of Formula I
Figure imgf000005_0003
or pharmaceutically acceptable salts thereof, wherein,
Rι =
Figure imgf000005_0004
Figure imgf000006_0001
R2 , 3 and R are independently hydrogen,
Figure imgf000006_0002
and wherein X = hydrogen, hydroxy, nitro, halo, Ci-ealkyl,
Figure imgf000007_0001
Figure imgf000007_0002
halo-substituted Ci. βalkyloxy, phenyl, Ct-ealkeneyl, C3.6cycloalkeneyl, C^alkeneoxy, or benzo; Be, R<,,RflfI , e, Rf. h^Rc L^ wjRή Ro^ independently = hydrogen, amino, halo, Ci-salkyl, Ci-salkyloxy, hydroxy, aryl, substituted aryl, substituted amino, mono or dialkyl-substituted amino, cycloalkyl-substituted amino, aryl-substituted amino,
Figure imgf000007_0003
Rg , Rj independently = hydrogen, hydroxy, halo, or C1.5 alkyl; j = hydrogen, amino, halo, Cι,5alkyl, Ci-jalkyloxy, hydroxy, aryl, substituted aryl, substituted amino, alkyl-s bstituted amino, cycloalkyl-substituted amino, aiyl-substituted amino, PrS,
Figure imgf000007_0004
Preferably, the compounds of the invention include the following: 5-(N,N-hcxamethylene)a iloride comprising the structure
Figure imgf000007_0005
Figure imgf000008_0001
hydrochloride comprising the structure
Figure imgf000008_0002
5-(N-methyl-N-isobutyl)amiloride comprising the structure
Figure imgf000008_0003
5-(N-ethyl-N-isopropyl)amiloride (herein referred to as EIPA), comprising the structure
Figure imgf000008_0004
N-(3,5-Diamino-6-chloro-pyrazine-2-carbonyl)-N4-phenyl-guanidine, comprising the structure
Figure imgf000009_0001
N-Benzyl -N'rtS.S-diamino-ό-chioro-pyizme^-carbony^-guamdine, comprising the structure
Figure imgf000009_0002
3-methoxy amiloride comprising the structure comprising the structure
Figure imgf000009_0003
3-methoxy-5-(N,N-Hexamethylene>amiloride comprising the structure
Figure imgf000009_0004
3-hydroxy-5-hexamethyleneimino-amiloride comprising the structure
Figure imgf000010_0001
Hexameth^yleneimino-6-phenyl-*2-ρyraxinecarboxamide comprising the structure
Figure imgf000010_0002
N-amidino-3,5-diamino-6-phemyl-2-pyrazinecarboxamide comprising the structure
Figure imgf000010_0003
5-(N,N-hexamethylene)amiloride comprising the structure
Figure imgf000011_0001
N-amidino-3-am o-5-phenyl-6-chloro-2-pyrazinecarboxamide comprising the structure
Figure imgf000011_0002
3*4 DichloroBenzamil comprising the structure
Figure imgf000011_0003
-II-
2'4 DichloroBenzamil HCI comprising the structure
Figure imgf000012_0001
5-(N-mefhyl-N-guanidinocarbonyl-methyl)amiloride comprising the structure
Figure imgf000012_0002
5-(N,N-Diethyl)amiloride hydrochloride comprising the structure
Figure imgf000012_0003
5-(N,N-Dimethyl)amiloride hydrochloride comprising the structure
Figure imgf000012_0004
5-tert-butylamino-amiloride comprising the structure
Figure imgf000013_0001
6-Iodoamiloride comprising the structure
Figure imgf000013_0002
Bodipy-FL Amiloride comprising the structure
Figure imgf000013_0003
5-(4-fluorophenyl)amiloride comprising the structure
Figure imgf000013_0004
1-napthoylguanidine comprising the structure
Figure imgf000014_0001
2-napthoylguanidine comprising the structure
Figure imgf000014_0002
N-(2-napthoyl)-N'-phenylguanidine comprising the structure
Figure imgf000014_0003
NJ T-bis(2-napthoyI)guanidine comprising the structure
Figure imgf000014_0004
N,N'-bis(l-napthoyl)guanidme comprising the structure
Figure imgf000015_0001
N,N'-bis(2-naρthoyl)-N"-phenylguaπidine comprising the structure
Figure imgf000015_0002
ό-meώoxy-2-naρhώoylguanidine comprising the structure
Figure imgf000015_0003
N-Cinnamoyl-N' N'-dimethylguanidine comprising the structure
Figure imgf000015_0004
3-quinolinoylguanidine comprising the structure
Figure imgf000016_0001
cinnamoylguanidine comprising the structure
Figure imgf000016_0002
4-ρhenylbenzoylguanidine comprising the structure
Figure imgf000016_0003
N-(citmamoyl)-N'phenylguanidine comprising the structure
Figure imgf000016_0004
(3-phenylpropanoyl)guanidine comprising the structure
Figure imgf000017_0001
N,N'-bis-(cinnamoyl)-N"-phenylguanidine comprising the structure
Figure imgf000017_0002
TSf-(3-phenylpropanoyl)-TS -phenylguanidine comprising the structure
Figure imgf000017_0003
NsN*-bis(3phenylρropanoyl>N"-phenylguanidine comprising the structure
Figure imgf000017_0004
trans-3-furanacryoylguanidine comprising the structure
Figure imgf000018_0001
N-(6-Hydroxy-2-napthoyl)-N'-phenylguamdine comprising the structure
Figure imgf000018_0002
(4-Phsnoxybenzoyl)guanidine comprising the structure
Figure imgf000018_0003
N,N'-Bis(amidino)napthalene-2,6-dica boxamide comprising the structure
Figure imgf000018_0004
6-bromo-2-napthoylguanidine comprising the structure
Figure imgf000019_0001
l-bromo-2-napthoylguaπidine comprising the structure
Figure imgf000019_0002
2-(2-naρthyl)acetoylguanidine comprising the structure
Figure imgf000019_0003
N"-Cinnamoyl-N,N'-diphenylguanidfoe comprising the structure
Figure imgf000019_0004
(Phenylacetyl)guamdine comprising the structure
Figure imgf000019_0005
N,N'-Bis(3-phenylρroρaaoyl)guamdine comprising the structure
Figure imgf000020_0001
Benzoylguanidine comprising the structure
Figure imgf000020_0002
(4-Chlorophenoxy-acetyl]guanidine comprising the structure
Figure imgf000020_0003
N-Benzoyl-N'-cinnamoylguanidine comprising the structure
Figure imgf000020_0004
[(E)-3-(4-Dimethylaιninophenyl)-2-methylacryloyl]guanidine comprising the structure
Figure imgf000020_0005
(4-Chlorocinnamoyl)guanidine comprising the structure
Figure imgf000021_0001
(4-Bromocinnamoyl)guanidine comprising the structure
Figure imgf000021_0002
(4-Methoxyoinnamoyl)guaπidine comprising the structure
Figure imgf000021_0003
(S-Phenyl-penta-2,4-dienoyl)guanidine comprising the structure
Figure imgf000021_0004
(3-Bromocinnamoyl)guanidine comprising the structure
Figure imgf000021_0005
(3-Methoxycinnamoyl)guanidine comprising the structure
Figure imgf000021_0006
(3-Chlorocinnamoyl)guanidine comprising the structure
Figure imgf000022_0001
(2-Chlorocinnamoyl)guanidine comprising the structure
Figure imgf000022_0002
(2-Brotnocinnamoyl)guanidine comprising the. structure
Figure imgf000022_0003
(2-Methoxycinnamoyl)guanidine comprising the structure
the structure
Figure imgf000022_0004
[3-(3-Pyridyl)acryloyl]guanidme comprising the structure
Figure imgf000022_0005
(4-Hydroxycinnamoyl)guanidine comprismg the structure
Figure imgf000023_0001
(Quinoline-2-carbonyI)guanidine comprismg the structure
Figure imgf000023_0002
(4-Nitrocinaamoyl)guanidme comprising the structure
Figure imgf000023_0003
(3-Nitrocinnamoyl)guanidine comprising the structure
Figure imgf000023_0004
(2-Nitrociπnamoyl)guanidine comprising the structure
Figure imgf000023_0005
(α-Methylcinnamoyl)guanidine comprising the structure
Figure imgf000024_0001
trans-3-(l-napthyl)acryloylguanidine comprising the structure
Figure imgf000024_0002
4-phenylcinnamoylguanidine comprising the structure
Figure imgf000024_0003
3-(trifluoromethyl)cinnamoylguanidine comprising the structure
Figure imgf000024_0004
-methylcinnamoylguanidine comprising the structure
Figure imgf000024_0005
4-(trifluoromethyl)cinnamoylguaπidine comprising thfe structure
Figure imgf000025_0001
2-methylcinnamoylguaradine comprising the structure
Figure imgf000025_0002
2-(trifluoromethyl)cinnamoylguanidine comprising the structure
Figure imgf000025_0003
4-mefhylcinnamoylguanidine comprising the structure
Figure imgf000025_0004
4-isopropyIcinnamoylguanidine comprismg the structure
Figure imgf000025_0005
-fluorocinnamoylguanidine comprising the structure
Figure imgf000026_0001
-fluorocinnamoylguanidine comprising the structure
Figure imgf000026_0002
-fluorocinnamoylguanidine comprising the structure
Figure imgf000026_0003
,4-dichlorocinnamoylguanidine comprising the structure
Figure imgf000026_0004
,4-dichlorocinnamolyguanjidine comprising the structure
Figure imgf000026_0005
2,6-dichlorocinnamoyiguanidine comprismg the structure
Figure imgf000027_0001
4-ethoxycinnamoylguanidine comprising the structure
Figure imgf000027_0002
3,4~(methylenedioxy)cinnamoylguanidine comprising the structure
Figure imgf000027_0003
3-(2-napthyl)acryloylguanidine comprising the structure
Figure imgf000027_0004
4-t-butylcinnamoylguanidine comprising the structure
Figure imgf000027_0005
3,4,5-trimethoxyciπnamoylguanidine comprising the structure
Figure imgf000028_0001
2-(l-napthyl)acetoylguanidine comprismg the structure
Figure imgf000028_0002
2,5-dimethylcinnamoylguanidine comprising the structure
Figure imgf000028_0003
2,3-difiuoroctnnamoylguanidine comprising the structure
Figure imgf000028_0004
3-phenylcinnamoylguanidine comprising the structure
Figure imgf000028_0005
-(trans-hept-l-en-l-yl)cinnamoylguanidine comprising the structure
Figure imgf000029_0001
-ethylcinnamoylguanidine comprising the structure
Figure imgf000029_0002
-chloro-6-fluorσcinnamoylguaπidine comprising the structure
Figure imgf000029_0003
-t-butylcinnamoylguanidine comprising the structure
Figure imgf000029_0004
3,4-difluorocinnamoylguanidine comprising the structure
Figure imgf000030_0001
5-bromo-2-fluorocinna oylguanidine comprising the structure
Figure imgf000030_0002
3-(trifluoromethoxy)cinnamoylguanidine comprising the structure
Figure imgf000030_0003
2-ethoxycinnamoylguanidine comprising the structure
Figure imgf000030_0004
2-t-butylcinnamoylguanidine comprising the structure
Figure imgf000030_0005
15 3-(cyclohex-l-en-l-yl)cinnamoylguanidine comprising the structure
Figure imgf000031_0001
cinnamoylguanidine hydrochloride comprising the structure
Figure imgf000031_0002
2,3,5,6,-tetramethylcinnamoylguanidine comprising the structure (Bit 134)
Figure imgf000031_0003
2-cyclohexylcinnamoylguanidine comprising the structure
Figure imgf000031_0004
5-bromo-2-methoxycinnamoylguanidine comprising the structure
Figure imgf000031_0005
,3-dimethylciπnamoylguamdine comprising the structure
Figure imgf000032_0001
-ethoxycinnamoylguanidine comprising the structure
Figure imgf000032_0002
-isoproρylciπnamoylguanidine hydrochloride comprising the structure
Figure imgf000032_0003
-pheuylciimamoylguanidine comprising the structure
Figure imgf000032_0004
-(cyclohex-l-en-lyl)cinnamoylguanidine comprising the structure
Figure imgf000032_0005
2,4,6-trimethylcinnamoylguanidine comprising the structure
Figure imgf000033_0001
(5-Phenyl-ρenta-2,4-dienoyl)guanidine comprismg the structure
Figure imgf000033_0002
5-(3'-bromophenyl)paita-2,4-dienoylguanicline comprising the structure
Figure imgf000033_0003
5-(2'-bromophenyl)penta-2,4-dienoyiguanidine comprismg the structure
Figure imgf000033_0004
Furanacryloyl comprising the structure
Figure imgf000033_0005
Preferably, the compounds of the invention are capable of reducing, retarding or otherwise inhibiting viral growth and/or replication.
Preferably, the antiviral activity of the compounds of the invention is against viruses such as those belonging to the Lentivirus family, and the Coronovirus family family of viruses. For example, the compounds of the invention exhibit antiviral activity against viruses such as Human Immunodeficiency Virus (HIV), Severe Acute Respiratory Syndrome virus (SARS), Mouse Hepatitis virus (), and Hepatitis C virus (HCV).
According to a fourth aspect of the present invention, there is provided a pharmaceutical composition comprising an antiviral compound according to any one of the first, second or third aspects, and optionally one or more pharmaceutical acceptable carriers or derivatives, wherein said compound is capable of reducing, retarding or otherwise inhibiting viral growth and/or replication.
Preferably, the antiviral activity of the compounds of the invention is against viruses such as those belonging to the Lentivirus family, and the Coronovirus family of viruses. For example, the compounds of the invention exhibit antiviral activity against viruses such as Human Immunodeficiency Virus (HIV), Severe Acute Respiratory Syndrome virus (SARS), Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV) and Equine Arteritis Virus (EAV).
Other Coronaviruses hich can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1.
The compositions of the invention may further comprise one or more known antiviral compounds or molecules.
According to a fifth aspect, there is provided a method for reducing, retarding or otherwise inhibiting growth and/or replication of a virus comprising contacting a cell infected with said virus or exposed to said virus with a compound according to any one of the first, second or third aspects. Preferably, the virus is from the Lentivirus family, or the Coronavirus family.
More preferably, the virus is Human Immunodeficiency Virus (HIV), Severe Respiratory Syndrome virus (SARS), Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Mouse Hepatitis virus (MHV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, the virus is HW-1, HIV-2, the SARS virus, Coronaviruse 229E, Coronavirus OC43, PRCV, BCV, HCV, or EAV.
Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1 ,
According to a sixth aspect, there is provided a method for preventing the infection of a cell exposed to a virus comprising contacting said cell with a compound according to any one of the first, second or third aspects.
Preferably, the virus is from the Lentivirus family, or the Coronavirus family. More preferably, the virus is Human Immunodeficiency Virus (HIV), Severe Respiratory Syndrome virus (SARS), Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Mouse Hepatitis virus (MHV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, the virus is HJV-1, HIV-2, the SARS virus, Coronaviruse 229E, Coronavirus OC43, PRCV, BCV, HCV, EAV.
Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1. According to a seventh aspect of the invention, there is provided a method for the therapeutic or prophylactic treatment of a subject infected with or exposed to a virus, comprising the administration of a compound according to any one of the first, second or third aspects, to a subject in need of said treatment.
Preferably, infection with a virus or exposure to a virus occurs with viruses belonging to the Lentivirus family, or the Coronovirus family. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with HlV-1, HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1.
The subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer). Preferably, the subject is a primate, or horse. Most preferably, the subject is a human.
According to a eighth aspect, there is provided a method of down regulating a membrane ion channel functional activity in a cell infected with a virus, comprismg contacting said cell with a compound according to any one of the first, second or third aspects,
. The membrane ion channel may be endogenous to the cell or exogenous to the celL
Preferably, the membrane ion channel of which functional activity is down τegulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral - replication and include, for example, the HIV membrane ion channel Vp , the HCV membrane ion channel P7, the Coronavirus E protein membrane ion channel, and the . SARS E protein membrane ion channel
Preferably, infection with a virus or exposure to a virus occurs with viruses belonging to the Lentivirus family, or the Coronovirus family. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with HIV-1, HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV).
According to anninth aspect of the present invention, there is provided a method of reducing, retarding or otherwise inhibiting growth and/or replication of a virus that has infected a cell, said method comprising contacting said infected cell with a compound according to any one of the first, second or third aspects, wherein said compound down regulates functional activity of a membrane ion channel derived from said virus and expressed in said infected cell. Preferably, infection occurs with a virus belonging to tiie Lentivirus family, or the Coronovirus family. More preferably, infection or exposure occurs withHTV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with HIV-1, HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV),
Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1. Preferably, the membrane ion channel of which functional activity is down regulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral replication and include, for example, the HIV membrane ion channel Vpu , the HCV membrane ion channel VI, and the Coronavirus E protein membrane ion channel. According to an tenth aspect, the present invention provides a method of reducing, retarding or otherwise inhibiting growth and or replication of a virus that has infected a cell in a mammal, said method comprising administering to said mammal a compound according to any one of the first, second or third aspects, or a pharmaceutical composition according to the fourth aspect, wherein said compound or said composition down regulates functional activity of a membrane ion channel expressed in said infected cell
Preferably, infection occurs with a virus belonging to the Lentivirus family, or the Coronovirus family. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with HIV-1, HIV-2, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV).
Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1. Preferably, the membrane ion channel of which functional activity is down regulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral replication and include, for example, the HIV membrane ion channel Vpu , the HCV membrane ion channel P7, and the Coronavirus E protein membrane ion channel.
The subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer). Preferably, the subjec is a primate, or horse. Most preferably, the subject is a human.
According to a eleventh aspect, the present invention provides a method for the therapeutic or prophylactic treatment of a subject infected with or exposed to a virus comprismg administering to said subject a compound according to any one of the first, second or third aspects, or a pharmaceutical composition according to the fourth aspect, wherein said compound or said composition down-regulates functional activity of a membrane ion channel derived from said virus.
Preferably, infection occurs with a virus belonging to the Lentivirus family, or the Coronovirus family of viruses. More preferably, infection or exposure occurs with HIV, SARS, Human Coronavirus 229E, Human Coronavirus OC43, Mouse Hepatitis virus (MHV), Bovine Coronavirus (BCV), Porcine Respiratory Coronavirus (PRCV), Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Most preferably, infection or exposure occurs with EtfV-1, HTV-2, SARS", Human Coronavirus 229E, Human Coronavirus OC43, Hepatitis C virus (HCV), or Equine Arteritis Virus (EAV). Other Coronaviruses which can be inhibited or their infections treated by the compounds of the invention are those listed in Table 1.
Preferably, the membrane ion channel of which functional activity is down regulated is that which Lentiviruses, and Coronaviruses utilise for mediating viral replication and include, for example, the HTV membrane ion channel Vpu , the HCV membrane ion channel P7, and the Coronavirus E protein membrane ion channel,.
The subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer). Preferably, the subject is a primate, or horse. Most preferably, the subject is a human. i
-38-
According to a twelfth aspect, the invention provides an antiviral compound selected from the group consisting of:
N-(3,5-l ammo-6-chloro-pyrazine-2-carbonyi)-N'-phenyl-guanidine,
N-Ben2yl-N'-(3,5-diamino-6-chlθϊθ-pyrazine-2-carbonyl)-guanidine, 3'4 DichloroBenzamil,
2'4 DichloroBenzamil,
5-(N-methyl-N-guanidinocarbonyl-methyl)amiloride,
5-(N-Methyl-N-isobutyl)amiloride,
5-(N-Ethyl-N-isopropyl)amiloride, 5-(N,N-Dimethyl)amiloride hydrochloride,
5-(N,N-hexamethylene)amiloride,
5-(N,N-Diethyl)amiloride hydrochloride,
6-Iodoamiloride,
Bodipy-FL amiloride, 3-hydroxy-5-hexamefhyleneimino-amiloride,
5-(4-fluorophenyl)amiloride,
5-tert-butylamino-amiloride,
N-aπύdinc^3-amino-5-phenyl-6-chloro-2-pyra necaιboxamide,
3-methoxy-5-(N,N-Hexamethylene)-amiloride, 3-methoxy-arailoride, hexamethyleneimino-6-phenyl-2-pyrazinecarboximide,
N-amidino-3,5-diamino-6-phenyl-2-pyrazinecarboxamide,
1-naphthoylguanidine,
2-naphthoylguanidine, N-(2-naphthoyl)-N1-phenylguanidine,
N,N,-bis(2-naphthoyl)guanidine,
N,N'-bis(l-naρhthoyl)guanidine,
N,N,-bis(2-naphthoyl)-N"-phenylguanidine, <
6-mefhoxy-2-naphthoylguanidine, 3-quinolinoylguanidine, cinnamoylguanidine,
4-phenylbenzoylguanidine, N-(cinnamoyl)-N'phenylguanidine,
(3-phenylpropanoyl)guanidine,
N,N'-bis-(cinnamoyl)-N"-pheιιylguanidine,
N-(3-phenylρropanoyl)-N,-phenylguanidine, N,:N'-bis(3phenylpropanoyϊ)-N"-ρhenylguanidine, trans-3-furanacryoylguanidine,
N-(6-Hydroxy-2-n^)hthoyl)-N'-phenylguamdine,
(4-Phenoxybenzoyl)guanidine,
N,N'-Bis(amidino)napthalene-2,6-dicarboxamide, ISP'-Cinnamoyl-NJSf'-diphenylguanidine,
(Pheήylacetyl)guanidine,
N^J'-Bis(3-phenylpropanoyl)guanidine, benzoylguanidine,
(4-Chlorophenoxy-acetyl]guanidine, N-benzoyl-N ciπnamoylguamdine,
[(E)-3-(4-Dimemylamincφhenyl)-2-memylacryloyl]guanidine,
(4-Chlorocinnamoyl)guanidine,
(4-Bromocinnamoyl)guanidine,
(Φ-Methoxyciπnamoyl)guanidine, (5-Phenyl-penta-2,4-dienoyl)guanidine,
(3-Bromochmamoyl)guanidine,
(3-Methoxycinnamoyl)guanidine,
(3-Chlorocinnamoyl)guamdine,
(2-Chlorocinnamoyl)guanidine, (2-BrDmocinnamoyl)guanidine,
(2-Methoxycinriamoyl)guaπidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
[3-(3-Pyridyl)acryloyl]guanidine,
(4-Hydroxycinnamoyl)guanidine, (Quinoline-2-carbonyl)guanidine, eutically acceptable salts thereof. According to a thirteenth aspect, the present invention provides a pharmaceutical composition comprising a compound according to the twelfth aspect, and optionally one or more pharmaceutical acceptable carriers or derivatives. ι
Preferably, the pharmaceutical composition may further comprise one or more known antiviral compounds or molecules.
Unless the context clearly requires otherwise, throughout the description and the claims, the words 'comprise', 'comprising', and the like are to be construed in an inclusive sense as opposed to an exclusive or exhaustive sense; that is to say, in the sense of "including, but not limited to",
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1 is a schematic representation of plasmids used for expression of Vpu in E. coli. A. The amino acid sequence ( < 400 > 1) encoded by the vpu open reading frame (ORF) generated by PCR from an HIV- 1 strain HXB2 cDNA clone. The vpu ORF was cloned in-frame at the 3' end of the GST gene in p2GEX to generate p2GEXVpu (B). It was subsequently cloned into pPL451 to produce the plasmid pPL + Vpu (Q.
Figure 2 is a photographic representation of the expression and purification of Vpu in E. coli. A. Western blotting after SDS-PAGE was used to detect expressed Vpu in E. coli extracts. Lanes 1-4 contain samples, at various stages of purity, of Vpu expressed from p2GEXVpu: lane 1, GST-Vpu fusion protein isolated by glutathione-agarose affinity chromatography; lane 2, Vpu liberated from the fusion protein by treatment with thrombin; lane 3, Vpu purified by HPLC anion exchange chromatography; lane 4, Vpu after passage through the im unoaffinity column. Lanes 5 and 6, membrane vesicles prepared from 42'C induced cells containing pPL+Vpu or pPL451 , respectively. B. Silver stained SDS-PAGE gelt lane 1, Vpu purified by HPLC anion exchange chromatography; lane 2, Vpu after passage through the immunoaffinity column.
Figure 3 is a graphical representation of ion channel activity observed after exposure of lipid bilayers to aliquots containing purified Vpu. In A and B, the CIS chamber contained 500mM NaCl and the TRANS chamber contained 50mM NaCl; both solutions were buffered at pH 6.0 with 10 mM MES. B shows a current versus voltage curve generated from data similar to that shown in A.
Figure 4 is a photographic representation of bacterial cross-feeding assays. For all plates, the Met*, Pro" auxotrophic strain was used to seed a soft agar overlay. Plates A and B contain minimal drop-out medium minus proline; in plate C the medium was minus methionine. To control for viability of the cells in the background lawn, the discs labelled P and M contained added proline or methionine, respectively. The discs labelled C and V were inoculated with Met+, Pro+ E, coli cells containing the plasmids ρPL451 orpPL+Vpu, respectively. Plates were incubated at 37°C (A and C) or 30dC (B) for two days and photographed above a black background with peripheral illumination from a fluorescent light located below the plate. The images were recorded on aNovaline video gel documentation system. light halos around the discs labelled P or M on all plates and around the disc labelled V on plate A indicate growth of the background lawn strain. Figure 5 is a graphical representation of the screening of drugs for potential Vpu channel blockers. The photograph shows a section of a minimal medium-lacking adenine - agarose plate onto which a lawn of XLrl-blue E. coli cells containing the Vpu expression plasmid pPLVpu has been seeded. Numbers 6-11 are located at the sites of application of various drugs being tested, which were applied in 3μl drops and allowed to soak into the agarose. The plate was then incubated at 37°C for 4Shr prior to being photographed. The background grey shade corresponds to areas of no bacterial growth. The bright circular area around " 10 " represents bacterial cell growth as a result of application of adenine at that location (positive control). The smaller halo of bacterial growth around "9" is due to the application of 5-(HN- heχamethylene)-amiloride at that location.
Figure 6. S RS E protein ion channel activity observed in NaCl solutions after exposure of lipid biϊayer to 3-10μg of E protein. A. The closed state is shown as solid line, openings are derivations from the line. Scale bar is 3 0ms and 5pA. The CIS chamber contained 50mM NaCl in 5mM HEPES buffer pH 7.2, the TRANS chamber contained 500mM NaCl in 5mM HEPES buffer pH 7.2. The CIS chamber was earthed and the TRANS chamber was held at various potentials between -100 to +1 OOmV. B. Largest single opening events of a single channel.
Figure 7. SARS E protein ion channel activity observed in NaCl solutions after exposure of lipid bilayer to 3-10μg of E protein. A. The closed state is shown as solid line, openings are derivations from the line. Scale bar is 300ms and 5pA. The CIS chamber contained 50nιM NaCl in 5mM HEPES buffer pH 7.2, the TRANS chamber contained 500mM NaCl in 5mM HEPES buffer pH 7.2. The CIS chamber was earthed and the TRANS chamber was held at various potentials between -100 to +1 OOmV. B. Largest single opening events of a single channel.
Figure 8. Cinnamoylguaπidine (Bit036) inhibits SARS E protein ion channel activity in NaCl solution. A, Representative currents at holding potential of-40mV, Scale bar is 30QmS and 5pA. E protein ion channel activity and E protein channel activity after the addition of lOOμM Bit036. B. All points histogram at holding potential of - 4QmV. E protein ion channel activity before and after the addition of lOOμM Bit036. C, Average current (pA), before formation of E protein ion channel, E protein ion channel activity and after addition of 1 OOμM Bit036.
Figure 9229E E protein Ion channel activity in lipid bilayers in KC1 solutions.
Figure 10: Part A shows raw currents generated by the 229E-E protein ion channel in a planar lipid bilayer. The top trace shows current activity prior to drug addition and the lower trace shows the effect of addition of lOOμM cinnamoylguanidine on channel activity. Part B is a graphical representation of the average current flowing across the bilayer (in arbitrary units), before and after addition of cinnamoylguanidine. Figure 11 : MHV E protein Ion channel activity in Hpid bilayers NaCl solutions.
Figure 12: Part A shows raw currents generated by the MHV-E protein ion channel in a planar lipid bilayer. The top trace shows current activity prior to drug addition and the lower trace shows the effect of addition of lOOμM cinnamoylguanidine on channel activity, Part B is a graphical representation of the average current flowing across the bilayer (in arbitrary units), before and after addition of cinnamoylguanidine.
DETAILED DESCRIPTION OF THE INVENTION
The present invention is based, in part, on the surprising determination that certain compounds that fall under the classification of substituted acylguanidines have antiviral activity against viruses from a range of different virus families. Without intending to be bound by any particular theory or mechanism of action, the negative impact of the compounds of the present invention on viral replication may be mediated by the inhibition or otherwise down-regulation of a membrane ion channel rehed upon by the virus for replication. This membrane ion channel may be a viral membrane ion channel (exogenous to the host cell) or a host cell ion channel induced as a result of viral infection (endogenous to the host cell).
As an example, the compounds of the present invention may inhibit Vpu or p7 function and thereby inhibit the continuation of the respective HIV or HCV life cycle. The SARS virus encodes an E protein which is shown for the first time, by the present inventors, to act as an ion channel. As similar E proteins are present in other coronaviruses, the compounds, compositions and methods of the present invention would have utility in the inhibition and/or treatment of infections by other coronaviruses. While the present invention is concerned with novel antiviral compounds falling under the classification of substituted acylguanidines, it does not include in its scope the use of compounds 5-(N,N-hexamethylene)amiloride and 5-(N,N-dimethyl)- amiloride for retarding, reducing or otherwise inhibiting viral growth and/or functional activity of HIV. It will be understood by those skilled in the art that the compounds of the invention may be administered in the form of a composition or formulation comprising pharmaceutically acceptable carriers and excipients.
The pharmaceutical compositions of the invention may further comprise one or more known antiviral compounds or molecules. Preferably, the known antiviral compounds are selected from the group consisting of Vidarabine, Acyclovir, Ganciclovir, Valgan clovir, Valacyclovir, Cidofovir, Fa ciclovir, Ribavirin, Amantadine, Rimantadine, Interferon, Oseltamivir, Palivizumab, Rimantadine, Zanamivir, nucleoside-analog reverse transcriptase inhibitors (NRTI) such as Zidovudine, Didanosine, Zalcitabine, Stavudine, Lamivudine and Abacavir, non- nucleoside reverse transcriptase inhibitors (NNRIT) such as Nevirapine, Delavirdine and Efavϊrenz, protease inhibitors such as Saquinavir, Ritonavir, Indinavir, Nelfinavir, Amprenavir, and other known antiviral compounds and preparations. Known antiviral compounds or molecules may in some cases act synesrgistically with the antiviral compounds of the invention.
Table 1 Known coronavirus isolates
Group 1 species
Canine coronavirus
Canine enteric coronavirus (strain INSAVC-l)
Canine enteric coronavirus (strain K378)
Feline coronavirus
Feline enteric coronavirus (strain 79-1683)
Feline infectious peritonitis virus (FIFV)
Human coronavirus 229E
Porcine epidemic diarrhea virus
Porcine epidemic diarrhea virus (strain Bri/87)
Porcine epidemic diarrhea virus (strain CV777)
Transmissible gastroenteritis virus
Porcine respiratory coronavirus
Porcine transmissible gastroenteritis coronavirus (STRAIN FS772/70)
Porcine transmissible gastroenteritis coronavirus (strain Miller)
Porcine transmissible gastroenteritis coronavirus (strain Neb72-RT)
Porcine transmissible gastroenteritis coronavirus (STRAIN PURDUE)!
Group 2 species
Bovine coronavirus
Bovine coronavirus (STRAIN F15)
Bovine coronavirus (strain G95)
Bovine coronavirus (STRAIN L9)
Bovine coronavirus (strain LSU-94LSS-0S1)
Bovine coronavirus (STRAIN LY-138)
Bovine coronavirus (STRAIN MEBUS)
Bovine coronavirus (strain O .-0514-3)
Bovine coronavirus (strain Ontario)
Bovine coronavirus (STRAIN QUEBEC)
Bovine coronavirus (STRAIN VACCINE)
Bovine enteric coronavirus (strain 98TXSF-110-ENT)
Canine respiratory coronavirus Chicken enteric coronavirus
Human coronavirus OC43
Murine hepatitis virus
Murine coronavirus (strain DVBVt)
Murine hepatitis virus (strain A59)
Murine hepatitis -virus (strain JHM)
Murine hepatitis virus (strain S)
Murine hepatitis virus strain l
Murine hepatitis virus strain 2
Murine hepatitis virus strain 3
Murine hepatitis virus strain 4
Murine hepatitis virus strain ML-11
Porcine hemagglutinating encephalomyelitis virus
Porcine hemagglutinating encephalomyelitis virus (strain 67N)
Porcme hemagglutinating encephalomyelitis virus (strain IAF-404) Puffinosis virus
Rat coronavirus
Rat coronavirus (strain 681)
Rat coronavirus (strain NJ)
Rat sialodacryoadenitis coronavirus
Group 3 species
Turkey coronavirus
Turkey coronavirus (strain Indiana)
Turkey coronavirus (strain Minnesota)
Turkey coronavirus (strain NC95)
Avian infectious bronchitis virus
Avian infectious bronchitis virus (STRAIN 6/82)
Avian infectious bronchitis virus (strain Arkansas 99)
Avian infectious bronchitis virus (strain Beaudette CK
Avian infectious bronchitis virus (strain Beaudette M42)
Avian infectious bronchitis virus (strain Beaudette US)
Avian infectious bronchitis virus (strain Beaudette)
Avian infectious bronchitis virus (strain D1466)
Avian infectious bronchitis virus (strain D274)
Avian infectious bronchitis virus (strain D3896)
Avian infectious bronchitis virus (strain D41)
Avian infectious bronchitis virus (strain DE072)
Avian infectious bronchitis virus (strain GRAY)
Avian infectious bronchitis virus (strain H120)
Avian infectious bronchitis virus (strain H52)
Avian infectious bronchitis virus (strain KB8523)
Avian infectious bronchitis virus (strain M41)
Avian infectious bronchitis virus (strain PQRTUGAL 322/82)
Avian infectious bronchitis virus (strain SAIB2Q) Avian infectious bronchitis virus (strain UK 123/82)
Avian infectious bronchitis virus (strain UK/142/86)
Avian infectious bronchitis virus (strain UK/1 7/84)
Avian infectious bronchitis virus (strain UK/183/66)
Avian infectious bronchitis virus (strain UK 68/84)
Avian infectious bronchitis virus (strain VI 8/91)
Avian infectious bronchitis virus (strain Vic S)
Avian infectious laryngotracheitis virus
[Preliminary Gronp 4 species
SARS coronavirus
SARS coronavirus Beijing ZY-2003
SARS coronavirus BJ01
SARS coronavirus BJ02
SARS coronavirus B J03
SARS coronavirus BJ04
SARS coronavirus CUHK-SulO
SARS coronavirus CUHK-W1
SARS coronavirus Frankfurt 1
SARS coronavirus GZ01
SARS coronavirus HKU-39849
SARS coronavirus Hong Kong ZY-2003
SARS coronavirus Hong Kong 03/2003
SARS coronavirus HSR 1
SARS coronavirus Sin2500
SARS coronavirus Sin2677
SARS coronavirus Sin2679
SARS coronavirus Sin2748
SARS coronavirus Sin2774
SARS coronavirus Taiwan
SARS coronavirus Taiwan JC-2003
SARS coronavirus Taiwan TCI
SARS coronavirus Taiwan TC2
SARS coronavirus Tor2
SARS coronavirus TW1
SARS coronavirus TWC
SARS coronavirus Urbani
SARS coronavirus Vietnam
SARS coronavirus ZJ-HZ01
SARS coronavirus ZJ01 unclassified coronaviruses
Bovine respiratory coronavirus (strain 98TXSF-110-LUN)
Human enteric coronavirus 4408
Enteric coronavirus
Equine coronavirus E uine coronavirus NC99
The present observations and findings now permit the use of agents such as certain substituted acylguanidines, as anti-viral agents for the therapy and prophylaxis of viral conditions caused by different viruses. The methods and compositions of the present invention may be particularly effective against viruses which rely on ion channel formation for their replication, however it will be understood that this is not the only mechanism .relied on by viruses for replication and that the compounds and methods of the present invention are not limited to agents which exert their action by - retarding or inhibiting the function of ion channels. Reference to "membrane ion channel" should be understood as a reference to a structure which transports ions across a membrane. The present invention extends to ion channels which may function by means such as passive, osmotic, active or exchange transport. The ion channel may be formed by intracellular or extracellular means. For example, the ion channel maybe an ion channel which is naturally formed by a cell to facilitate its normal functioning. Alternatively, the ion channel ma be formed by extracellular means. Extracellular means would include, for example, the formation of ion channels due to introduced chemicals, drugs or other agents such as ionophores or due to the functional activity of viral proteins encoded by a virus which has entered a cell. The ion channels which are the subject of certain embodiments of the present invention facilitate the transport of ions across membranes. Said membrane may be any membrane and is not limited to Ihe outer cell wall plasma membrane. Accordingly, "membrane" as used herein encompasses the membrane surrounding any cellular organelle, such as the Golgi apparatus and endoplasmic reticulum, the outer cell membrane, the membrane surrounding any foreign antigen which is located within the cell (for example, a viral envelope) or the membrane of a foreign organism which is located extracellularly. The membrane is typically, but not necessarily, composed of a fluid lipid bilayer. The subject ion channel may be of any structure. For example, the Vpu ion channel is foiflied by Vpu which is an integral membrane protein encoded by HTV-1 which associates with, for example, the Golgi and endoplasmic reticulum membranes of infected cells. Reference hereinafter to "Vpu ion channels" is a reference to all related ion channels for example P7 HCV and M2 cf influenza and the like.
Reference to "HIV", "SARS", "Coronavirus" or "HCV" should he understood as a reference to any HTV, SARS, Coronavirus or HCV virus strain and including homologues and mutants.
Reference to the "functional activity" of an ion channel should be understood as / a reference to any one or more of the functions which an ion channel performs or is involved in. For example, the Vpu protein encoded ion channel, in addition to facilitating the transportation of Na+, K+, CI* and P0 3", also plays a role in the degradation of the CD4 molecule in the endoplasmic reticulum. Without wishing to be bound by a particular theory, the Vpu protein encoded ion channel is also thought to play a role in mediating the HIV life cycle. The present invention is not limited to treating HIV infection via the mechanism of inhibiting the HIV life cycle and, in particular, HIV replication. Rather, the present invention should be understood to encompass any mechanism by which the compounds of the present invention exert fheir anti-viral activity and may include inhibition of HIV viability or functional activity. This also applies to HCV, Coronaviruses, and to other viruses.
Reference to the "functional activity" of a virus should be understood as a reference to any one or more of the fonctions which a virus perfoπns or is involved in.
Reference to the " viral replication" should be understood to include any one or more stages or aspects of the viral life cycle, such as inhibiting the assembly or release of virions. Ion channel mediation of viral replication may be by direct or indirect means. Said ion channel mediation is by direct means if the ion channel interacts directly with the virion at any one or more of its life cycle stages. Said ion channel mediation is indirect if it interacts with a molecule other than those of the virion, which other molecule either directly or indirectly modulates any one or more aspects or stages of the viral life cycle. Accordingly, the method of the present invention encompasses the mediation of viral replication via the induction of a cascade of steps which lead to the mediation of any one or more aspects or stages of the viral life cycle. Reference to "down-regulating' ion channel functional activity, should be understood as a reference to the partial or complete inhibition of any one or more aspects of said activity by both direct and indirect mechanisms. For example, a suitable agent may interact directly with an ion channel to prevent replication of a virus or, alternatively, may act indirectly to prevent said replication by, for example, interacting with a molecule other than an ion channel. A further alternative is that said other molecule interacts with and inhibits the activity of the ion channel.
Screening for molecules that have antiviral activity can be achieved by the range of methodologies described herein. Reference to a "cell" infected with a virus should be understood as a reference to any cell, prokaryotic or eukaryotic, which has been infected with a virus. This includes, for example, immortal or primary cell lines, bacterial cultures and cells in situ. In a suitable screening system for antiviral compounds, the preferred infected cells would be macrophages monocytes or hepatocytes/lymphoid cells infected with either HIV or HCV respectively.
Without limiting the present invention to any one theory or mode of action, the compounds of the present invention are thought to inhibit viral replication or virion release from cells by causing ion channels, namely VPU of HIV, the E protein of SARS and other Coronaviruses, or P7 of HCV to become blocked. The present invention encompasses antiviral compounds that are substituted acylguanidines. The present invention also includes the use of compounds 5-(N,N- hexamethylene)amiloride and 5-(N,N-dimethyl)-amiloride in the control of viral replication and/or growth other than HTV.
The subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer). Preferably, the subject is a human or primate. Most preferably, the subject is a human.
The method of the present invention is useful in the treatment and prophylaxis of viral infection such as, for example, but not limited to HTV infection, HCV infection and other viral infections. For example, the antiviral activity may be effected in subjects known to be infected with HIV in order to prevent replication of HIV thereby preventing the onset of AIDS. Alternatively, the method of the present invention may be used to reduce serum viral load or to alleviate viral infection symptoms. Similarly, antiviral treatment may be effected in subjects known to be infected with, for example, HCV, in order to prevent replication of HCV, thereby preventing the further hepatocyte involvement and the ultimate degeneration of liver tissue.
The method of the present invention may be particularly useful either in the early stages of viral infection to prevent the establishment of a viral reservoir in affected cells or as a prophylactic treatment to be applied immediately prior to or for a period after exposure to a possible source of virus.
Reference herein to "therapeutic" and "prophylactic" is to be considered in their broadest contexts. The term "therapeutic" does not necessarily imply that a mammal is treated until total recovery. Similarly, "prophylactic" does not necessarily mean that the subject will not eventually contract a disease condition. Accordingly, - therapy and prophylaxis include amelioration, of the symptoms of a particular condition or preventing or otherwise reducing the risk of developing a particular condition. The terin "prophylaxis" may be considered as reducing the severity of onset of a particular condition. Therapy may also reduce the severity of an existing condition or the frequency of acute attacks. In accordance with the methods of the present invention, more than one compound or composition may be co-administered with one or more other compounds, such as known anti-viral compounds or molecules. By "co- administered" is meant simultaneous administration in the same formulation or in two different formulations via the same or different routes or sequential administration by the same or different routes. By "sequential" administration is meant a time difference of from seconds, minutes, hours or days between the administration of the two or more separate compounds. The subject antiviral compounds may be administered in any order.
Routes of administration include but are not limited to intravenously, intraperitionealy, subcutaneously, intracranialy, intradermally, intramuscularly, intraocularly, intrathecaly, intracerebrally, intranasally, transmucosally, by infusion, orally, rectally, via iv drip, patch and implant. Intravenous routes are particularly preferred.
Compositions suitable for injectable use include sterile aqueous solutions (where water soluble) and sterile powders for the extemporaneous preparation of sterile injectable solutions. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol and liquid polyethylene glycol, and the like), suitable mixtures thereof and vegetable oils. The prevention of the action of microorganisms can be brought about by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thiπnerosal and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin. Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by, for example, filter sterilization or sterilization by other appropriate means. Dispersions are also contemplated and these may be prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, a preferred method of preparation includes vacuum drying and the fieeze-drying technique which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution. When the active ingredients are suitably protected, they may be orally administered, for example, with an inert diluent or with an assimilable edible carrier, or it may be enclosed in hard or soft shell gelatin capsule, or it may be compressed into tablets. For oral therapeutic administration, the active compound may be incorporated with excipients and used in the form of ingestible tablets, buccal tablets, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.01 % by weight, more preferably 0.1 % by weight, even more preferably 1% by weight of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 1 to about 99%, more preferably about 2 to about 90 %, even more preferably about 5 to about 80% of the weight of the unit. The amount of active compound in such therapeutically useful compositions in such that a suitable dosage will be obtained. Preferred compositions or preparations according to the present invention are prepared so that an oral dosage unit form contains between about 0.1 ng and 2000 mg of active compound.
The tablets, troches, pills, capsules and the like may also contain the components as listed hereafter: A binder such as gum, acacia, com starch or gelatin; excipients such as dicalcium phosphate; a disintegrating agent such as corn starch, potato starch, alginic acid and the like; a lubricant such as magnesium stearate; and a sweetening agent such as sucrose, lactose or saccharin may be added or a flavouring agent such as peppermint, oil of wintergreen, or cherry flavouring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar or both. A Syrup or elixir may contain the active compound, sucrose as a sweetening agent, methyl and propylparabens as preservatives, a dye and flavouring such as cherry or orange flavour. Any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compound(s) may be incorporated into sustained-release preparations and formulations.
The present invention also extends to forms suitable for topical application such as creams, lotions and gels. In such forms, the anti-clotting peptides may need to be modified to permit penetration of the surface barrier. Procedures for the preparation of dosage unit forms and topical preparations are readily available to those skilled in the art from texts such as Pharmaceutical Handbook. A Martindάle Companion Volume Ed. Ainley Wade Nineteenth Edition The Pharmaceutical Press London, CRC Handbook of Chemistry and Physics Ed. Robert C. Weast Ph D. CRC Press Inc.; Goodman and Gϊlm n's; The Pharmacological basis ofTherapeutics. Ninth Ed McGraw Hill; Remington- and The Science and Practice of Pharmacy. Nineteenth Ed. Ed. Alfonso R. Gennaro Mack Publishing Co. Easton Pennsylvania.
Pharmaceutically acceptable carriers and/or diluents include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, use thereof in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of active material calculated.to produce the desired therapeutic effect in association with the required pharmaceutical carrier. The specification for the novel dosage unit forms of the invention are dictated by and directly dependent on (a) the unique characteristics of the active material and the particular therapeutic effect to be achieved and (b) the limitations inherent in the art of compounding.
Effective amounts contemplated by the present invention will vary depending on the severity of the pain and the health and age of the recipient. In general terms, effective amounts may vary from 0.01 ng/kg body weight to about 100 g kg body weight.
Alternative amounts include for about 0. 1 ng kg body weight about 100 mg kg body weight or from 1.0 ng/kg body weight to about 80 mg kg body weight. The subject of the viral inhibition is generally a mammal such as but not limited to human, primate, livestock animal (e.g. sheep, cow, horse, donkey, pig), companion animal (e.g. dog, cat), laboratory test animal (e.g. mouse, rabbit, rat, guinea pig, hamster), captive wild animal (e.g. fox, deer). Preferably, the subject is a human or primate. Most preferably, the subject is a human. The methods of the present invention is useful in the treatment and prophylaxis of viral infection such as, for example, but not limited to HTV infection, HCV infection and other viral infections. For example, the antiviral activity may be. effected in subjects known to be infected with HIV in order to prevent replication of HIV thereby preventing the onset of AIDS. Alternatively, the methods of the present invention may be used to reduce serum viral load or to alleviate viral infection symptoms. Similarly, antiviral treatment may be effected in subjects known to be infected with, for example, HCV, in order to prevent replication of HCV, thereby preventing the further hepatocyte involvement and the ultimate degeneration of liver tissue.
The methods of the present invention may be particularly useful either in the early stages of viral infection to prevent the establishment of a viral reservoir in affected cells or as a prophylactic treatment to be applied immediately prior to or for a period after exposure to a possible source of virus.
The present invention will now be described in more detail with reference to specific but non-limiting examples describing studies of viral membrane ion channels and screening for antiviral activity. Some examples involve the use of the SARS virus. It will be clear from the description herein that other lentiviruses, and coronaviruses and other compounds may be used effectively in the context of the present invention. It is to be understood, however, that the detailed description is included solely for the purpose of exemplifying the present invention. It should not be understood in any way as a restriction on the broad description of the invention as set out above.
Example 1. Synthesis of the Compounds of the Invention.
The compounds of the present invention may be made from the corresponding acid chlorides or methyl esters as shown in Scheme 1. Both of these methods are well described in the literature.
Figure imgf000055_0001
acid chloride aeylgπaaidϊne ester
Scheme ! The following examples show synthetic schemes for some compounds of the invention.
Example 2. Synthesis of Cinttamnylfliιaτridiτιe from Cinnamic acid Chmamoyl chloride
Figure imgf000056_0001
To a solution of trώ«s-cinnamic acid (1.50 g, 10.12 mmol) in dry benzene (30mL) containing a drop of N,iV-dimethylformamide was added oxalyl chloride (5.14 g, 40.5 mmol) causing the solution to effervesce. After refluxing for 2 h, the solution was evaporated to dryness under reduced pressure. The resulting solid was dissolved in dry trirahydrofuran (20mL) and added slowly to a solution of guanidine hydrochloride in 2M aqueous sodium hydroxide (25mL). The reaction was stirred at room temperature for lh then extracted with ethyl acetate (3x50mL). The combined extracts were dried over magnesium sulfate and evaporated to give an orange oil. The crude product was purified by column chromatography. Elution with 10% to 20% methanol in dichloromethane gave Cinnamoylguanidine as a cream solid (0.829 g, 43%). ,
Example 3 Synthesis of N-aιnidinn--^-aτniτιo-5-phenvI-6-chloro-2-pyrazineearboxamϊde Par i
Figure imgf000056_0002
To a solution of methyl 3-amino-S,6-dichloro-2-pyra2inecarboxylate (0.444 g, 2.0 mmol) in tetrahydrofuran (5 L) / water (10 mL) / toluene (20 mL) was added phenyl borαπic acid (0.536 g, 4.4 mmol), sodium carbonate (0.699 g, 6.6 mmol) and tetrakis(triphenylphosphine)- paUadium(0) (0.116 g, 0.10 mmol). The reaction was evacuated and purged with nitrogen several times before being refluxed for 6 h. The organic layer was separated and the aqueous layer extracted with toluene (3 x 20 mL). The combined organic extracts were dried over magnesium sulfate, filtered and evaporated under reduced pressure to give methyl 3-a ino-6-chloroS- ph nyI-2-pyra∑inecarboxylats as a yellow solid (0.43 g, S2%).
Part 2
røj»jc= nα/Na05fe
Figure imgf000057_0001
Figure imgf000057_0002
Figure imgf000057_0003
To a solution of sodium (0.040 g, 1.74 mmol) dissolved in methanol (5 mL) was added guahidine hydrochloride (0.258 g, 2.70 mmol) and the mixture refluxed for 30 min after which it was filtered, To the filtrate was added methyl 3-amino-6- chloro-5-phenyl-2-pyrazinecarboxylate (0.264 g, 1.0 mmol) in N,N- dimethylfoimamide (5 mL) and the solution heated at 75oC for 12 h. The solvent was removed under reduced pressure and the residue chromatographed on silica gel eluting with 1 % triethylamine / 5% methanol / dichloromethane. The resulting solid was suspended in chloroform, filtered and dried under high vacuum to give N- Amidmo~3 ammo-5-phenyl-δ-chforo>-2''pyrazinecarboxamide as a yellow solid (0.04
Example 4. Synthesis of hexamethylflHftiinino.fi.phepyl^pyrazmecarbQxaniidje Part i
Figure imgf000057_0004
To a solution of methyl 3-amino-5,6-dichloro-2-pyrazinecarboxylate (1 , 11 g, 5.0 mmol) in tetrahydrof ran (50 mL) was added hexamethyleneimine (1.49 g, 15.0 mmol) and the reaction was refluxed for 1 h. The reaction was allowed to cool and the solid hexamethyleneimine hydrochloride removed.by filtration. The filtrate was evaporated and the residue chromatographed over silica gel. Elutiσn with dichloromethane gave methyl 3-amino-6-chloro-5'hexametkyIeneimino-2- pyrazinecarboxylaie as an off-white solid (1.20 g, 85%).
Part 2
Figure imgf000058_0001
To a solution of methyl 3-amino-6-chloro-5-hexamethyleneimino-2- pyrazinecarboxylate (0.350g, 1.23 mmol) in dimethylsulfoxide (5 mL) was added phenyl boronic acid (0.166 g, 1,35 mmol), potassium carbonate (0.511 g, 3.70 mmol) and [l,l'-bis(diphenylphosphino)ferrocene]dichloropalladium(Il)-dichloromethane complex (0.041 g, 0.05 mmol). The reaction was heated at 90oC for 16 h before being poured into water (50mL) and extracted with ethyl acetate (3 x 5ftmL). The combined extracts were dried over magnesium sulfate, filtered and evaporated to give a brown oil which was purified by chromatography on silica gel. Elution with dichloromethane followed by 10% ethyl acetate dichloromeihane gave methyl 3- amino-5-hexamethyIeneimino-6-pkenyI-2-pyrazinecarbσxyIate as a yellow solid (0.309 g, 77%).
Part 3.
Figure imgf000058_0002
To a solution of sodium (0.090 g, 6.17 mmol) dissolved in methanol (8 mL) was added guanidine hydrochloride (0.598 g, 6.26 mmol) and the mixture was refluxed for 30 min after which it was filtered. To the filtrate was added methyl 3- amino-5-hexamethyleneimino-6-phenyl-2-pyrazinccarboxylate (0.310 g, 0.95 mmol) in tetrahydrofuran (10 mL) and the solution refluxed for 72 h. The solvent was removed under reduced pressure and the residue chromatographed on silica gel. Elution with 5% methanol dichloromethane gave N-amidino-3-amino-5- hexamethyleneimino~6phenyl-2-pyra∑inecarb χamide as a yellow solid (0.116 g, 35%).
Example 5. Viral Studies
Construction of recombiuant plasmids containing open reading frames encnriiTiP various virus proteins.
Complimentary DNA (cDNA) fragments for the various viral proteins listed in Table 2 were obtained either by PCR amplification from a parental virus genome clone, or by direct chemical synthesis of the polynucleotide sequence. For example, the open reading frame encoding Vpu (Fig la) was amplified by PCR from a cDNA clone of an Nde I fragment of the HIV-1 genome (isolate HXB2, McFarlane Burnet Centre, Melbourne, Australia) as follows: Native PfU DNA polymerase (Stratagene; 0.035 U/ D) was chosen to catalyse the PCR reaction to minimise possible PCR introduced errors by virtue of the enzyme's proofreading activity. The 5 sense, primer AGTAGGATCCATGCAACCTATACC (< 400 > 2) introduces a BamHl site (underlined) for cloning in-frame with the 3' end of the GST gene in p2GEX (41). This primer also repairs the start codon (bold T replaces a Q of the vpu gene which is a threonine codon in the HXB2 isolate. The 3', antisense, primer
TCTGGAATTLTACAGATCAT CAAC (< 400 > 3) introduces an EcoRl site (underlined) to the other end of the PCR product to facilitate cloning. After 30 cycles of 94°C for 45 sec, 55°C for 1 min and 72°C for 1 min in 0.5 ml thin-walled eppendorf tubes in a Perkin-Ehner thermocycler, the 268bp fragment was purified, digested with BamHl and EcoRl and ligated to p2GEX prepared by digestion with the same two enzymes. The resultant recombinant plasmid is illustrated in Fig lb. The entireVpu open reading frame and the BamHl and EcoRl ligation sites were sequenced by cycle sequencing, using the Applied Biosystems dye-terminator kit, to confirm the DNA sequence. Other cDNAs were synthesised for us using state of the art methods by GenScript Corporation (New Jersey, USA). Codon sequences were optimised for expression in bacterial, insect or mammalian cells, as appropriate. Restriction endonuclease enzyme recognition sites were incorportated at the 5' and 3' ends of the synthetic cDNAs to facilitate cloning into plasmid expression vectors, ρcDNA3.1, pFastBac and pPL451 for expression of the encoded virus proteins in mammalian, insect or bacterial cells, respectively.
Standard techniques of molecular biology were used in cloning experiments. For example, to prepare the Vpu open reading frame for insertion into the ρPL451 expression plasmid, p2GEXVpu was first digested with BamHl and the 5' base overhang was filled in the Klenow DNA poly erase in the presence of dNTPs. The , Vpu-encoding fragment was then liberated by digestion with EcoRl, purified from an agarose gel and ligated into pPL451 which had been digested with Hpal and EcoRl. Western blots subsequently confirmed that the pPLVpu construct (Fig lc) expressed Vpu after induction of cultures at 42°C to inactivate the cI857 repressor of the PR and PL promoters.
Table 2 Source of viral cDNA or peptide sequences.
Figure imgf000060_0001
Example 6. Purification of Recombinant VPU from E» Colt
Cultures of E. coli strain XU-blue cells containing p2GEXVpu were grown at 30°C with vigorous aeration in LB medium supplemented with glucose (6g L) and ampicillin (50mg L) to a density of approximately 250 Klett units, at which time IPTG was added to a final concentration of O.OhnM and growth was continued for a further 4hr. The final culture density was approximately 280 Klett units. Since early experiments revealed that the majority of expressed GST-Vpu fusion protein was associated with both the cell debris and 30 membrane fractions, the method of Varadhachary and Maloney (Varadhachary and Maloney, 1990) was adopted to isolate osmotically disrupted cell ghosts (combining both cell debris and membrane fractions) for the. initial purification steps. Cells were harvested, washed, weighed and resuspended to lftml/g wet weight in MTPBS containing DTT (ImM) and MgCl2 (lOmM). Lysozyme (0.3 mg/ml; chicken egg white; Sigma) was added and incubated on ice for 30 min with gentle agitation followed by 5 min at 37°C. The osmotically sensitised cells were pelleted at 12,000g and resuspended to the original volume in water to burst the cells. The suspension was then made up to lxMTEBS/DTT using a lOx buffer stock and the ghosts were isolated by centrifugation and resuspended in MTPBS/DTT to which was then sequentially added glycerol (to 20 % wt vol) and CHAPS (to 2 % wt/vol) to give a final volume of one quarter the original volume. This mixture was stirred on ice for 1 hr and then centrifuged at 400,000g for lhr to remove insoluble material. The GST-Vpu fusion protein was purified from the detergent extract by affinity chromatography on a glutathione agarose resin (Sigma). The resin was thoroughly washed in 50mM Tris pH 7.5 containing glycerol (5 %), DTT (ImM), and CHAPS (0.5 %) (Buffer A) and then the Vpu portion of the fusion protein was liberated and eluted from the resin-bound GST by treatment of a 50% (v v) suspension of the beads with human thrombin (lOOU/ml; 37°C for lhr). PMSF . (0.5mM) was added to the eluant to eliminate any remaining thrombm activity. This Vpu fraction was further purified on a column of MA7Q anion exchange resin attached to a BioRad HPLC and eluted with a linear NaCl gradient (0-2M) in buffer A. The Vpu was purified to homogeneity - as determined on silver stained gels - on an immunoaffinity column as follows: HPLC fractions containing Vpu were desalted on a NAP 25 column (Pharmacia) into buffer A and then mixed with the antibody- agarose beads for lhr at room temperature. The beads were washed thoroughly and Vpu was eluted by increasing the salt concentration to 2M. Protein was quantitated using the BioRad dye binding assay.
Example 7. Expression and Purification of Vpu in E.Coli.
The plasmid p2GEXVpu (Fig. 1) was constructed to create an in-frame gene fbsion between the GST and Vpu open-reading frames. This system enabled IPTG-inducible expression of the Vpu polypeptide fUsed to the C-teπninus of GST and allowed purification of the fusion protein by affinity chromatography on glutathione agarose. Optimal levels of GST-Vpu expression were obtained by growing the cultures at 30°C to a cell density of approximately 250-300 Klett units and inducing with low levels of IPTG (O.Ol M). To purify the GST-Vpu, a combined cellular fraction containing the cell debris and plasma membrane was prepared by lysozyme treatment of the induced cells followed by a low-speed centrifugation. Approximately 50% of the GST-Vpu protein could be solubilised from this fraction using the zwitterionic detergent CHAPS. Affinity chromatography using glutathione-agarose beads was used to enrich the fusion protein and thrombm was used to cleave the fusion protein at the high affinity thrombin site between the fusion partners, liberating Vpu (Fig. 2A). In fractions eluted from the anion exchange column Vpu was the major protein visible on silver stained gels (Fig.2B, lane 1). Finally, Vpu was purified to apparent homogeneity on an immunoaffinity column (Fig.2B, lane 2). The N-terminal amino acid sequence of the protein band (excised from SDS-PAGE gels) corresponding to the immunodetected protem confirmed its identity as Vpu.
Example S. Reconstitutlon of Vpu in Phospholipid Vesicles.
Proteoliposomes containing Vpu were prepared by the detergent dilution method (New, 1990). A mixture of lipids (PEiPC:PS; 5:3:2; lmg total lipid) dissolved in chloroform was dried under a stream of nitrogen gas and resuspended in 0.1 ml of potassium phosphate buffer (50mM pH 7.4) containing DTT (ImM). A 25μl aliquot containing purified Vpu was added, followed by octylglucoside to a final concentration of 1.25 % (wt vol). This mixture was subject to three rounds of freezing in liquid nitrogen, thawing and sonication in a bath type sonicator (20-30 sec) and was then rapidly diluted into 200 volumes of the potassium phosphate buffer. Proteoliposomes were collected by centrifugation at 400,000g for lhr and resuspended in approximately 1 0μl of phosphate buffer.
Example 9. Assaying VPΠ Ion Channel Activity
Purified Vpu was tested for its ability to induce channel activity in planar lipid bilayers using standard techniques as described elsewhere (Miller, 1986; and Piller et al, 1996). The solutions in the CIS and TRANS chambers were separated by a Delrin™ plastic wall containing a small circular hole of approximately lOOμm diameter across which a lipid bilayer was painted so as to form a high resistance electrical seal. Bilayers were painted from a mixture (8:2) of palmitoyl-oleoly- phosphatidyl-ethanolamine and pahnitoyl-oleolyphosphatidyl-choline (Avanti Polar Lϊpids, Alabaster, Alabama) in n-decane. The solutions in the two chambers contained MES buffer (lOmM, pH 6.0) to which various NaCl or KCl concentrations were added. Currents were recorded with an Axopatch™ 200 amplifier. The electrical potential between the two chambers could be manipulated between +/-200mV (TRANS relative to grounded CIS). Aliquots containing Vpu were added to the CIS chamber either as a detergent solution or after incorporation of the protein into phospholipid vesicles. The chamber was stined until currents were observed.
Example 10. Vpu Forms Ion Channels in Lipid Bilayers.
To assay for ion-channel formation by Vpu, reconstitution into planar lipid bilayers was performed. When samples (containing between 7 and 70ng of protein) of purified recombiαant Vpu were added to the 1ml of buffer in the CIS chamber of the bilayer apparatus, current fluctuations were detected after periods of stirring that varied from 2 to 30 min (Fig.3). This time taken to observe channel activity approximately correlated with the amount of protein added to the chamber. No channels were detected when control buffer aliquots or control lipid vesicles were added to the CIS chamber. In those control experiments the chambers could be stirred for more than an hour without appearance of channel activity.
Example 11. Properties of The VPU Channels.
Channel activity was observed in over 40 individual experiments with Vpu samples prepared from five independent purifications. In different experiments, the amplitude of the currents varied over a large range and, again, seemed to approximately correlate with the amount of protein added. The smallest and largest channels measured had conductances of 14 pS and 280 pS, respectively. The channels were consistently smaller when lipid vesicles containing Vpu were prepared and fused to the bilayer rather than when purified protein in detergent solution was added. This may be because the former method included treatment with high concentrations of detergent and a dilution step that may have favoured the breakdown of large aggregates into monomers. The relationship between current amplitude and voltage was linear and the reversal potential in solutions containing a ten-fold gradient of NaCl (500mM CIS; 50mM TRANS) was +3GmV (Fig. 3B). A similar reversal potential was obtained when solutions contained KCl instead of NaCl. In 5 experiments with either NaCl or KCl in the solutions on either side of the membrane, the average reversal potential was 31.0 +/-1.2mV (+/-SEM). This is more negative than expected for a channel selectively permeable for the cations alone. Using ion activities in the Goldman- Hodgkin-Katz equation gives a Pjfø d ratio of about 5.5 indicating that the channels are also permeable to chloride ions. An attempt was made to reduce the anion current by substituting phosphate for chloride ions. When a Na-phosphate gradient (15QmM Na+ & lOOmM phosphate CIS; 15mM Na+ & lOmM phosphate TRANS, pH 6.8) was used instead of the NaCl gradient, the reversal potential was 37.1 +/- 0.2 (+/-SEM, n-2) again indicating a cation/anion permeability ratio of about 5. (For calculations involving the phosphate solutions, the summed activities of the mono and bivalent anions were used and it was assumed that the two species were equally permeable). - The current-voltage curve now exhibited rectification that was not seen in the NaCl solutions. It can be concluded that the channels formed by Vpu are equally permeably to Na+ and K+ and are also permeable, though to a lesser extent, to chloride as well as phosphate ions.
Example 12. Bacterial Bio-Assav for Screening Potential Ion Channel-Blocking Drugs.
This bio-assay is based on the observation that expression of Vpu in E. coli results in an active Vpu channel located in the plasmalemma that dissipates the transmembrane sodium gradient. As a consequence of this Vpu channel activity, metabolites whose accumulation within the cells is mediated by a sodium dependent co-transporter (for example proline or adenine) leak out of the cell faster than they can be synfhesised so that the metabolites' intracellular levels become limiting for growth of the cell. Thereby, an E. coli cell expressing Vpu is unable to grow in minimal drop-out media lacking adenine or proline. However, in the presence of a drug that blocks the Vpu channel, the cell is once again able to re-establish its transmembrane sodium gradient - due to the action of other ion pumps in the membrane - and the leakage of metabolites is prevented enabling growth. Experi ents to demonstrate that Vpu can foπn sodium channels in the plasma membrane of E. coli were performed as follows.
To express unftised Vpu in E. coli, the vpu open-reading frame was cloned into the plasmid pPL451 to create the recombinant plasmid pPL-Vpu (Fig. lb). In this vector the strong P and P lambda promoters are used to drive expression of Vpu under control of the temperature sensitive cl857 represser, such that when grown at 30°C expression is tightly repressed and can be induced by raising the temperature to between 37*C and 42βC. On agar plates, cells containing pPL-Vpu grew when incubated at 30°C and 37°C but not at 42°C, while control strains grew well at 42°C. Liquid cultures of cells containing pPL-Vpu were grown at 30°C to ODsoof=0.84 then moved to grow at 42°C for two hours (the final cell density was OD6oo,=0.75). The plasma membrane fraction was prepared and western blotting, using an antibody that specifically binds to the C-terminus of Vpu, detected a single band at approximately lόfcDa, indicating that Vpu was expressed an associated with the membranes (Fig. 2A, lane 5).
Example 13. Cross-Feeding Experiments Reveal That Proline Leaks _Oμ _of Cells Expressing VPU.
Uptake of proline by E. coli is well characterised and active transport of the amino acid into the cells is known to use the sodium gradient as the energy source (Yamato et al, 1994). To detect whether proline leakage occurs, the following cross- feeing assay was used: A lawn of an E. coli strain auxotrøph c for proline and methionine (Met" PκO, was seeded and poured as a soft agar overlay on minimal drop-out media plates lacking proline but containing methionine. Sterile porous filter discs were inoculated with a Met+ Pro+ strain (XL-1 blue) containing either the pPL451 control plasmid or pP! -Vpu and placed onto the soft agar. The plates were then incubated at 37°C or 30°C for two days. After than time a halo growth of the Met" Pro" strain was clearly visible surrounding the disc inoculated with the cells containing pPL-Vpu incubated at 37eC (Fig. 4A). This growth can only be due to the leakage of proline from the Vpu-expressing cells on the disc. No such leakage was apparent from the control strain at 37βC nor around either strain on plates grown at 30*C (Fig. 4B). Iα contrast to proline transport, the E. coli methionine permease is known to belong to the ABC transporter family (Rosen, 1987) and hence be energised by ATP. Identical crossfeeding experiments to those described above were set us except that the Met" Pro" strain was spread on minimal drop-out plates lacking methionine but containing proline. No growth of this strain was evident around any of the discs (Fig. 4C), indicating that methionine was not leaking out of the XL-1 blue cells even when Vpu was being expressed.
Example 14. E.Coli Cells Expressing Vpn Require Adenine in the External Medium for Growth. It was observed that, due to an uncharacterised mutation in the adenine synthesis pathway, growth of E. coli cells of the XLI-blue strain expressing Vpu at 37°C was dependant on the presence of adenine in the medium. This allowed the development of an even simpler bioassay for Vpu ion-channel activity than the proline cross-feeding assay described above: A lawn of XLl-blue cells containing the pPL-Vpu plasmid is seeded onto an agarose plate lacking adenine in the medium, small aliquots of drugs to be tested for inhibition of the Vpu channel are spotted onto the agarose in discrete locations and the plates are incubated at 37°C for a suitable period of time (12-36 hours). Halos of growth around a particular drug application site indicate that the drug has inhibited expression of the Vpu ion channel activity that prevents growth in the absence of the drug. (Figure 5).
Example 15
Assay of Compounds in Planar Lipid Bilayers for Vpn Channel Blocking Activity Comopunds were characterized for their ability to block Vpu ion channel activity reconstituted into planar lipid bilayers. Vpu N-terminal peptide (residues 1- 32) dissolved in trifluoroethanol was added to the CIS chamber of the bilayer apparatus and the solutions was stirred until ion currents were observed, indicating incorporation of one or more Vpu ion channels into the bilayer. After recording the channel activity for a few minutes, drugs were added to the solutions in the CIS and TRANS chambers - with stirring - to a final concentration of lOOμM. Channel activity was then recorded for at least a further three minutes and the effect of drug addition on ion current was determined by comparing the channel activity before and after drug addition. For each experiment, drug effect was classified into four categories: "Stong block", if current was inhibited approximately 90-100%; "weak block", approx. 50-90% inhibition; "partial block", <50%; and "no effect". Experiments were disregarded if currents larger than ±50pA were generated after . addition of Vpu N-peptide because in such cases it is possible that non-native peptide aggregates contribute to bilayer breakdown. Such aggregates, by virtue of their disorganized structure may not be specifically blocked by the drugs at the concentrations tested.
Table 3 summarises the results of the bilayer experiments. A novel outcome of these experiments was the strong blocking of Vpu channels observed with Phenamil. Phenamil has a phenyl group derivative at the guanidine group of amiloride. Amiloride itself is not a blocker of Vpu, whereas addition of the hexamethylene group at the 5- position of the pyrazine ring created a structure (HMA) that blocks the channel at concentrations as low as 2SμM. These new results with Phenamil, however, now show that a bulky hydrophobic derivative at the opposite end of the molecule can also turn amiloride into an effective Vpu channel blocker. Interestingly, benzamil, with a very similar structure was much less effective at blocking the Vpu channel.
Table 3: Summary of Compounds Inhibiting the Vpu Ion Channel in Bilayers
Compound No. of Results Expts.
Phenamil 3 3x Strong block
MIA 2 lx Strong block; lx weak
Benzamil 10 3x partial block; 7x no effect
EIPA 3 3x weak block;
HMA 1 lx Strong block;
(5-Phenyl-penta-2,4-dienoyl)guanidiπe 6 6x strong block
6-methoxy-2-naphthoylguanidine 5 5x strong block
(2-Chlorocinnamoyl)guanidine 6 4χ strong; 2x partial blocks
3-(trifluoromethyl)cinnamoylgua dine 5 4x strong blocks; lx no effect N-{5-[3-(5-Guanidino-pentyloxymethyl benzyloxy]-pentyl}-guanidine 4 3x strong block; lx no effect
4-phenylbenzoylguaπidine 3 3x strong block
3-methylcinnamoylguanidine 4 2x strong block; 2x partial
(3-Chlorocinnamoyl)guanidine 4" 2x strong block; 2x partial
N-(3-phenylρroρanoyl)-N'- phenylguanidine 1 lx strong blocks
(3-Bromocinnamoyl)guanidine 3 3x partial-strong block
5-tert-butylamino-aαύloride 3 3x partial block
N-amidino-3-amino-5-phenyl-6-chloro-2- pyrazύiecarboxamide 3 3x partial block
3-methoxy-HMA 3 3x partial block
5-(N-Methyl-N-isobutyl)amiloride 1 lx partial block
5-(N-Efhyl-N-isopropyl)amiloride 1 lx partial block
2-napthoylguanidine 7 7x weak block
N,N-bis(3phenylpropanoyl)-N"- phenylguanidine 7 7x weak block cinnamoylguanidine 3 3x weak block
(5-Phenyl-penta-2,4-dienoyl ^uanidine 6 6x strong block
Example 16 Compound Screening using the Bacterial Bio-Assav for the Vpu protein.
The halos of growth around the site of application of particular drugs - as described in example 14— were given a score between zero and six reflecting the size and density of the zone of bacterial cell growth. Scores greater than 3 represent strong inhibition of the Vpu protein; scores between 1.5 and 3. represent moderate inhibition and scores between 0.01 and 1.5 represent fair inhibition.
Table 4 lists the scores for inhibition of Vpu protein in the bacterial bio-assay.
Table 4
Figure imgf000068_0001
5-bromo-2-fluorocinnamoylguamdine 3.5/2
3-methylcinnamoylguanidine 3.4/2
2-methylcinnamoylguanidine 3.1/2
2,3-dimethylcinnamoylguaπidine 3.1/2 cinnamoylguanidine 2.96/12
S-methoxy-2-naphthoylguanidine 2.9/4 ixans-3-(l-napthyl)acryloylguanidine 2.9/3
3,4-dicMorocinnamoylguanidine 2.9/3
2,6-dichlorocinnamoylguanidine 2.88/2"
4-ρhenylbeazoylguanidine 2.75/5
2-ethylcinnamoylguanidine 2.75/2
(4-Chlorocinnamoyl)guanidine 2.7/5
2-napthoylguanidine 2.7/11
2,5-dimethyleianamoylguanidine 2.69/2
3-isopropylcinnamoylguanidine hydrochloride 2.6/2
(5-Phenyl-penta-2,4-dienoyl)guanidine 2.56/2
3-phenylciunamoylguanidine 2.54/3
(4-Bromocinnamoyl)guauidine 2.5/4
5-(3'-bromophenyI)penta-2,4-dienoylguanidme 2.5/2
3-(cyclohex-l -en-l-yl)cinnamoylguanidine 2.5/2
3-(1rifluoromethoxy)cinnamoylguanidine 2.44/2
2-(trifluoromethyl)cinnamoylguanidine 2.4/2
N,N1-bis(3phenylρropanoyl N"-ρhenylguanidine 2.25/3
2-ethoxyciπnamoylguanidine 2.25/2
N-(3-phenylpropanoyl)-N,-phenylguanidine 2.21/3
4-(trifluoromethyl)cinnamoylguanidine 2.2/2
C4-Mefeoxy nnamoyl guamdine 2.13/3
2-t-butylcinnamoylguanidine 2.13/2
4-methylcinnamoylguanidine 2.1/2
2-fluorocinnamoylguanidine 2.1/2
2-phenylcinnamoylguanidinje 2.1/2
N-(6-Hydroxy-2-napthoyl)-N'-phenylguanidine 2.06/2
3-t-butylcinnamoylguanidine 2.06/2
3,4-difluorocinnarnoylguanidine 2.06/2
5-(N»N-hexamethylene)amiloride 1.9/31
3-fluorocinnamoylguanidine 1.9/2
5-bramo-2-methoxycinnamoylguanidine 1.9/2
3-ethoxycinnamoylguanidine 1.9/2
3,4-(methylenedioxy)cinnamoylguanidine 1.88/2
(2-Methoxyoiπnamoyl)guaπidine 1.7/4
2'4 DichloroBenzamil HCI 1.7/2
2,3,5,6,-tetramethylcinnamoylguaπidine 1.6/2
3-(2-napthyl)acryloylguanidine 1.56/2
2-(l-napthyl)acetoylguanidine 1.56/2
2,3-difluorocinnamoylguanidine 1.56/2
(3-Methoxycinnamoyl)guanidine 1.52/6
4-isopropylcinnamoylguaπidine 1.4/2
2,4,6-trimethylcinnamoylguanidine 1.4/2
Figure imgf000070_0001
Human monocytes were isolated from peripheral blood and cultured either for 24hr (one day old monocytes) or for 7 days to allow differentiation into monocyte derived acrophages (MDM). These cells were then exposed to cell-free preparations of HIV isolates and allowed to absorb for 2hr before complete aspiration of the medium, washing once with virus-free medium and resuspension in fresh medium. The cells were exposed to various concentration of compound either 24 hr prior to infection or after infection. Subsequent HIV replication, at various times after infection, was compared in cells exposed to drugs and in cells not exposed to drugs (controls). The progression and extent of viral replication was assayed using either an HIV DNA PCR method (Fear et al, 1998) or an ELISA method to quantitate ρ24 in culture supernatants (Kelly et al, 1998).
Table 5 provides examples of results obtained using this assay and test antiviral compounds.
Table s
Figure imgf000071_0001
Example IS. SARS Coronavirus.
SARS E protein forms an ion channel Peptide Synthesis
A peptide corresponding to the full-length SARS-CoV (isolate Tor2 and Urbani) E protein (MYSFVSEETGTLIVNSVLLFLAFVVi iVTLAϊLTAlRLCA YCCNIVNVSLVKPTVYVYSRVKNLNSSEGVPDLLV) and a second peptide comprising the first 40 amino acids of the full length E protein which correspond to the transmembrane domain (MYSFVSEETGTLIVNSVLLFLAFVVF LLVTLAILTALRLC) were synthesized manually using FMOC chemistry and solid phase peptide synthesis The synthesis was done at the Biomolecular Resource Facility (John Curtin School of Medical Research, ANU, Australia) using a Symphony11 Peptide Synthesiser from Protein Technologies fcic.(Tucson, AZ, USA) according to the manufacturers instructions.
Example 19. Peptide purification Mass spectral analysis of the synthetic peptide revealed that the preparation contained significant amounts of material with lower na/z ratio than expected for the full-length product. The majority of these are presumably truncated peptides generated during the peptide synthesis process. To enrich the full-length E protein, the following procedure was used, which relies on differential solubility of the smaller molecules and full-length peptide. The crude preparation was suspended at 12 mg ml in 70% CH3CN, 0.1 %TFA and voitexed for 10 minutes. This suspension was centrifiiged at 10,000g for 10 minutes at 20°C. The supernatant was discarded and the insoluble fractions was extracted with 70% CH3CN, 0.1 % TFA, as above, two . more times. The insoluble material containing the E protein was dried using Speedvac an the weight of the final product was used to calculate the yield. The purified peptide was analysed by Bruker Qmniflex MALDKTOF mass spectrometry in HABA matrix at 2.5mg ml in methanol at a 1 :1 ratio and spectra were obtained in the positive linear mode. A clear peat at m/z ratio of 8,360.1 was seen as expected for the calculated molecular weight of full-length E protein and 4422.3 for the N-terminal E protein. Example 20. Planar Lipid Bilavers
The SARS virus E protein was resuspended at lmg/ l in 2,2,2-trifluoroethanoL The SARS virus E protein's ability to form ion channels was tested on a Warner (Warner instruments, Inc. 1125 Dixwell Avenue, Hamden, CT 06 14) bilayer rig as follows; A lipidmix of 3:1:1, l-Palmitoyl-2-oleolyi phosphatidyl Ethanolamine: l-Pahnitoyl-2- oleolyl phosphatidyl Serine: l-Pahnitoyl-2-oleolyl phosphatidyl choline in CHC13 was dried under 2 gas and resuspended to 50mg ml in n-decane, Bilayers were painted across a circular hole of approximately lOOμm diameter in a Delrin™ cup separating aqueous solution in the CIS and TRANS chambers. The CIS chamber contained a solution of 500mM NaCl or KCl, in a 5mM HEPES buffer pH 7.2, the TRANS chamber contained a solution of 50mM NaCl or KCl, in a 5mM HEPES buffer pH 7.2. Silver electrodes coated in chloride with 2% agarose bridges are placed in the CIS and TRANS chamber solutions. The SARS E protein full-length or N- terminal peptides (3-1 Ou ) were added to the CIS chamber, which was stύτed until channel activity was detected. The CIS chamber was earthed and the TRANS chamber was held at various holding potentials ranging between +100 to -lOOmV. Currents were recorded using a Warner model BD-525D amplifier, filtered at lfcHz, sampling at 5 kHz and digitally recorded on the hard disk of a PC using software developed in house. Drugs to be tested for their ability to inhibit SARS E protein ion channel activity were made up at 50mM in a solution of 50% DMSO: 50% methanol. For experiments testing the ability of compounds to inhibit E protein ion channel activity, 100 μM to 400 μM of compound was added to the CIS chamber while stirring for 30 seconds. Bilayer currents were recorded before channel activity, during channel activity and after the addition of the drug.
Among the compounds tested was cinnamoylguanidine (Bit036), a compound which was shown in earlier experiments to be antiviral and to inhibit ion channel proteins from other viruses.
Example 20.1. Polvacrylamide gel electrophoresis Purified E protein was dissolved to 1 mg/ml, 5 mg/ml and 10 mg ml in, 6 M
Urea, 10% Glycerol, 5% SDS, 500 mM DTT, 0.002% Bromophenol Blue, 62.5 mM Tris HCI (pH 8.3). Peptides in solutions were heated at 1 0°C for 20 minutes before 30 μL samples were run on stacking gel 4-20% (Gradipore). SeeBlue® pre-stained standard (Invitrogen) was used for molecular weight markers.
Example 20.2 Results
To test if the SARS E protein forms ion channels the purified synthetic peptide was reconstituted into planar lipid bilayers' (21). Typically, 3 μg of SARS full-length E protein was added to the CIS chamber, while stirring. This CIS chamber contained 500 mM NaCl and the TRANS chamber contained 50 mM NaCl. In 60 experiments, ion currents due to SARS E protein ion channel activity were observed after about 5 -15 minutes of stirring. Activity was detected more rapidly and reliably with a holding potential of approximately —1 OOmV across the bilayer. Currents recorded at -lOOmV, (A) and at -60mV (B) in one of these experiments are shown in Figure 6. In that experiment the reversal potential was about +48mV and the channel conductances were calculated to be 52pS and 26pS, respectively. This indicates that the current-voltage (IV) relationship is not linear. In ten other experiments, where no protein was added to the CIS chamber, no ion channel activity was detected, even after recording for over 1 hour.
Figure 7a shows typical current traces recorded over a range of potentials in NaCl solutions. In that experiment the direction of current flow reversed at +48mV (Fig 7b). The IV curve shows that at the lower voltages the average current flow across the bilayer is small but at higher potentials there is an increase in average current across the bilayer, resulting in a non-linear IV relationship. In seven independent experiments, the average reversal potential was +48.3 + 2.3 mV (mean + 1SEM), indicating that the channels were about 37 times more permeable to Na+ ion than to CI" ions. The reversal potential is close to the Na+ equilibrium potential (+53mV), therefore the channel is selective for Na+ ions. For these 7 experiments the channel conductance varied between 95-164 pS; the average conductance was 130 ± 13 pS. SARS E protein ion channel is slightly less selectivity for K+ ions than Na+ ions. Figure 8b shows recording of cuπrents in KCl solutions at a range of potentials. In this experiment the currents reversed at +31 mV. In seven similar experiments E protein ion channel average reversal potential was +34.5 ± 2.5 mV. Therefore the SARS E protein ion channel is about 7.2 times more permeable to K+ ions than CY ions. In seven experiments, the channel conductance varied ranging between 24-166 pS, the average conductance was 83.4 ± 26 pS.
Similar results were obtained with a second synthetic peptide, which corresponded to the first forty N-terminal amino acids of the SARS E protein "N-terminal peptide" (21). The average reversal potential in NaCl solution in four experiments was +46.3 + 2.5 V, indicating that the ion channel formed by N-terminal peptide is about 25 times more permeable to Na+ ion than to CI- ions. The SARS E protein N-terminal peptide was sufficient for the formation of ion channels with properties like those of the full length SARS E protein. Therefore, the selectivity filter for the SARS E protein is most likely contained within the first forty amino acids of the N-terminal. SARS E protein N-terminal peptide also formed ion channels in KCl solution that were similarly selective for K+ ions compared to the full-length E protein. In five independent experiments the average channel reversal potential was +39.5 ± 3.6 V, therefore the channel is about 11 times more permeable to + ions than CI" ions. SDS-PAGE of the purified full-length E protein peptide showed bands corresponding to the full-length E protein (Data not shown). Larger bands of varying size up to about 20 kDa were detected, suggesting that SARS E protein may form homo- oligomers.
Example 21. SARS E protein ion channel is blocked by cinnamoylguanidine and other compounds
E protem ion channel activity in NaCl solutions was significantly reduced (p≥ 0.01, n=6 experiments) by addition of 100 to 200 μM cinnamoylguanidine to the CIS chamber. The average current across the bilayer was reduced to baseline by 1 OOμM cinnamoylguanidine. In experiments when E protein ion channels had higher conductance, 100 to 200 μM ciπnamoulguanidine reduced the average current across the bilayer about 4 fold'. Similarly, in four other experiments, 100 to 200 μM cinnamoylguanidine blocked channels formed by full-length E protein in KCl solutions. In two additional experiments, the SARS E protein N-tetminal peptide was blocked by 100 to 200 μM cinnamoylguanidine, demonstrating that the cinnamoylguanidine drug-binding site is located within the first forty amino acids of the E protein N-teπninal domain. Other compounds tested in bilayers for their effect on the SARS E protein are shown iri below in Table 6.
Table 6
Figure imgf000076_0001
Example 21.1 Results and Discussion.
We have shown that SARS E protein can form ion channels in lipid bilayer membranes. Hie ion currents reversed at positive potentials, which demonstrates that E protein ion channels are selective for monovalent cations over mαnovalent anions. " E protein Ion channels were about 37 times more selective for Na+ ions over Cl-ions and about 7.2 times more selective for K+ ions over CI- ions. In over 60 experiments the Na+ conductance of the E protein ion channel varied from as low as 26 pS to as high as 164 pS. SDS-PAGE showed that the E protein forms homo-oligomers, and we surmised that the larger conductances were probably due to aggregation of the E protein peptide leading to larger ion channels or the synchronous opening of many ion channels. Single channel currents were observed in several experiments and from these the channel conductance was calculated to be voltage dependent. The first 40 amino acids of the N-terminal which contains the hydrophobic domain of the SARS virus E protem is sufficient for the formation of ion channels on planar lipid bilayers. The N-terminal E protein ion channel has the same selectivity and conductance as the full-length E protein i n channel.
The SARS virus full length E protem ion channel activity and N-terminal domain E protem ion channel activity on planar lipid bilayers in NaCl and KCl solutions was inhibited by addition of between lOOμM to 200μM cinnamoylguanidine to the CIS chamber. Inhibition or partial, inhibition of the E protein ion channel activity by cinnamoylguanidine has been observed in seven independent experiments in NaCl solution and four independent experiments in KCl solution.
All known coronaviruses encode an E protein with a hydrophobic N-terminus transmembrane domain therefore all coronaviruses E proteins could form ion channels on planar lipid bilayers. This indicates that the E protein could be a suitable target for antiviral drugs and potentially stop the spread of coronavirus from infected host cells. Drugs that block the E protein ion channel could be effective antiviral therapy for the treatment of several significant human and veterinary coronavirus diseases including SARS and the common cold.
Example 22. Bacterial Bio-Assav for Screening Potential SARS-CoV E protein Ion Channel-Blocking Drugs. SARS-CoV E protein Ion Channel inhibits Bacterial Cell growth.
A bio-assay of SARS-CoV E protem function in bacterial cells was developed. A synthetic cDNA fragment encoding SARS-CoV E protein was cloned into the expression plasmid ρPL451, creating a vector in which E protein expression is . temperature inducible, as described in Example 4. Inhibition of the growth of E.coli cells expressing E protein at 37°C was observed as an indicator of ρ7 ion channel function dissipating the normal Na+ gradient maintained by the bacterial cells.
Example 23. Compound Screening using the Bacterial Bio-Assay for SARS coronavirus E protein.
The halos of growth around the site of application of particular drugs - as described in example 14 - were scored as decribed in example 15.
Table 7 lists the scores for inhibition of SARS-CoV E protein in the bacterial bio- assay.
Table 7
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Example 24. SARS Antiviral Assay for testing compounds against replication of SARS coπmavirαs (SARS-CoV).
Compounds were tested against SARS-CoV (Hong Kong strain) using virus plaque purified three times in Vero cells. Stock virus was generated by infecting Vero cells at MOI = lx TCID50 per 100 cells. Example 24.1 Screening for anti-viral activity using the virus microtitre assay Monolayers of Vero cells grown in 25cm2 flasks were infected at a multiplicity of 1 :50 and treated immediately post infection with compounds at two concentrations, lQuM and 2uM. A control infected monolayer remained untreated. Samples of culture media were taken at 48 hours post infection. Two aliquots from each of the samples (titrations 1 and 2) were serially log diluted and 12 replicates of log dilutions -4 to -7 added to cells in microtitre plates. Four days later, wells in the microtitre plates were scored for cytopathic effect (CPE) and the titration values calculated based on the number of CPE positive wells at the 4 dilutions. Control titres were 4.8 and 5.9 TCID50 x 106 (average 5.35 x 106)
Example 25; Effect of compounds in SARS CoV antiviral assay;
Three selected compounds were tested for activity against SARS-CoV according to the method described in example 21, For trans-3-(l- napthyl)aαryloylguaπidine and cinnamoylguanidine a decrease in virus titre of approximately 80% was observed at a concentration of 1 OuM and a reduction of approximately 50% was seen to persist at 2μM trans-3-(l-napthyl)acryloylguanidine.
Table 8 provides Virus titration data presented as % of a control (SARS CoV grown for 48 hours in the absence of compounds).
Table 8
Figure imgf000080_0001
Example26. Human 229E Coronavirus
Synthesis and Purification of a Peptide Corresponding to the 229E-E Protein
A peptide corresponding to the full-length 229E-E protein (sequence: MFLKLVDDHALVVIWLLWCVVLIVILLVCΠΉKLIKLCFTCHMFCNRTVYG^^
KNVYHIYQSYMHIDPFPKRVIDF; accession number NP_073554) was synthesized manually using FMOC chemistry and solid phase peptide synthesis. The synthesis was done at the Biomolecular Resource Facility (John Curtin School of Medical Research, ANU, Australia) using a Symphony*1 Peptide Synthesiser from Protein Technologies Inc.(Woburn, MS, USA) according to the manufacturers instructions to give C-terminal amides, the coupling was done with HBTU and hydroxybenzotriazole in N-methylpyrrolidone. Each of the synthesis cycles used double coupling and a 4-fold excess of the amino acids. Temporary α-N Fmoc- protecting groups were removed using 20% piperidine in DMF. The crude synthetic peptide was purified using the ProteoPIus™ kit (Qbiogene inc. CA), following manufactures instructions. Briefly, the peptides were diluted in loading buffer (60mM Tris-HCl pH 8.3, 6M urea, 5% SDS, 10% glycerol, 0.2% Bromophenol blue, + 100 mM β-tnercaptoethanol) and run on 4-20% gradient polyacrylamide gels (Gradipore, NSW, Australia) in tris-glycine electrophoresis buffer (25 mM Tris, 250 M glycine, 0.1% SDS). The peptides were stained with gel code blue (Promega, NSW) and the bands corresponding to the full-length peptide were excised out of the gel.
The gel slice was transferred to the ProteoPLUS™ tube and filled with tris-glycine electrophoresis buffer. The tubes were emerged in tris-glycine electrophoresis buffer and subjected to 100 volts for approximately 1 hour. The polarity of the electric current was reversed for 1 minute to increase the amount of protein recovered. The peptides were harvested and centrifuged at 13, 000 rp for 1 minute. The purified peptides were dried in a Speedvac and the weight of the final product was used to calculate the yield. Examnle 27.229E-E protein forms ion channels in planar lipid bflavers.
Lipid bilayer studies were performed as described elsewhere (Sunstrom, 1996; Miller, 1 86). A lipid mixture of pahnitoyl-oleoyl-phospihatidylethanolamine, palmitoyl-oleoyl-phosphatidylserine and pakmtoyl-oleoyl-phosphatidylcholine (5:3:2) (Avanti Polar Lipids, Alabaster, Alabama) was used. The lipid mixture was painted onto an aperture of 150-200 μm in the wall of a 1 ml deidn cup, The aperture separates two chambers, cis and trans, both containing salt solutions at different concentrations. The cis chamber was connected to ground and the trans chamber to the input of an Axopatch 200 amplifier. Normally the cis chamber contained either 500mMNaClor500mM Cl an thetrans 50 mMNaCl or50mMKCl. The bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis. The synthetic peptide was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 5000 Hz and steered on magnetic disk.
The 229E E synthetic peptide was dissolved in 2,2,2-trifluorethanol (TFE) at 0.05mg ml to 1 mg ml. 10 μl of this was added to the cis chamber (1ml aqueous volume) of the bilayer apparatus, which was stirred via a magnetic "flea". Ionic currents, indicating channel activity in the bilayer, were typically detected within 15- 30 min. After channels were detected the holding potential across the bilayer was varied between -lθOmV and +100mV to characterise the size and polarity of current flow and enable the reversal potential to be determined.
In 15 experiments where the cis chamber contained 50QmM NaCl solution and the trans chamber contained 50 mM NaCl solution, the average reversal potential of the channel activity was calculated to be 22 ±7 (SEM) mV. In 13 experiments where the cis chamber contained 500mM KCl solution and the trans chamber contained 50 mM KCl solution, the average reversal potential of the channel activity was calculated to be 38 ±4 (SEM) mV. These results indicate that the 229E E protein forms cation selective ion channels that are slightly more selective for K+ than for Na+ ions. Figure 9 shows examples of raw current data for the 229E E ion channel at various holding potentials (cis relative to trans) in asymmetrical KCl solutions (500/50 mM).
The graph is a representative plot of average bilayer current (pA; y-axis) versus holding potential (mV; x-axis).
Example 28. Chemical compounds inhibit the ion channel activity of the 229E E protein synthetic peptide.
To test compounds for their ability to block or otherwise inhibit the ion channel formed by 229E E protem, small aliquots of solutions containing the compounds were added to the aqueous solutions bathing planar lipids in which the peptide channel activity had been reconstituted and the effect of the compound addition on the ionic currents was recorded and measured.
Compound stock solutions were typically prepared at 500 mM in DMSO. This solution was further diluted to 50 M, or lower concentration in 50% DMSO/50% methanol and 2 μl of the appropriately diluted compound was added to the cis and/or trans chambers to yield the desired final concentration.
In the example shown in Figure 10, addition of lOOμM nnamoylguanidine to the cis chamber greatly reduced current flow through the 229E E ion channel.
Example. 29. Bacterial Bio-Assay for Screening Potential 229E-CoV E protein Ion Channel-Blocking Drugs.
229E-CoV E-protein Ion Channel inhibits Bacterial Cell growth. A bio-assay of 229E-CoV E-protein function in bacterial cells was developed. A synthetic cDNA fragment encoding 229E-CoV E-protein was cloned into the expression plasmid ρPL451, creating a vector in which E protein expression is temperature inducible, as described in Example 4. Inhibition of the growth of Kcoli cells expressing E protein at 37°C was observed as an indicator of p7 ion channel function dissipating the normal Na+ gradient maintained by the bacterial cells. Example 30 Compound Screening using the Bacterial Biq-Assay for 229E-CoV E-protein.
The halos of growth around the site of application of particular drugs - as described in example 14 -were scored as decribed in example 15.
Table 9 list the scores for inhibition of 229E-CoV E-protein in the bacterial bio-assay. Table 9
Figure imgf000084_0001
Figure imgf000085_0001
Example 31; Antiviral Assay for testing compounds against replication of human coronavirus 229E (229E1
To determine the antiviral activity of compounds against human coronavirus 229E replication (ATCC VR-740), an assay measuring reduction in the number of plaques formed in monolayers of 229E infected MRC-5 cells (human lung fibroblasts ;ATCC CCL-171) was developed: First, a virus working stock was prepared by amplification in MRC-5 cells. This was then used to infect confluent monolayers of MRC-5 cells grown in 6-well tissue culture plates by exposure to the virus at an MOI of approx. 0.01 pfu/cell for 1 hour at 35βC in 5%COa- The infective inoculum was removed and replaced with fresh medium (DMEM supplemented with 10% fetal calf serum) containing various test concentrations of compounds or the appropriate level of solvent used for the compounds (control). Plates were subsequently i cubated at 35°C (in 5% C j) for 3 -5 days post infection, after which time culture supernatant was removed and the cells were stained with 0.1 % crystal violet solution in 20% ethanol for 10 minutes. Plaques were counted in all wells and the percentage reduction in plaque number compared to solvent control was calculated. Measurements were performed in duplicate to quadruplicate wells.
Table 10
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Example 32 Human OC43 Coronavirus
OC43 Antiviral Assay for testing compounds against replication of human coronavirus OC43.
To determine the antiviral activity of compounds against human coronavirus OC43 repHcation (ATCC VR-759), an ELISA assay was developed measuring the release of the viral N-protein into culture supernatants from monolayers of OC43- infected MRC-5 cells (human lung fibroblasts ;ATCC CCL-171): First, a virus working stock was prepared by amplification in MRC-5 cells. This was then used to infect confluent monolayers of MRC-5 cells grown in 6-well tissue culture plates by exposure to the virus at an MOI of approx. 0.01 pfu/cell for 1 hour at 35°C in 5%Cθ2. The infective inoculum was removed and replaced with fresh medium (DMEM supplemented with 10% fetal calf serum) containing various test concentrations of compounds or the appropriate level of solvent used for the compounds (control). Plates were subsequently incubated at 35°C (in 5% CO2) for 5 days post infection, after which time culture supernatant was harvested and cellular debris removed by centrifugation at 5000 x g for 10 minutes. For N-antigen detection, lOOμl samples of clarified culture supernatant were added to duplicate wells of a 96-well Maxi-Sorb plate; lOOμl of RIPA buffer was added per well with mixing and the plate was covered and incubated at 4°C overnight to enable protein binding to the plastic wells. The next day, the coating solution was discarded, wells were washed thoroughly with PBST, and blocking of unoccupied protein binding sites was performed by incubation in 1% BSA in PBS for 1.5 hours. The antibody recognising OC43 N-protein was used at 1/800 dilution in PBS (lhr at 3 C) and the secondary antibody (goat-anti- mouse alkaline phosphatase) was used for the colour development reaction. Optical density of the wells was read at 405 mn and the effect of compounds determined by comparison of the level of signal in presence of compound to level of signal from the solvent control,
Example 33: Effect of compounds in OC43 antiviral assay
Compounds were screened for activity against OC43 replication according to the method described in example 22. Results are shown in Table 11. Table 11
Figure imgf000090_0002
Example 34. Mouse Hepatitis Virus (MHV).
Synthesis and Purification of a Peptide Corresponding to the MHV-A59 E Protean.
A peptide corresponding to the full-length MHV-A59 E protein (sequence:
Figure imgf000090_0001
VLSPSIYLYDRSKQLYKYYNEEMRLPLLEVDDI; accession number NP_068673) was synthesized manually using FMOC chemistry and solid phase peptide synthesis The synthesis was done at the Biomolecular Resource Facility (John Curtin School of Medical Research, A I, Australia) using a Symphony11 Peptide Synthesiser from Protem Technologies Inc.(Woburn, MS, USA) according to the manufacturers instructions to give C-terminal amides, die coupling was done with HBTU and hydroxybenzotriazolc in N-mcthylpyrrolidone. Each of the synthesis cycles used double coupling and a 4-fold excess of the amino acids. Temporary α-N F oc- protecting groups were removed using 20% piperidine in DMF. The crude synthetic peptide was purified using the ProteoPlus™ kit (Qbiogene inc. CA), following manufactures instructions. Briefly, the peptides were diluted in loading buffer (60mM Tris-HCl H 8.3, 6M urea, 5% SDS, 10% glycerol, 0.2% Bromophenol blue, ± 100 mM β-mercaptoethanol) and run on 4-20% gradient polyacrylamide gels (Gradipore, NSW, Australia) in tris-glycine electrophoresis buffer (25 mM Tris, 250 M glycine, 0.1 % SDS). The peptides were stained with gel code blue (Promega, NSW) and the bands corresponding to the full-length peptide were excised out of the gel.
The gel slice was transferred to the ProteoPLUS™ tube and filled with tris- glycine electrophoresis buffer. The tubes were emerged in tris-glycine electrophoresis buffer and subjected to 100 volts for approximately 1 hour. The polarity of the electric curreait was reversed for 1 minute to increase the amount of protein recovered. The peptides were harvested and centrifiiged at 13, 000 rp for 1 minute. The purified peptides were dried in a Speedvac and the weight of the final product was used to calculate the yield.
Example 35; MHV-E protein forms ion channels in planar lipid bilayers.
Lipid. bilayer studies were performed as described elsewhere (Sunstrom, 1996; Miller, 1986). A lipid mixture of palmitoyl-oleoyl-phosphatidylethanolamine, palntitoyl-oleoyl-j^sphatidylserineandpahnitoyl-oleoyl-phosphatidylcholine (5:3:2) (Avanti Polar Lipids, Alabaster, Alabama) was Used, The lipid mixture was painted onto an aperture of 150-200 μm in the wall of a 1 ml delrin cup. The aperture separates two chambers, cis and trans, both containing salt solutions at different concentrations. The cis chamber was connected to ground and (he trans chamber to the input of an Axopatch 200 amplifier. Normally the cis chamber contained either 500 mM NaCl or 500mM KCl and the trans 50 mM NaCl or 50mM KCl. The bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis. The synthetic peptide was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 5000 Hz and stored on magnetic disk. The MHV E synthetic peptide was dissolved in 2,2,2-trifluorethanol (TFE) at 0.05mg ml to 1 mg ml. 10 μl of this was added to the cis chamber (1ml aqueous volume) of the bilayer apparatus, which was stirred via a magnetic "flea". Ionic currents, indicating channel activity in the bilayer, were typically detected within 15- 30 min. After channels were detected the holding potential across the bilayer was varied between -lOOmV and +100mV to characterise the size and polarity of current flow and enable the reversal potential to be determined.
In 14 experiments where the cis chamber contained 500mM NaCl solution and the trans chamber contained 50 mM NaCl solution, the average reversal potential of the channel activity was calculated to be 49 ±1 (SEM) mV. In 11 experiments where the cis chamber contained 500mM KCl solution and the trans chamber contained 50 mM KCl solution, the average reversal potential of the channel activity was calculated to be 13 ±6 (SEM) mV. These results indicate that the MHV E protein forms cation selective ion channels that are more selective for Na+ than for K* ions. Figure 11 shows examples of raw current data for the MHV E ion channel at various holding potentials (cis relative to trans) in asymmetrical NaCl solutions (500/50 M). The graph is a representative plot of average bilayer current (pA; y- axis) versus holding potential (mV; x-axis).
Example 36. Chemical compounds inhibit the ion channel actrvity of the MHV F. protein synthetic peptide.
To test compounds for their ability to block or otherwise inhibit the ion channel formed by MHV E protein, small aliquots of solutions containing the compounds were added to the aqueous solutions bathing planar lipids in which the peptide channel activity had been reconstituted and the effect of the compound addition on the ionic currents was recorded and measured. Compound stock solutions were typically prepared at 500 M in DMSO. This solution was further diluted to 50 mM, or lower concentration in 50% DMSO/50% methanol and 2 μl of the appropriately diluted, compound was added to the cis and/or trans chambers to yield the desired final concentration. hi the example shown in Figure 12 below, addition of lOOμM cinnamoylguanidine to the cis chamber greatly reduced current flow through the MHV E ion channel.
Example 37.Bacterial Bio-Assay for Screening Potential MHV E-protein Ion Channel-Blocking Drugs. MHV E-protein Ion Channel inhibits Bacterial Cell growth.
A bio-assay of MHV E-protein function in bacterial cells was developed. A synthetic cDNA fragment encoding MHV E-protein was cloned into the expression plasmid pPL451, creating a vector in which E protein expression is temperature inducible, as described in Example 4. Inhibition of the growth of E.coli cells expressing E protein at 37°C was observed as an indicator of p7 ion channel function dissipating the normal Na+ radient maintained by the bacterial cells.
Example 38. Compound Screening using the Bacterial Bio-Assav for MHV E protein.
The halos of growth around the site of application of particular drugs - as described in example 14 -were scored as decribed in example 15.
Table 12 lists the scores for inhibition of MHV E protein in the bacterial bio-assay.
Table 12
Figure imgf000093_0001
(trifluoromethyl)cinna oylguanidine 2.9 3-(cyclohex-l-en-l-yl)cinnamoylguanidine 2.9 trans-3-(l -napthyl)acryloylguanidine 2.8 4-(trifluorome!thyl)cinnamoylguanidine 2.8 3-(2-naρthyl)acryloylguanidine 2.8 2-(trifluoromethyl)cinnamoylguanidiήe 2.7 (4-Phenoxybeπzoyl)guanidine 2.4 (3-Bromocinnamoyl)guanidine 2.4 2,5-dimemyldnnamoylguanidine 2.3 5-bromo-2-fluorocinnamoylguanidine 2.1 6-methoxy-2-naphthoylguanidine 1.8 4-phenyIbenzoylguanidine 1.8 ' ( -Bromocinnamoyl)guanidine 1.8 1-napthoylguanidine . 1.7 (5-Phenyl-penta-2,4-dienoyl)guanidine 1.4 (2-Bromocinnamoyl)guanidine 1.4 (4-Chlorocinnamoyl)guanidine 1.3 2-methylcinnamoylguanidine 1.2 2,6-dichlorocinnamoylguanidine 1.2 2,4,6-trimethylcinnamoylguanidine 1,2 5-(N,N-hexamethylene)amiloride 1.1 cinnamoylguanidine 1.1 cinnamoylguanidine hydrochloride 1,1 (a-Methylcinnamoyl)guanidine 1.0 2,3-dimemylcinnamoylguanidine 1.0 2-cycIohexylcinnamoylguanidine 0.9 N-(3-phenylpropanoyl)-N'-ρhenylguanidine 0.8 N,N,-bis(3phenylpropanoyl)-N"-phenylguanidine 0.8 (3-Methoxycinnamoyl)guanidine 0.8 (2-Methoxycinnamoyl)guanidine 0.8 3-fluorocinnamoylguanidine 0.8 2-fluorocinnamoylguanidine 0.8 2,4-dichlorocinnamolyguanidinβ 0.8 2-ethylcinnarøoylguanidine 0.8 (2-Chlorocinnamoyl)guanidine 0.7 (4-Hydroxycinnamoyl)gHaπidine 0.7 2-ethoxycinnamoylguanidine 0.7 2-naρthoylguanidine 0.6
(traπs-2-PhenyIcyclopropanecarbonyl)guanidine 0.6 5-(N,N-Dimethyl)amiloride hydrochloride 0.5 5-(4-fluorσphenyl)amiloride 0.5 3-methylcinnamoylguanidiαe 0.5 (3-Chlorocinnamoyl)guanidine 0.4 4-methylcinnamoylguanidine 0.4 4-ethoxycinnamoylguanidine 0.4 2-(l-napthyl)acetoylguanidine
Figure imgf000094_0001
0.4
Figure imgf000094_0002
Figure imgf000095_0001
Example 39. MHV Antiviral Assay for testing compounds against replication of mouse hepatitis virus (MHV).
To determine the antiviral activity of compounds against MHV replication (strain MHV-A59: ATCC VR-764), an assay measuring reduction in the number of plaques formed in monolayers of MHV infected L929 cells (ATCC CCL-a) was developed: First, a virus working stock was prepared by amphfioation in NCTC clone 1469 cells (ATCC CCL-9.1). This was then used to infect confluent monolayers of L929 cells grown in 6-well tissue culture plates by exposure to the virus at an MOI of 0.01 pfu/cell or 1 pfu/cell for 30 minutes at 37°C in 5%C02. The infective inoculum was removed and replaced with fresh medium (DMEM supplemented with 10% horse serum) containing various test concentrations of compounds or the appropriate level of solvent used for the compounds (control). Plates were subsequently incubated at 37°C (in 5% CO2) for 16 - 24 hours post infection, after which time culture supernatant was removed and the cells were stained with 0.1% crystal violet solution in 20% ethanol for 10 minutes. Plaques were counted in all wells and the percentage reduction in plaque number compared to solvent control was calculated. Measurements were performed in duplicate to quadruplicate wells.
Example 40. Effect of compounds in MHV antiviral assay.
Table 13 provides the results obtained from this study. Tablel3
Figure imgf000096_0001
trans-3-Furanacryoylguanidine
N-amidmo-3-amino-5-hexamethyleneύnino-6- ρhenyl-2-ρyrazinecarboxamide
(2-Nitrocinnamoyl)guanidine
4-(trifluorθmethyl)cinna oyIguanidine
3,4-(methylenedioxy)cinnamoylguanidine
5-(N-Methyl-N-isobutyl)amiloride
(4-Chlorocinnamoyl)guanidine
2,4-dichlorocinnamolyguanidine
N-tS-phenylpropanoylJ-N -phenylguanidine
(3-Nitrociήπamoyl)guanidine
2-phenylcinnamoylguanidine
4-isoρroρylcinnamoylguanidine
3-(trifluoromethoxy)cinnamoylguanidine
3-(trifluoromethyl)cinnamoylguanidine
(4-Nitrocinnamoyl)guaπidine
3-(2-napthyl)acryloylguanidine
4-ethoxycinnamoylguanidine
2,6-dichlorocinnamoylguanidine
2,5-dimethylcinnamoylguanidine
(3-Bromocinnamoyl)guanidine
(3-ChlorocinnamoyI)guanidine
Figure imgf000097_0001
3-methylcinnamoylguanidine 90/1 88/ 1 39/1
(3-Methoχycinnamoyl)guanidine 92/2 87/2 37/3
2-t-butylcinnamoylguanidine N/D 98/2 37/1
[(E)-3-( -DimethylaminoρhenyI)-2- methylacryIoyl]guanidine 56/1 45/1 37/1
NjN'-bis(l -napthoyl)guanidine 58/1 52/2 35/1
3-methoxy -HMA 15/1 31/1 35/1
5-tert-butylamino-amiloridc 89/4 84/4 34/4
trans-3-(l -naρthyl)acryloylguanidine 95/2 86/3 34/3
6-methoxy-2-naphthoylguanidine 88/3 56/3 34/3
2-napthoylguanidine 67/2 36/2 34/2
2-ethylcinnamoylguanidine 96/ 1 81/2 34/1
2,3-dimethylcinnamoylguanidine 95/1 79/2 34/1
N'-Cinnamoyl-NJSf'-diphenylguanidine 97/1 72/2 34/1
3-isoρropylcinnamoylguanidine hydrochloride N/D 99/2 32/1
(4-Phenoxybenzoyl)guanidine 73/1 65/1 32/1
(trans-2-Phenylcyclopropanecarbonyl)guanidine 77/2 64/2 31/2
3-fluorocinnamoylguanidine 100/1 93/2 31/1
5-bromo-2-f!uorocinnamoylguanidine Toxic 81/2 31/1
N,N'-bis-(cinnamoyl)-N"-phenylguanidine 16/1 38/ 2 31/1
3-quinolinoylguanidine 27/1 36/2 30/1
2,4,6-trimethylcinnamoylguanidine 91/2 61/3 27/2 l-bromo-2-napthoylguanidine 31/1 27/2 27/1
N-amidino-3,5-diamino-6-ρhynyl-2- pyrazinecarboxamide 53/1 39/2 25/1
N-Cinnamoyi-N',N'-dimethylguanidine 92/2 65/3 24/2
(2-Methoxycinnamoyl)guaπidine 90/2 85/2 23/2
2-(2-napthyl)acetoylguanidine 52/1 20/2 23/1
4-ρhenyIcinnamoylguanidine 53/1 36/1 21/3
[3-(3-Pyridyl)acryloyl]guanidine 81/2 73/2 21/2
3,4,5-trimethoxycinnamoylguanidine 84/1 84/1 21/1
4-methylciιmamoylguamdine 93/1 89/1 20/1
4-fluorocinnamoylguanidine 86/1 83/1 20/1
2-methylcinnamoylguanidine 91/1 82/1 20/1
6-bromo-2-napthoylguanidine 65/1 37/2 19/1
5-(N,N-Dimethyl)amiloride hydrochloride 4274 7/4 17/4
(5-Phenyl-penta-2,4-dieήoyI)gύanidine 27/1 24/1 17/1
2-cyclohexylcinnamoylguanidine 100/1 74/2 16/1
5-(4~fluorophenyl)amilαride 4/1 25/1 16/1
Benzyoylguanidine 22/1 39/2 14/1
N-Benzoyl-NC-cinnamoylguanidine 0/1 0/1 14/1
5-(N,N-hexamethylene)amiloride 84/2 89/1 13/2
N-(cinnamoyI)-Nϊ>henylguanidine 83/1 88/ I 13/1
(4-Hydroxycinnamoyl)guanidine 19 /1 15/1 13/1
Figure imgf000100_0001
Figure imgf000101_0001
Example 41. Porcine Respiratory Coronavims (PRCV) Antiviral Assay for testing compounds against replication of porcine respiratory coronavirns fPRCV).
To determine the antiviral activity of compounds against porcine respiratory coronavirus replication (ATCC VR-2384), an assay measuring reduction in the number of plaques formed in monolayers of PRCV infected ST cells (procine fetal testis cell line, ATCC CRL-1746) was developed: Confluent ST cells in 6 well plates eϊe infected with a quaternary passage of porcme respiratory virus (PRCV) strain AR310 at three dilutions 10*1, 50"1 and 10"2 in PBSto provide a range of plaques numbers to count. lOOul of diluted virus was added r well in a volume of lml of media. Plates wereincubated for one hour on a rocking platform at room temperature to allow virus to adsorb to cells. The viral supernatant was removed and 2ml well of overlay containing 1% Seaplaque agarose in lx MEM, 5% FCS was added to each well. Compounds to be tested were added to the overlay mixture by diluting the compounds from frozen stock to a concentration so that the same volume of compound/solvent would be added to the overlay for each conceaitration of compound. The volume of compound/solvent never exceeded 0.07% of the volume of the overlay. The solvent used to dissolve compounds was DMSO and methanol mixed in equal proportions. Compounds were tested for anti-plaque forming activity at four concentrations, 0. luM, luM, lOuM and 20uM. Either duplicates or quadruplicates were performed at each concentration. Controls were performed where the same volume of solvent was. added to the overlay. The overlay was allowed to set at room temp for 20 ins. The plates were then incubated at 37°C for 2 days. The monolayers were then fixed and stained overnight at room temperature by adding Iml/well of 0.5% methylene blue, 4% formaldehyde. Overlay agarose and stain was then rinsed off to visualize stained and fixed monolayer
Example 42: Effect of compounds in PRCV antiviral assay
Compounds were screened for activity against PRCV replication according to the method described in example 29. Table 14 provides EC50 values for some tested compounds.
Table 14
Figure imgf000102_0001
Example 43. Bovine Coronavirus.
Antiviral Assay for testing compounds against replication of bovine coronavirus
(RCV\. To determine the antiviral activity of compounds against bovine coronavirus replication (ATCC VR-874), an assay measuring reduction in the number ofplaques formed in monolayers of BCV infected MDBK cells (bovine kidney cell line ;ATCC CCL-22) was developed: Confluent MDBK cells in 6 well plates were infected with a secondary passage of BCV with serially diluted virus diluted to 10"3, 5"5 and 10"4 in PBS to provide a range ofplaques numbers to count. lOOul of diluted virus was added pea* well in a volume of 1ml of media. Plates were incubated for one hr to allow virus to adsorb to cells. The viral supernatant was removed and 2ml well of overlay containing 1% Seaplaque agarose in lx MEM, 5% FCS was added to each well. Compounds to be tested were added to the overlay mixture by diluting the compounds from a 0.5M frozen stock to a concentration so that the same volume of compound/solvent would be added to the overlay for each concentration of compound. The volume of compound/solvent never exceeded 0.07% of the volume of the overlay. The solvent used to dissolve compounds was DMSO and raiethanol mixed in equal proportions. Compounds were tested for anti-plaque forming activity at four concentrations, O.luM, luM, lOuM and 20uM. Quadruplicates were performed at each concentration. Controls were performed where the same volume of solvent was added to the overlay. The overlay was allowed to set at room temp for 20 mins. The plates were then incubated at 37°C for 7 days. The monolayers were then fixed and stained by adding Iml well of 0.5% methylene blue, 4% formaldehyde.
Example 44: Effect of compounds in BCV antiviral assay
Compounds were screened for activity against BCV replication according to the method described in example 31. Table 15 provides EC50 values for some tested compounds.
Table IS
Figure imgf000103_0001
Examnle 45 Hepatitis C Virus
Ion channel activity of Hepatitis C vims P7 Protein
Testing of a Synthetic P7 Peptide for channel activity in artificial linid bilayers
A peptide mimicking the protein P7 encoded by the hepatitis C virus (HCV) 5 was synthesised having the following amino acid sequence:
AIENLVILNAASLAGTHGLVSFL FCFAWYLKGRWVPGAVYAFYGMWPLL LLLLALPQRAYA
Lipid bilayer studies were performed as described elsewhere (Miller, 1986). A lipid mixture of palrmtoyl-oleoyl-pho^hatidylethamolamine, pahnitoyl-oleoyl-
10 phosphatidylserme and palmitoyl-oleoyl-phosphatidylcholine (5:3:2) (Avanti Polar Lipids, Alabaster, Alabama) was used. The lipid mixture was painted onto an aperture of 150-200 m in the wall of a 1 ml dehrin cup. The aperture separates two chambers, s and trans, both containing salt solutions at different concentrations. The cis chamber was connected to ground and the trans chamber to the input of an Axopatch
15 200 amplifier. Normally the cis chamber contained 500 mM KCl and the trans 50 mM KCl. The bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis. The protein was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 0 2000 Hz and stored on magnetic disk. The P7 peptide was dissolved in 2,2,2- trifluorethanol (TFE) at lGmg ml. 10 ul of this was added to the cis chamber of the bilayer which was stirred. Channel activity was seen within 15-20 min.
When the P7 peptide was added to the cis chamber and stirred, channel activity was recorded. The potential in the trans chamber was -80 mV and the currents are 5 downwards. The currents reversed at +50 mV close to the potassium equilibrium potential in these solutions indicating that the channels were cation-selective. The amplitude of the open-channel peak is 1.7 pA corresponding to a channel conductance of about 14 pS. In most experiments, "single channels'* had a much larger size, presumably because of aggregation of the P7 peptide. The currents 0 reversed at about +40 mV in this experiment, hi some experiments the solution in the cis chamber was 1 0 mM KCl and 15 M KCl in the trans chamber. The P7 peptide again produced currents that reversed.
Similar results were obtained when both chambers contained NaCl. Currents recorded in an experiment when the cis chamber contained 500 M NaCl and the trans chamber 50 mM NaCL Again the currents reversed between +40 and +60 mV, close to the Na+ equilibrium potential indicating that channels were much more permeable to Na+ than to K+.
The channels formed by the P7 peptide were blocked by 5-(N,N- hexamethylene) amiloride (HMA),
Addition of the P7 peptide produced channel activity. Addition of 2 μl of 50 μM HMA to the cis chamber followed by stirring resulted in disappearance of the channel activity. Block of channel activity produced by the P7 peptide with 100 μM HMA was recorded in 4 experiments. In 2 experiments, sodium channels (500/50) were blocked by 500 μM HMA
When 10 mM CaCls was added to the cis chamber (K solutions) the reversal potential of the currents produced by P7 peptide shifted to more negative potentials indicating a decrease in the Pκ/Pα ratio,
When the cis chamber contained 500 mM CaCfe and the trans chamber 50 mM CaCfe, both positive and negative currents were seen at potentials around +20 mV and it was not possible to determine a reversal potential.
Example 46. Recombinant Expression of HCV P7 protein.
Two cDNA fragments, each encoding the same polypeptide corresponding to the amino acid sequence of the HCV-la p7 protein, were synthesised commercially by GeneScript. The two cDNAs differed in nucleotide sequence such that in one cDNA ("cDp7.coli") the codons were optimised for expression of the p7 protein in . E.coli while in the other cDNA (κcDp7,marø)" codons were biased for expression in mammalian cell lines. cDρ7.coli was cloned into the plasmid pPL451 as a BamHI/EcoRI fragment for expression in E.coli. cDp7.mam was cloned into vectors (for example, pcDNA3.1 vaccinia virus, pfasfBac-1) for expression of ρ7 in mammalian cell lines.
Example 47. Role of p7 in enhancement of Gag VLP Budding.
The budding of virus-like particles (VLP) from cultured HeLa cells results from the expression of retroviral Gag proteins in the cells and co-expression of small viral ion channels, such as M2, Vpu and 6K, with the Gag protein enhances budding. Interestingly, the viral ion channels can enhance budding of heterologous virus particles. Therefore, to assess budding enhancement by ρ7 it was co-expressed with the HIV-1 Gag protein in HeLa cells, and VLP release into the culture medium was measured by Gag ELISA. To achieve this, the plasmids pcDNAρ7 (pc DNA3.1 = pcDp7.mam as described in Example 20, p7 expressed under control of the T7 , promoter) and pcDNAGag (HTV-l Gag protein expressed under control of the T7 promoter) were cotransfected into HeLa cells infected with the vaccinia virus strain vTF7.3 (expresses T7 RN polymerase) and culture supernatauts were collected for ELISA assay after 16 hours incubation.
Example 48. Assay of the ability of compounds to inhibit p7 ion channel functional activity.
The two methods of detecting p7 ion channel functional activity, described in Examples 33-35, were employed to assay the ability of compounds to inhibit the p7 channel. In the case of Example 33, compounds were tested for their abihty to inhibit p7 channel activity in planar lipid bilayers. In the case of Example 35 compounds were tested for their ability to reduce the number ofVLPs released from cells expressing both p7 and HTV-l Gag. Example 49.
Bacterial Bio-Assav for Screening Potential HCV P7 protein Ion Channel- Blocking Drugs.
HCV D7 Ion Channel inhibits Bacterial Cell growth. A bio-assay of p7 fonction in bacterial cells was developed. The p7-encoding synthetic cDNA fragment cDp7.coli was cloned into the expression plasmid pPL451, creating the vector pPLp7, in which p7 expression is temperature inducible, as described in Example 4. Inhibition of the growth of E.coli cells expressing p7 at 37'C was observed as an indicator of p7 ion channel function dissipating the normal Na+ gradient maintained by the bacterial cells.
Example 50 Compound Screening using the Bacterial Bio-Assav for HCV p7 protein.
The halos of growth around the site of application of particular drugs - as described in example 14 - were scored as decribed in example 15.
Table 16 lists the scores for inhibition of HCV ρ7 protein in the bacterial bio-assay.
Table 16
Figure imgf000107_0001
Figure imgf000108_0001
2-(cyclohex-l -eιι-lyl)cinnamoylguanidine 1.00 / 1
2-napthoylguanidine 1.0 /3
3-phenylcinnamoylguanidine 1.0/1
5-(N,N-Dimethyl)amiIoride hydrochloride 1.0 /1
5-(4-fluorophenyl)amiloride 1.0 /1
(3-Methoxycinnamoyl)guanidine 1.0 /1
2-fluorocinnamoylguanidine 1.0/1
5-(3'-bromophenyl)penta-2,4-dienoylguanidine 1,0/1
[(4-Chlorophenoxy-acetyl]guanidine 1.0 /1
(3-phenylprøpanoyl)guamdine 1.0 / 1
2"chloro-6-fluorocinnamoylguanidirie 0.88 /1
3-fluorødnnamoyIguanidine 0.S6/1
2-methylcinnamoylguanidine 0.75 /1
(2-Methoxycinnamoyl)guanidine 0.75 /1
1 -brømo-2-napthoylguanidine 0.75/1
3,4,5-trimethoxycinnatnoylguanidine 0.71 /l
3-methylcittnamoylguanidine 0.63 /l
3-(trans-hept-l-en-l-yl)cinnamoylguaπidine 0.50/1
AmiloridcHCl 0.5 /2
Phenamil methanesulfonate salt 0.5 /1
2,4-dichlorocinna olyguanidine 0.38/1
(4-Nitrocinnamoyl)guanidine 0.25/1
3,4-difluorociπnamoylguanidine 0.13 /1
[(E)-3-(4-Dimethylaminophenyl)-2-
{taethylacryloyl]guanidine 0.03 /4
Example 51: Equine Arteritis Virns (EAV)
Antiviral Assay for testing compounds against replication of equine arteritis virus (EAV).
5 To determine the antiviral activity of compounds against EAV replication
(strain Bucyrus; ATCC VR-796), an assay measuring reduction in the number of plaques formed in monolayers of EAV infected BHK-21 cells (ATCC CCL-10) was devebped: A virus stock was amplified in RK-13 cells (ATCC CCL-37) and this was then used to infect confluent monolayers of BHK-21 cells grown in 6-well tissue .
10 culture plates by exposure to the virus at an MOI of 5X10"3 pfii cell for 1 hour at 37°C 5% COj. The infective inoculum was removed and nd the cells were overlayed with a 1% sea plaque overlay (Cambrex Bio Science) in MEM containing 10% FCS containing and 10, 5 or 1 μM of compounds to be tested or the appropriate level of solvent used for the compounds (control). Plates were subsequently incubated at
15 37βC (in 5% C02) for 3 days post infection, after which time culture supernatant was . removed and the cells were stained with 0.1% crystal violet solution in 20% ethanol for 10 minutes, Plaques were counted in all wells and the percentage reduction in plaque number compared to solvent control was calculated. Measurements were performed in duplicate to quadruplicate wells.
Example 52: Effect of compounds in EAV antiviral assay
Compounds were screened for activity against EAV replication according to the method described in example 35. Results expressed as IC50 values are shown in Table 17.
Table 17
Figure imgf000110_0001
Example 53 Dengue Flavivirπs
Peptide Synthesis of Dengue virus M Protein The C- terminal 40 amino acids of the M protein of the Dengue virus type 1 strain Singapore S275/90 (Fu et al 1992)
(ALRHPGFWIALFLABAIGTSITQKGHFH MLVTPSMA) was synthesised using the Fmoc method. The synthesis was done on a Symphony Peptide Synthesiser form Protein Technologies Inc (Tucson, Arizona) as used to give C-terminal amides, the coupling was done with HBTU and hydroxybenzotriazole in N-methylpyrrolidone. Each of the synthesis cycle used double coupling and a 4-fold excess of the amino acids. Temporary α-N Fmoc-protecting groups were removed using 20% piperidine in DMF.
Example 54. Incorporation of Dengue M virus protein into lipid bilayers. Lipid bilayer studies were performed as described elsewhere (Sunstrom, 1996;
Miller, 1986), A lipid mixture of palmitoyl-oleoyl-phosphatidylethanolantine, palmitoyi-oleoyl-phosphatidylserine and palmitoyl-oleoyl-phαsphatidylcholin . (5:3:2) (Avanti Polar Lipids, Alabaster, Alabama) was used. The lipid mixture was painted onto an aperture of 150-200 μm in the wall of a 1 ml delrin cup. The aperture separates two chambers, cis and trans, both containing salt solutions at different concentrations. The cis chamber was connected to ground and the trans chamber to the input of an Axopatch 200 amplifier. Normally the cis chamber contained 500 M KCl and the trans 50 mM KCl. The bilayer formation was monitored electrically by the amplitude of the current pulse generated by a current ramp. The potentials were measured in the trans chamber with respect to the cis. The protein was added to the cis chamber and stirred until channel activity was seen. The currents were filtered at 1000 Hz, digitized at 5000 Hz and stored on magnetic disk.
The dengue virus M protein C-terminal peptide (DMVC) was dissolved in 2,2,2- trifluorethanol (TFE) at 0.05mg ml to I mg ml. 1 μl of this was added to the c s chamber of the bilayer which was stirred. Channel activity was seen within 15-30 min,
Eτamτιle 55: Hexamethylene amiloride (HMA) to inhibits ion channel activity of the dengue virus M protein C-termfnal peptide.
Solutions of 50 mM HMA were prepared by first making a 500 inM solution in DMSO. This solution was further diluted to 50 mM HMA using 0.1 M HCI.2 μl of the 50 mM HMA was added to the cis chamber after channel activity was seen. The cis chamber contained 1 ml of solution making the final concentration of HMA 100 μM.
Example 56. Antiviral Assay for testing compounds against Effects of Dengne flavivirns against cvtoproliferation.
Compounds were tested at 10, 5, 2.5, 1.25 and 0.625 μM for activity against Dengue 1 strain Hawaii using a cytoproliferation assay which measures the effect of dengue virus infection on proliferation of LLC-MK2, rhesus macaque monkey kidney cells. The LLC-MK2 cells were infected with a predetermined amount of vims, titrated such that cell proliferation in infected cultures would be significantly reduced -Ill- compared to uninfected controls. The infected cells were then plated at 1.5xl03 cells per well in a 96 well plate. Negative controls (no virus, no experimental compound), positive controls (virus, no experimental compound), and cytotoxicity controls (experimental compound, no virus) were run with each drug assay. The cultures were allowed to grow for 7 days and then Alamar Blue, a fluorescent dye that measures the metabolism of the cultures (red/ox), was added to each culture and the fluorescence value for each culture was measured. The negative control without experimental compound or virus was fixed at 100%. The positive controls and the cultures with compound were scored by calculating their average fluorescence as a percentage of the negative control. At least six replicate wells were measured for each experimental condition.
Example 57 Effect of compounds in Dengue antiviral assay:
Table 18
Figure imgf000112_0001
412-
Figure imgf000113_0001
N.A. - not applicable
Example 58: Positive correlation between Bacterial Assay and Anti-viral Assays
Example 58.1 Positive correlation between Vpn Bacterial Assay and antl-HIV-1 Data.
A correlative study was performed to measure correlation between the activity scores assigned to compounds tested in the Vpu bacterial assay and the abihty of these compounds to inhibit HIV-1 in the anti-viral assay.
Example 58.2. Methodology
The p24-antigeώ data for twelve compounds representing various substituted acylguanidines was compared with the activity scores obtained for those compounds in the Vpu bacterial assay. The data from each assay was initially rank ordered for effectiveness. The rank order for the Vpu bacterial assay was determined from all activity scores, the highest score indicating the greatest effectiveness. The rank order for the anti-HIV-1 assay was determined based on the overall average value of p24 antigen measured in culture supβmatants at all of the drug concentrations tested, with the lowest score indicating the greatest effectiveness. The two rank orders generated ware then compared statistically by generating the Spearman's Rank correlation coefficient.
Example 58.3. Results and Conclusion
The Spearman's correlation coefficient was 0.785 which, by comparison with a statistical table of critical values (for n=l 2), indicates that the two rank orders are significantlypositivel cotrel^ed (P<0.01) (Table 19a).
In addition, a different comparison of the Vpu Bacterial assay rank order with a yes/no score for whether the anti-viral data indicated a p24 reduction of at least one order of magnitude, aligned the 'yes' group of compounds with the top 6 compounds by the bacterial assay (Table 19b). These results are indicative that a positive correlation exists between bacterial assays and the antiviral assays as perfoπnedaccording to the present invention. The bacterial assay may therefore be a useful tool in screening for compounds that exhibit anti-viral activity.
Table 19a. Comparison of Rank order of efficacy of 12 substituted acylguanidines in the Vpu bacterial assay and anti-HTV assay.
Figure imgf000114_0001
Table 19b.
Figure imgf000114_0002
Figure imgf000115_0001
Example 58.4. Correlation Between Percent inhibition of MHV plaque formation and MHV-E bacterial bio-assav score.
A positive correlation was seen between the activity scores assigned to compounds when tested in the Mouse Hepatitis Virus E-protein bacterial bio-assay and the percent inhibition exhibited by these compounds in the Mouse Hepatitis Virus plaque assay.
Example 58.5. Method:
MHV plaque reduction activity data for 96 compounds screened were sorted from greatest to least percent plaque reduction and rank orders were assigned to the list of compounds. This was performed for the data generated by exposure to both lOμM and 1 μM concentrations of the compounds, giving rise to two rank order lists. Similarly, a rank order list was generated for the MHVE bacterial bioassay scores for the same 96 compounds. Where one or more compounds had the same score, the rank values for that group were averaged.
Spearman's statistical test for [as described in "Mathematical Statistics with
Applications" (2nd edn): Mendenhall, W., Scheaffer, RL.,& Wackerly, DD. Duxbuiy Press, Boston Massachusetts - 1981] was used to compare rank orders. Briefly, this involved calculating the Sum of squares (SS) of the differences between rank values for . each compound, and then generating the Spearman's Rank Correlation coefficient (Rs) according to the formula: Rs * l-(6.SS n(n2-l)), where n is the number of compounds ranked (96 in this case). Rs is then compared to a Table of critical values to determine statistical significance (P values).
Example 58.6. Summary of Results: This table summarises the Rs and P values generated as a result of the indicated pairwise comparisons between rank orders. Table 20
Figure imgf000116_0001
Example 58.7. Conclusions:
The rank order comparison of 96 compounds assayed in the bacterial bioassay and the antiviral assay show that MHVE bacterial assay rank order for the compounds tested is significantly positively correlated with the rank orders generated by the MHV plaque reduction assay. The significant correlation between the assays is highly indicative that either assay may be utilised to identity compounds that may be useful. The bacterial assay may thereby be a useful tool in screening for compounds that exhibit anti-viral activity. -
Example 58.8. Correlation Between Percent inhibition of 229E plaque formation and 229E-E bacterial bio-assay score.
A positive conelation was seen between the activity scores assigned to compounds when tested in the Human Coronavirus 229E E-protein bacterial bioassay and the percent inhibition exhibited by these compounds in the Human Coronavirus 229E plaque assay.
Example 58.9. Method:
229E plaque reduction activity data for 97 compounds screened against 2.5 μM compound concentration were sorted from greatest to least percent plaque reduction and rank orders were assigned to the list of compounds. Similarly, a rank order list was generated for the 229E E bacterial bioassay scores for the same 97 compounds. Where one or more compounds had the same score, the rank values for that group were averaged.
Spearman's statistical test for [as described in "Mathematical Statistics with Applications" (2nd edn): Mendenhall, W., Scheaffer, RL.,& Wackerly, DD. Duxbury Press, Boston Massachusetts- 1981] was used to compare rank orders. Briefly, this involves calculating the Sum of squares (SS) of the differences between rank values for each compound, and then generating the Spearman's Rank Correlation coefficient (Rs) according to the formula: Rs = l-(6.SS/n(n2-l)), where n is the number of compounds ranked (97 in this case). Rs is then compared to a Table of critical values to determine statistical significance (P values).
Example 58.9.1. Summary of Results and Conclusions This table summarises the Rs and P values generated as a result of the indicated pairwise comparisons between rank orders. Table 21
Figure imgf000117_0001
The results above indicate that the 229E bacterial assay rank order for the compounds tested is significantly positively correlated with the rank orders generated by the 229E plaque reduction assay. This result combined with that shown in Examples 49.1 and 49.4, provides strong evidence that either assay may be utilised to identity compounds that may be usefiil. The bacterial assay may thereby be a useful tool in screening for compounds that exhibit anti-viral activity.
Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described. It is to be understood that the invention includes all suoh variations and modifications The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations of any two or more of said steps or features. BIBLIOGRAPHY
Barr , M„ Mulcahy, F. and Back, D - J., Br J Clin Pharmacol, 45: 221 (1998)
Deeks, S,G„ West J Med, 168:133 (1998)
Miles, S,A„ J Acquir Immune Defic Syndr Hum Retrovirol, 16 Suppl 1 : S36 (1 97) Miles, S.A., J. Acquir Immune Defic Syndr Hum Retrovirol, 16 Suppl 1: S I (1998)
Moyle, G.J., Gazzard, B.G., Cooper, D.A. and Gatell, L, Drugs, 55:383 (1998)
Rachlis, A.R. and Zarowny, D.P., Cmaj, 158:496 (1998)
Vella, S., Fragola, V. and Palmisano, L., J Biol Regul Homeost Agents, 11 :60 (1997)
Volberding, P.A. and Deeks, 5.G., Jama, 279:1343 (1998) Volberding, P.A., Hosp Pract (Off Ed), 33:81-4, 87-90, 5-6 passim. (1998)
Miller, R.H. and Sarver, N., Nat Med, 3:389 (1997)
Mitsuya, H., Enzyme Inhibition, 6:1 (1992)
Moore, IP., Science, 276:51 (1997)
Thomas, M. and Brady, L., Trends Biotechnol., 15:167 (1997) Balliet, J. W., Kolson, D.L., Eiger, G„ Kim, F.M., McGann, K. A., Srinivasan, A. and
Collman, R., Virology, 200:623 (1994)
Wcstcrvclt, P., Hcnkcl, T., Trowbridgc, D.B., Orcnstcin, J., Heuser, , Gendclman,
H.Rand Ratner, L,, J Virol, 66:3925 (1992)
Ewart, G.D., Sutherland, T., Gage, P.W. and Cox, G.B., J Virol, 70:7108 (1996) Schubert, U„ Henklein, P., Boldyreff, B., Wingender, E., Strebel, K. and Porstmann,
T. J Mol Biol, 236:16 (1994)
Friborg, J., Ladha, A., Gottlinger, H., Haseltine, W.A. and Cohen, E.A., Journal of
Acquired Immune Deficiency Syndromes & Human Retrovirology, 8:10 (1995)
Fu ., Tan»B-H., Yap,E.H., Chan,Y.C. and Tan,Y.H. (1992) Full-length cDNA sequence of dengue type 1 virus (Singapore strain S275/90). Virology 188 (2), 953-
958
Love, C.A., Lilley, P.E. and Dixon, N,E„ Escherichia coli. Gene, in press. (1996) Yamato, I., Kotani, M., Oka, Y. and Anraku, Y., Escherichia coli. Journal of Biological Chemistry, 269:5729 (1994) Rosen, B,R„ ATP-coupled solute transport systems. Escherichia coli and Salmonella typhimuriu : Cellular and molecular biology 1: (1987) Editor: Neidhardt, F.C. American Society for Microbiology.
Piller, S.C., Ewart, G.D., Premkumar, A., Cox, G B. and Gage, P.W., Proceedings of the National Academy of Sciences of the United States of America, 93:111 (1996) Lu, Y.A., Clavijo, P., Gatantino, M., Shen, Z.Y., Liu, W. and Tarn, J.P., Molecular Immunology, 28:623 (1991)
Harlow, E, and Lane, D., (1988) Antibodies: A laboratory manual, (ed). Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 'Vol:'.
Varadhachary, A. and Maloney. P -C, Molecular Microbiology, 4:1407 (1990) - 44 New, R.C.C., (1990). Liposomes: A practical approach. The Practical Approach Series.
Kuhn RJ, Zhang W, Rossmann MG, Pletnev S V, Corver J, Lenches E, Jones CT, Mukhopadhyay S, Chipman PR, Strauss EG, Baker TS, Strauss JH. (2002). Structure of dengue virus: implications for flavivirus organization, maturation, and fusion. Cell.2002 Mar 8;108(5):717-25. Rickwood, D., Harries, B. D. (eds). IRL Press, Oxford.
Miller, C, (1986) Ion channel reconstitution. (ed). Plenum Press, New York and London.
Fear, W.R., Kesson, A.M., Naif, H., Lynch, G.W. and Cunningham, A.L., J. Virol,
72:1334 (1998) Kelly, MJD., Naif, H., Adams, S.L., Cunningham, A.L. and Lloyd, A.R., J. Immunol, 160:3091 (1998)
New, R.C.C. (ed.), Liposomes: a practical approach. IRL Press, Oxford (1990) Grice, A.L., Kerr, I.D. and Sansom, M.S., FEBSLett, 405(3):299-304 (1997) Moore, P.B., Zhong, Q., Husslein, T. and Klein, M.L., FEBS Lett, 431(2): 143-148 (1998) Schubert, U., Bour, S., Ferreπnαntiel, A.V., Montal, M., Maldarelli, F. and Strebel, K.,Joumal of Virology, 70(2):809-819 (1996a)
Sunstrom NA, Premkumar LS, Premkumar A, Ewart G, Cox GB, Gage PW (1 96), Ion channels formed by NB, an influenza B virus protein. J Merøbr Biol. 1996 Mar,150(2):127-32 Willbold, D., Hofftøann, S. and Rosch, P., Eur J Biochem, 245(3):581-8 (1997) Wray, V., Kinder, R., Federau, T., Herύdein, P. . Bechmger, B. and Schubert, U., Biochemistry, 38(lό):5272-82 (1999)

Claims

THE CLAIMS DEFINING THE INVENTION ARE AS FOLLOWS:-
1. An acylguanidine with antiviral activity.
2. An antiviral compound of Forumula I
Figure imgf000121_0001
wherein 1-R4 are independently aromatic groups, heteroaromatic groups, alkylaromatic groups, alkylheteroaromatio groups, alkenylaromatic groups, alkenylheteroaromatic groups, cycloalkylaromatic groups, cycloalkylheteroaromatic groups, aryloxyalkyl groups, heteroaiyloxyalkyl groups, said groups are mono or polycyclic, and are optionally substitiited with one or more substitutents independently selected from hydrogen, hydroxy, nitro, halo, amino, substituted amino, alkyl-substituted amino, cycloalkyl-substituted amino, aryl-substituted amino,
Figure imgf000121_0002
. βalkyloxy, C3.6cycloaJkyl, halo-substituted Chalky!, halo-substituted Ci. - eal ylo y, phenyl, C^alkeneyl, Cs-^cycloalkeneyl, Ci-ealkeneoxy, benzo,
aryl, substituted aryl, PrS,
Figure imgf000121_0003
3. An antiviral compound of Formula I
Figure imgf000121_0004
or pharmaceutically acceptable salts thereof, wherein,
Rι =
Figure imgf000122_0001
R2 , R3 and 4 are independently hydrogen,
Figure imgf000123_0001
and wherein
X = hydrogen, hydroxy, nitro, halo, Chalky!,
Figure imgf000123_0002
C3-6cycloalkyl, halo-substituted Ct^alkyl, halo-substituted Ci- 6aJkyloxy, phenyl, Cι.6alkeneyl, C3-<$cycloalkeneyl,
Figure imgf000123_0003
or bønzo; s , Et , Re , d , RB , Rf , Rh , Rk , RL , IM , a , Ro , RP independently * hydrogen, amino, halo, d-salkyl, Ci-salkyloxy, hydroxy, aryl, substituted aryl, substituted amino, mono or dialkyl-substituted amino, cycloalkyl-substituted amino, aryl-substituted amino,
Figure imgf000123_0004
Rg , Ri independently = hydrogen, hydroxy, halo, or Ci^ alkyl; j = hydrogen, amino, halo, Cι.salkyl,
Figure imgf000123_0005
hydroxy, aryl, substituted aryl, substituted amino, alkyl-substituted amino, cycloalkyl-substituted amino, aryl-substituted amino, PrS,
Figure imgf000123_0006
423-
4. A pharmaceutical composition comprising an antiviral compound according to any one of claims 1 to 3, and optionally one or more pharmaceutical acceptable carriers or derivatives,
5. The pharmaceutical composition according to claim 4, further comprising one or more known antiviral compounds or molecules.
6. A method for reducing, retarding or otherwise inhibiting growth and/or replication of a virus comprising contacting a cell infected with said virus or exposed to said virus with a compound according to any one of claims 1 to 3.
7. The method according to claim 6, wherein said virus is a Lentivirus.
8. . The method according to claim 7, wherein said Lentivirus is Human
Immunodeficiency Virus (HIV).
9. The method according to claim 8, wherein said compound is selected firom the group consisting o :
(3-Chlorocinnamoyl)guanidine, (3-Bromocinnamoyl)guanidine, (2-Chlorocinnamoyl)guanidine, (2-Bromocinnamoyl)guanidine, 3-(trifluoromethyl)cinιιamoyIguanidine, 5-brαmo-2-fluorocinnamoylguanidine, 3-methylcinnamoylguanidine, 2-methylcinnamoylguanidine, 2,3-dimethylcinnamoylguanidine, Cinnamoylguanidine, 6-methoxy-2-naphthoylguanidine, trans-3-(l-napthyl)acryloylguanidine, 3 ,4-dichlorocinnamoylguanidine, . 2,6-dichlorocinnamoylguanidine, 4-phenylbenzoylguanidine, 2-ethylcinnamoylguanidine, (4-C orocinnamoyl)guanidine, 2-napthoylguanidine, 2,5-dimefhylcinnamoylguanidihe, 3-isoρropylcinnamoylguanidine hydrochloride, (5-Phenyl-penta-2,4-dienoyl)guanidine, 3-phenylcinnamoylguanidme, (4-Bromocinnamoyl)guanidine, 5-(3-bromophenyl)ρenta-2,4-dienoylguanidine, 3-(cyclohe5£-l-ea-l-yl)cinnamoylguanidine, 3-(trifluoromefhoxy)cinnamoylguanidine, 2-(trifluoromethyl)ciιmamoylguanidine, N,N'-bis(3ρhenylρroρanoyl)-N"-phenylguanidine, 2-ethoxycannamoylguanidine, N-tS-phenylpropanoyl^N'-phenylguanidine, 4-(trifluoromethyl)cinnamoylguanidinc, (4-Methoxycinnamoyl)guanidine, , 2-t-butylcinnamoylguanidine, 4-methylcinnamoylguamcline, 2-fluorocinnamoylguanidine, 2-phenylcinnamoylguanidine, N-fβ-Hydroxy^-napthoyl N'-phenylguanidine, 3-t-butylcinnamoylguanidine, 3,4-difluαrocinnamoyIguanidine, 5-(N,N-hexamethylene)amiloride, 3-fluorocinnamoylguanidine, 5-bromo-2-methoxycήmamoylguanidine, 3-ethoxycinnamoylguamdine, 3,4-(meihylenediόxy)cinnamoylguanidine, (2-Methoxycinnamoyl)guanid e, 2*4 DichloroBenzamil HCI, 2,3,5,6,-tetramethylcinnamoylguanidine, 3-(2-napthyl)acryloylguanidine, 2-(l-naρthyl)acetoylguanidine, 2,3-difluorocinnamoylguanidine, (3-Methoxyciππamoyl)guanidine, 4-isopropylcinnamoylguanidine, 2,4,6-ϋimethylcinnamoylguanidine, N-ζcinnamoylJ-N^phenylguanidine, 2-(cyclohex-l-e!n-lyl)cinnamoylguanidine, 2-(2-napthyl)acetoylguanidine, (4-Hydroxycinnamoyl)guanidine, 4-phenyiciπmamoylguanidine, 4-fluoroeinnamoylguamdine, N^N'-bis-(cinnamoyl)-N"-ρhenyIguanidine, (2-Furanacryloyl)guanidine, Phenamil methanesυlfonate sal , Benzamil hydrochloride, (3-Nitrocinnamoyl)guanidine, Benzyoylgπanidine, (4-Phenoxybenzoyl)guanidine, 3-(trans-heρt-l-en-l-yl)cinnamoylguanidine, 5-(N-Me l-N-isobutyI)amiloride, 2-cycϊohexylcύmamoylguanid e, 4-ethoxycinnamoylguanidine, 2,4-dichlorocinnamolyguanidine, 5-(N-Ethyl-N-isopropyl)amiloride, N-amidino-3-amino-5-hexamethyleneimino-6-ρhenyl- 2-pyrazinecarboxamide, (a-Methylcinnamoyl)guaniditte, cinnamoylguanidine hydrochloride, [(4-Chloroρhenoxy-acetyl]guanidine, N-amidino-3-amino-5-phenyl-6-chloro-2- pyrazinecarboxamide, 5-(4-fluorophenyl)a iloride, (trans-2-Phenylcycloρropanecarbonyl)guanidinc, (2-Nltiocinnamoyl)guanidine, trans-3-Furanacryoylguanidiue, 1-napthoylguanidine, 5-tert-butylamino-amiloride, 3-methoxy -HMA, (3-ρhenylρroρanoyl)guanidine, 4-t-butylcinnamoylguanidine, 5-(N,N-Dimethyl)amiloride hydrochloride, - N,lNF-Bis(3-phenylpropanoyl)guanidine, N-Benzoyl-N-cir amoylguanidineand l-bromo-2-napthoylguanidine.
10. The method according to claim 8, wherein said compound is selected from the group consisting of 4-phenylbenzoylguanidine, (3- bromocinnamoyl)guanidine, 3-(trifluoromethyl)cinnamoylguanidine, 5- (NjN-hexamethylene)amiloride, and (5-Phenyl-penta-2,4- dienoyl)guanidine.
11. The method according to any one of claims 8 to 10, wherein said HIV is HIV-1.
12. The method according to claim 6 wherein said virus is a Coronavirus.
13. The method according to claim 12, wherein said Coronavirus is the Severe Acute Respiratory Syndrome virus (SARS).
4. The method according to claim 13, wherein said compound is selected from the group consisting of
2,3-difluorocinnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
4-t-butylcinnamoylguanidine,
3-(2-napthyl)acryloylguanidine,
(3-Chlorocinnamoyl)guanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
2,5-dimethylcinnamoylguanidine, trans-3-(l-napthyl)acryloylguamdine,
4-isopropylcinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
6-methoxy-2-naphthoylguanidihe,
5-(N-Methyl-N-isobutyl)amiloride,
3-phenylcinnamoylguanidine,
(2-Chlorocinnamoyl)guanidine,
2'4 DichloroBenzamil HCI,
4-ρhenylcinnamoylguamdine,
4-(trifluoromethyl)cinnamoylguanidine,
3-(trifluoromethoxy)cinnamoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
2-ethoxycinnamoylguanidine, cinnamoylguanidine hydrochloride,
4-ethoxycinnamoylguanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
3,4,5-trimefhoxycinnamoylguamdine,
5-tert-butylamino-amiloride,
3-t-butylcinnamoylguanidine,
5-bromo-2-fluorocinnamoylguarιidine,
(4-Chlorocinnamoyl)guanidine,
2-t-butylcinnamoylguanidine,
2-cyclohexylcinnamoylguanidine, .
6-Iodo amiloride,
3-(trans-hcpt-l-cn-l-yl)cinnamoylgttani<ϋne,
(4-Bromocinnamoyl)guanidine,
(4-Hydroxycinnamoyl)guanidine,
N-tS-phenylpropanoylJ-N'-phenylguanidine,
(3-Nitrocinnamoyl)guanidine,
3-fluoro nnamoylguanidine,
2-(l -napthyl)acetoylguanidine,
2-ethylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
2-napthoylguanidine,
5-(4-fluoroρhenyl)amiloride,
2-(trifluoromethyl)cinnamoylguanidine, N-(6-Hydroxy-2-napthoyl)-N'-phenylguamdine,
(trans-2-Phenylcycloρroρanecarbonyl)guanidine,
N,N'-bis(3phenylpropanoyl)-N"-phenylguanidine,.
1-napthoyϊguanidine,
Benzamil hydrochloride,
3-mefhoxy -HMA,
4-methylcinnamoylguanidine,
4-fluorocinnamoylguanidine,
3,4-(methylenedioxy)cinnamoylguanidine,
5-(N,N-hexamethylene)amiloride,
N-(cinna oyl)-N'phenylguanidine,
5-(N-Ethyl-N-isoρropyl)amiloride,
3-methylcinnamoylguanidine,
2-methylcinnamoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine, trans-3-Furanacryoylguanidine,
(4-Methoxycinnamoyl)guanidine,
(2-Furanacryloyl)guanidine,
(3-phenylpropanoyl)guanidine,
2-(2-napthyl)acetoylguanidine,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
[3-(3-Pyridyl)acryloyl]guanidine,
4-phenylbenzoylguanidine,
2,4-dichlorocinnamolyguanidine,
(3-Methoxycirmamoyl)guanidine,
2-fluorocinna oylguanidine,
(4-Phenoxybenzoyl)guanidine,
(a-Methyloinnamoyl)guanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
(5-Phenyl-ρenta-2,4-dienoyl)guanidine,
(Quinoline-2-carbonyl)guanidine,
(Phenylacetyl)guanidme,
N,N'-Bis(amidino)napthalene-2,6-dicarboxaraide,
6-bromo-2-napthoylguanidine, l-bromo-2-napthoylguanidine,
2-chloro-6-fluorocinnamoylguanidine,
[(4-Chlόrophenoxy-acetyl]guanidine,
Phenamil methanesulfonate salt ,
N-Benzoyl-N'-cinnamoylguanidine and
N-(2-naρthoyl)- -phenylguanidine.
15. The method according to claim 13, wherem said compound is selected from the group consisting of cinnamoylguanidine, trans-3-(l- napthyl)acryloylguanidine, and 6-methoxy-2-naphthoylguanidine.
16. The method according to claim 12, wherein said Coronavirus is human Coronavirus 229E.
17. The method according to claim 16, wherein said compound is selected from the group consisting of
4-isopropylcinnamoylguanidine,
3,4-dicMorocinnamoylguanidine,
3-(trifluoromethoxy)ciπnamoylguanidhιe,
4-t-butylcinnamoylguanidine,
3-isoρroρylcinnamoylguanidine hydrochloride,
3-t-butylciππamoylguanidine,
2-t-butylcinnamoylguanidine, trans-3-(l -naρthyl)acryloylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
2,3-difluorocinnamoylguanidine,
3-(2-napthyl)acryloylguanidine,
2-phenylcinnamoylguanidine,
3-phenylcinnamoylguanidine,
3-(cyclohex-l-en-l -yl)cinnamoylguanidine, ' 4-phenylbenzoylguanidine,
3-(trifluonmιelhyl)cinnamoylguamdine,
(4-Phenoxybenzoyl)guanidine,
4-(trifluoromethyl)cinnamoylguamdine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
1 -napthoylguanidine,
5-(4-fluorophenyl)amiloride,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
(3-Bromocinnarøoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
2-(trifluoromethyl)cinnamoyIguanidine,
6-methoxy-2-naphthoylguanidine, . (4-Chlorocinnamoyl)guanidine,
(3-Methoxycinnamoyl)guanidine,
5-bromo-2-fluorocinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
(a-Methylcinnamoyl)guanidine,
4-phenylcinnamoylguanidine,
2,6-dichlorocinnamoylguanidine,
(2-Bromocinnamoyl)guanidine,
2,4,6-trimethylciιuιamoylguanidine, (trans-2-Phenylcyclopropanecarbonyl)guanidine,
(3-Chlorocinna oyl)guanidine,
2-(l-napthyl)acetoylguanidine,
2-ethylcinnamoylguanidine,
2-cyclohexylcinnamoyIguanid e,
(4-Hydroxycinnamoyl)guanidine,
2-ethoxydnnamoylguamdiπe,
3-methyiciαnaraoylguanidine,
2-methylciπnamoylguanidine,
3-fluorocinnamoylguanidine, cinnamoylguanidine hydrochloride,
2,3-dimefhylcinnamoylguamdine>
2-fluorocdnnamoylguanidine,
4-fluorocinnamoyϊguanidine,
3,4-difluorocinnamoylguanidine,
5-tert-butylamino-amiloride,
2-napthoylguam'dine,
N,N'-Bis(amidino)napthalene-2,6-dicarboxamide,
N,N'-Bis(3-ρhenylρrqpanoyl)guanidine,
4-methylcinnamoylguanidine,
5-(3'-bromophenyl)pe!nta-234-dienoylguanidine,
2,3,5,6,-tetramethylcmnamoylguanidine,
3-ethoxycinnamoylguanidine,
N,N'-bis(3ρhenylρropanoyl)-N"-phenylguanidine,
(4-Methoxycirmamoyl)guanidine,
(2-Chlorocinnamoyl)guanidine,
(3-Nitrocinnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
2-(2-napthyl)acetoylguanidioe,
N-(3-phcnylpropanoyl)-Nl-phcnyIguanidine,
5-(2'-bromophenyi)penta-2,4- dienoylguanidine,
(4-Bromocinnamoyl)guanidine,
(2-Nitrocinnamoyl)guanidine,
(3-Chlorocinnamoyl)guanidine, ;
(4-Methoxyciπnamoyl)guanidine,
4-(trifluoromethyl)oinnamoylguanidme,
[(E)-3-(4-Dimethylaminophenyl)-2- methylacryloyl]guanidine, . (
N-Benzoyl-N'-cinnamoylguanidine, j
4-ρhenylbenzoylguanidine, trans-3-Furanacryoylguahidine,
N-amidino-3-aminθ'5-phenyl-6-chloro-2- . |
Pyraziπecarboxamide, i i N- iimamoy -N'phenylguamdine,
Cinnamoylguanidine,
3-methoxy-amiloride,
(3-phenylpropanoyl)guanidine,
3rmetfaoxy -HMA,
Benzyoylguanidine,
N-amidino-3,5-diamino-6-phynyl-2-
Pyrazinecarboxamide,
(Quinoline-2-carbonyl)guanidine,
[3-(3-Pyridyl)acryIoyl]guanidine,
N-Cinnamoyl-N^N-dimethylguamdine,
N- -naptboyty-N'-phenylguanidine and
(Phenylacetyl)guanidine.
18. The method accordrag to claim 16, wherein said compound is selected from the group consisting of
2-t-butylcinnamoylguanidine,
4-isoρropylcinnamoylguaπidine,
3,4-ώchlorocinnamoylguanidine,
3 -(trifluoromethoxy)dnnamoylguanidine,
2,6-dichlorocinnamoylguani<Ene,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
2-cyclohexyIcinnamoylguanidhιe,
5-bromo-2-methoxycinnamoylguanidine,
2-phenylcinnamoylguanidine,
4-t-butylcinnamoylguanidine,
3-phenylcinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride, trans-3-(l-napthyl)acryloylguanidine,
3-(2-napthyl)acryloylguanidine,
2,4-dichlorocinnamolyguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidiue,
4-methylcinnamoylguanidinβ,
(4-CWorocitmamoyl)guanidine,
3-fluorociπnamoylguanidine,
3-(cyclohex-l-cn-l -yl)cinnamoylguanidάnc,
(a-Methylci«namoyl)guanidine,
2,3,5,6,-tetramethylcinnamoylguamdine,
2-fluorocinnamoylguanidine,
(3-Nitrociιmamoyl)guanidme,
2,5-dimethylciππamoylguanidine,
3-t-butylcinnamoylguanidine,
(3-Methoxycinnamoyl)guanidine,
3-methylcinnamoylguaπidine,
3-isopropylcinnamoylguanidine hydrochloride, (2-Bromocinnamoyl)guanidine,
3-ethoxyciιmanιoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
(2-Chlorøcinnamoyl)guanidine,
4-ethoxy nnamoylguanidine,
4-fluorocinnamoylguanidine,
3,4-difluorociαnamoyiguanidine,
N-β-pbeaylpropano -N1-
Phenylguanidine,
2,4,6-trimethylcirmamoylguanidine,
2-methylcinnamoylguanidine,
(trans-2-Phenylcyclopropanecarbpnyl)- guanidine,
(4-Phenoxybenzoyl)guanidine,
(2-MethoxycinnamoyI)guanidines
Cinnamoylguanidine,
3,4-(memylenedioxy)cinnamoylguanidine,
NJ^-Bis(amidino)napthalene-2,6-
Dicarboxa ide,
2,3-dimethyl nnamoylguanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
N,N,-Bis(3-phenylpropanoyl)guanidine,
2.3-difhιorocinnamoylguanidine,
1-naρthoylguanidine,
6-methoxy-2-naphthoylguanidine,
S-(N,N-Dimethyl)amiloride hydrochloride,
2-ethoxycinnamoylguanidine,
2-napthόylguauidine,
3,4,5-trimethoxycinnamoylguanidine,
2-(trifluoro ethyl)cinnamoylguanidine, cinnamoylguanidine hydrochloride,
(4-Hydroxycinnamoyl)guanidine,
5-(4-fluor phenyl)amiloride,
2-(l-napthyl)acetoylguanidine,
(2-Furanacιyloyl)guanidine,
N-Cinnamoyl-N',N'-dimethyIguanidπιe,
2-(2-napthyl)acetoylguanidine and '
N,N-bis(3phenylprapanoyl)-N' -
Phenylguanidine.
19. The method according to claim 12, wherein said Coronavirus is human Coronavirus OC43.
20. The method according to claim 19, wherein said compound is selected from the group consisting of
3-methylcinnamoylguanidine, traπs-3-(l-napthyl)acryloylguanidine,
(3-Bromocinnamoyl)guanidinc,
(2-Chlorocinnamoyl)guanidine,
3,4-dichlorø nnamoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
4-isopropylcinnamoylguanidine,
Cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
2,4-dichlorocinnamolyguanidine,
(4-Chlorocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
(4-Bromocirmamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
(5-Phenyi-penta-2,4-diernoyl)guanidine,
3-(trifluoromethoxy)cinnamoylguanidine and
2-t-butylcinnamoylguanidine.
21. The method according to claim 12, wherein said Coronavims is porcine respiratory Coronavirus (PRCV).
22. The method according to claim 21, wherein said compound is selected from the group consisting of
5-(N,N-hexamethylene)amiloride,
6-methoxy-2-naphthoylguanidine,
Cinnamoylguanidine,
N-(3-phenylpropanoyl)-N'-phenylguanidine,
3-methylcinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
(trans-2-Phenylcycloρropanecarbonyl)guanidine, trans-3-(l-napthyl)acryloylguanidine and
2-(2-naρthyl)acetoylguanid e,
23. The method according to claim 12, wherein said Coronavirus is bovine
Coronvirus (BCV).
24. The method according to claim 23, wherein said compound is selected from the group consisting of
(3-Bromocinnamoyl)guanidine,
3-(trifluoromethyl)cinnamoylguanidtne,
6-methoxy-2-naphthoylguanidine,
5-(N^ϊ-hexametiιylene)amiloride, tians-3-(l-napthyl)acryloylguanidine,
Cinnamoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
2-(2-napthyl)acetoylguanidine,
(traήs-2-Phenylcyclopropanecarbonyl)guanidine,
N-(3-phenylproρanoyl)-N-phenylguanidincand
4-phenylbenzoylguanidine.
25. The method according to claim 12, wherein said Coronavirus is any one of the known corqnavirus isolates listed in Table 1.
26. The method according to claim 25, wherem said compound is selected from the group consisting of
4-isopropylcinnamoylguanidine, 3,4-dichlorocinnamoylguaniditte, 3-(1rifluoromethoxy)cinnamoylguamdine, 4-t-butylcinnamoylguanidine, 3-isopropylcinnamoylguanidine hydrochloride,
27. The method according to claim 6, wherein said virus is the Hepatitis C virus.
28. The method according to claim 27, wherein said compound is selected from the group consisting of
2,3-dimethylcinnamoyiguamdine,
2,4,6-trimethylcinnamoylguanidine,
5:bϊorao-2-fiuorocinnamoylguanidine,
(4-Bromocmnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
4-(triflupromethyl)cinnamoylguanidine,
6-methoxy-2-naphthoylguaιudine,
(2-Chlorocinnamoyl)guanidine,
(4-Chlorocinnamoyl)guanidine,
(2-Bromocinnamoyl)guanidine, 2,6-dichlorocinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
2-(triftuoromethyl) nnamoylguanidine5
(4-Phenoxybenzoyl)guanidine,
3,4-dichlorocinnamoylguanidine,
4-isoρropylcinnamoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
4-t-butylciιmamoylgαanidine,
2-t-butylciιmamoylguamdine,
2-cthylcinnamoylguanidine,
4-methylcinnamoylguanidine,
5-bromo-2-methoxycinnamoyIguanidinc,
3-(trifluoromethoxy)cinnamoylguanidine,
2-cyclohexyldnnamoylguanidine,
1-napthoylguanidine,
3-t-butylciιmamoylguanidine,
4-phenylbenzoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
N-(cinnamoyl)-]Srphenylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
Benzamil hydrochloride,
N-(3-phenylpropanoyl)-N'-phenylguanidine,
N,N'-bis(3phenylpropanoyl)-N1'-ρhenylguaπidine,
3-(2-napthyl)aeryloylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
2'4 DichloroBenzamil HCI,
5-tert-butylamino-amiloride,
5-(N-Ethyl-N-isopropyl)amiloride,
(4-Methoxycinnamoyl)guanidine,
4-fluorocinnamoylguanidine,
(3-MtrocinnamoyI)guanidine,
4-ethoxycinnamoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
3-ethoxycinnamoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
4-ρhenylcinnamoylguanidine, trans-3-Furanacryoylguanidine,
N-(6-Hydroxy-2-napthoyl)-N1-phenylguanidine,
(2rFuranacryloyl)guanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidin<t, cinnamoylguanidine hydrochloride,
5-(N,N-hexamethylene)amUo:ride,
2,3-difluorocinnamoylguanidiπe,
2-(l-napthyl)acetoylguanidine,
(a-Methylcinnamoyl)guanidine,
(2-Nitrocinnamoyl)guanidine, 6-IodoamiIoride,
3,4-(methylenedioxy)cinnamoylguaπidine,
2-ethoxycinnamoylguanidine,
Cinnamoylguanidine,
2-ρhenylcinnamoylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
2-napthoylguanidine,
3-phenylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
5-(4-fluorophenyl)amiloride,
(3-Methoxycinnamoyl)guanidine,
2-fluorocinnamoylguamdine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidinc,
[(4-Chlorophenoxy-acetyl]guanidine,
(3-ρhenyIρroρanoyl)guanidine,
2-chlαro-6-fluorocinnamoylguanidme,
3-fluorocinnamoylguanidine,
2-methylcinnamoylguanidine,
(2-Mefhαxycinnamoyl)gπanidine, l-bromo-2-napthoylgua∞dine,
3,4,5-trimethoxycinnamoylguanidine,
3-inethylcinnamoylguanidine,
3-(trans-hept-l-en-l-yl)cinnamoylguanidine,
Phenamil methanesulfonate salt ,
2,4-dicblorocinnamolyguanidine,
(4-Nitrocinnamoyl)guanidi»e,
3,4-difluorocinnamoylguanidine and
[(E)-3-(4-Dimethylaminophenyl)-2- methylacryloyljgπanidine.
29. The method according to claim 6, wherein said virus is Equine Arteritis virus.
30. The method according to claim 29, wherein said compound is selected from the group consisting of
5-(N,N-hexame1hylene)amiloride, (3-Bromocinnamoyl)guanidine, trans-3-(l-naρthyl)acryloylguanidine, 2-t-butylcinnamoylguanidine and 2-(cyclohex-l-en-lyl)cinnamoylguanidine.
31. A method according to any one of claims 6 to 30, wherein said compound is provided as a pharmaceutical composition accordmg to claim 4 or claim 5.
32. A method for preventing the infection of a cell exposed to a virus comprismg contacting said cell with a compound according to any one of claims 1 to 3,
33. The method according to claim 32, wherein said virus is a Lentivirus.
34. The method according to claim 33, wherein said Lentivirus is Human Immunodeficiency Virus (HIV).
35. The method according to claim 34, wherein said compound is selected from the group consisting of
(3-Chlorocinnamoyl)guamdine,
(3-Bromocinnamoyl)guanidine,
(2-ChIorocinnamσyl)guanidine,
(2-Bromocinnamoyl)guanidine,
3-(trifluoromethyl)cinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
3-methylάnnamoylguanidϊne,
2-mβthylcinnamoylguanidine,
2,3-dimethylcinnamoylguanidine,
Cinnamoylguanidine,
6-methoxy-2-naphfhoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
3,4-dichlorocinnamoylguanidine,
2,6-dicHorodnnamoylguanidine,
4-phenylbenzoylguanidine,
2-ethylcirmamoylguanidiue,
(4-Chlorocinnamoyl)guanidine,
2-napthoylguanidine,
2,5-dimethylcinnamoylguanidine,
3-isopropylcinnamoylgua»idine hydrochloride,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
3-phenylcinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(3 -bromophenyl)penta-2,4-dienoylguanid e,
3-(cycloh «-l-en-l-yl)cinnamoylguanidine,
3-(trifluoromethoxy)ctnnamόylguanidine,
2-(trifluoromethyl)cinnamoyIguamdine,
N,N'-bis(3phenylpropanoyl)-N"-ρhenylguanidiQe,
2-ethoxycinnamoylguanidine,
N-(3-ρhenylρropanoyl)-N,-phenjdguanidine, 437-
4-(trifluoromethyl)cinnamoylguanidine,
(4-Methoxycinnamoyl)guanidine,
2-t-butylcitmamoylguanidine,
4-methylcinnamoylguanidine,
2-fluorocinnamoyIguanidine,
2-phenylciπnamoyIguanidine,
N-(6-Hydroxy-2-napthoyl)-N,-phenylguanidine,
3-t-butylcinnamoylguanidine,
3,4-difluotrocinnamoylguanidine,
5-(N,N-hexamethylene)amiloride,
3-fiuorocinnamoyiguanidine,
5-bromo-2-methoxycinnamoylguanidine,
3-ethoxycinnamoylguanidinc,
3,4-(methylenedioxy)cinnamoylguanidine,
(2-Methosycinnamoyl)guanidine,
2'4 DichloroBenzamil HCI,
2,3,5,6,-tetramethylcinnamoylguanidine,
3-(2-napthyl)acryloylguanidme,
2-(l-napthyl)acetoylguanidine,
2,3-difluorocinnamoylguanidine,
(3-Methoxyciιmainoyl)guanidine,
4-isopropylcinnamoylguanidine,
2,4,6-trimethylcinnamoylguanidine,
N-(cinnamoyl)-N"phenylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
2-(2-napthyl)acetoylguamidine,
(4-Hydroxycinnamoyl)guanidine,
4-phenylcinnamoylguanidine,
4-fluorocήmamoylguanidine,
N^-bis-^innamoyl^-N'-phenylguanidine,
(2-FuranacryloyDguanidine,
Phenamil methanesulfonate salt ,
Benza il hydrochloride,
(3-Nitrocinnaraoyl)guanidine,
Beπzyoylguanidine,
(4-Phenoxybenzoyl)guanidine, -(trans-hept-l-en-l-yl)cirmamoylguanidine,
5-(N-Metbyl-N-isobutyl)amiloride,
2-cyclohexyldnnamoylguanidine,
4-ethoxycinnamoylguanidine,
2,4-dichlorocinnamolyguanidine,
5-(N-Ethyl-N-isopropyl)amiloride,
N-amidino-3-amino-5-hexamethyleneimino-6-phenyl-
2-pyrazinecarboxamide,
(a-Methylcinnamoyl)guanidine, cinnamoylguanidine hydrochloride,
[(4-Chlorophenoxy-acetyl]guanidine,
N-amidino-3-amino-5-phenyl-6-chloro-2- pyrazinecarboxamide, '
5-(4-fluorophenyl)amiloride,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
(2-Nitrocinnamoyl)guanidine, trans-3-Furanacryoylguanidine,
1-napthoylguanidine,
5-tert-butylamino-amiloride,
3-methoxy -HMA,
(3-phenylpropanoyl)guanidine,
4-t-butylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
N,N-Bis(3-phenylpropanoyl)guaπidine,
N-Benzoyl-N'-cinna oylguanidine and
1 -bromo-2-napthoylguanidine.
36. The method according to claim 34, wherein said compound is selected from the group consisting of 4-phenylbenzoylguanidine, (3- bromocinnamoyl)guanidine, 3-(trifluoromethyl)ciπnamoylguanidme, 5- (NJSf-hexamethylene)amiloride, and (5-Phenyl-penta-2,4- dienoyl)guanidine.
37. The method according to any one of claims 34 to 36, wherein said HIV is HIV-1. 1
38. The method according to claim 32 wherem said virus is a Coronavirus.
39. The method according to claim 38, wherein said Coronavirus is the Severe Acute Respiratory Syndrome virus (SARS).
40. The method according to claim 39, wherein said compound is selected from the group consisting of
2,3-difluDrocinnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
4-t-butylcinnamoylguanidine,
3-(2-napthyl)acry ylguanidine,
(3-Chlorocimιaraoyl)guanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
2,5-dimethylcinnamoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
4-isopropylcinnamoylguanidine, (3-Bromo nnamoyl)guanidine,
6-methoxy-2-naphthoylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
3-phenylcinnamoylguanidine,
(2-Chlorocinnamoyl)guanidine,
2'4 DichloroBenzamil HCI,
4-phenylcinnamoylguanidine,
4-(trifϊuoromethyl)cinnamoylguanidiπe,
3-(trifϊuoromemoxy)cinnamoylguarιidine,
3-(trifluoromethyl)cimιamoylguanidine,
2-ethoxycinnamoylguanidine, cinnamoylguanidine hydrochloride,
4-cthόxycinnamoylguanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
5-tert-butylamino-amiloride,
3-t-butylcinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
(4-Oιlorocinnamoyl)guanidine,
2-t-b.utylcinnamoylguanidine,
2-cyclohexylcinnamoylguanidine,
6-Iodoamiloride,
3-(trans-hept-l -en-1 -yl)cinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
(4-Hydroxyciπnamoyl)guanidine,
N-(3-phenyIpropanoyl>N'-phenylguanidine,
(3-Nitrocinnamoyl)guanidine,
3-fiuorocinnamoylguanidine,
2-(l-napthyl)acetoylguanidine,
2-ethylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
2-napthoylguanidine,
5-(4-fluorophenyl)amiloride,
2-(trifluor ιethyl)c namoylguanidine,
N-fe-Hydroxy^-napthoyiyN'-phenylguanidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
N,N'-bis(3phenylpropanoyl)-N"-phenylguanidine,.
1-napthoylguanidine,
Benzamil hydrochloride,
3-methoxy-HMA,
4-methylcinnamoylguanidinc,
4-fluorocitmamoylguanidine,
3,4-(methylenedioxy)cinnamoylguanidinc,
5-(N,N-hexamethylene)amiloride,
N-(cinnamoyl)-N'phemylguanidine>
5-(N-Ethyl-N-isopropyl)amiloride,
3-methylcinnamoylguanidine, 2-methylcinnamoylguanidine,
2,3,5,6,-tetramethylcinnamoyiguanidine, trans-3-Furanacιyoylguanidine,
(4-Methoxycinnamoyl)guanidine,
(2-Furanacryloyl)gua dine,
(3-phenylpropanoyl)guanidine,
2-(2-napthyl)acetoylguanidine,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
[3-(3-Pyridyl)acryloyl]guanidine,
4-phenylbenzoylguanidine,
2,4-dic orociιmamolyguaιιidine,
(3-Methoxycinnamoyl)guamdinc,
2-fluorocinnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
(a-Methylcinnamoyl)guanidine,
5-(3'-bramophenyl)penta-2,4-dienoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidme,
(Quinoline-2-carbonyl)guanidine,
(Phenylacetyl)guanidine,
N,N'-Bis(amidino)napthalene-2,6-dicarboxamide,
6-bromo-2-napthoylguanidinej,
1 -bromo-2-napthoylguanidine,
2-chloro-6-fluorocinnamoylguanidine,
[(4-Chlorophenoxy-acetyl]guanidine,
Phenamil methanesulfonate sal ,
N-Benzoyl-N'-cinnamoylguaniditie and
N-(2-napthoyi)-N'-phenylguanidine.
41. The method according to claim 39, wherein said compound is selected from the group consisting of cinnamoylguanidine, trans-3-(l- napthyl)acryloylguanidine, and 6-methoxy-2-naphthoylguanidine.
42. The method according to claim 38, wherein said Coronavirus is human Coronavirus 229E
43. The method according to claim 42, wherein said compound is selected from the group consisting of
4-isoproρylcinnamoylguanidine, 3,4-dichlorocinnamoylguanidine, 3-(trifluoromethoxy)ciιmamoylguani<ϋne,
4-t-butylcinnamoylguanidine,
3-isopropylcinnamoyIguanidine hydrochloride,
3-t-butyl nnamoylguanidine,
2-t-butylcinnamoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
5-bromo-2-methoxycitmamoylguanidine,
2,3-difluorocinnamoylguani ine,
3-(2-napthyl)acryloylguanidine,
2-phenylcinnamoylguanidine,
3-phenyϊcinnamoylguanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
4-phenylbenzoylguanidine,
3-(trifϊuoro ethyl)cinnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
4-(trifluorome yl)ciimamoylguanidine,
2-(cyclohex-l -en- lyl)cinnamoylguaήidine,
(4-Bromociιmamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
1-naρthoylguanidine,
5-(4-fluorophβuyl)amiloride,
(5-Phenyl-panta-2,4-dienoyl)guanidine,
(3-Bromocinna oyl)guanidine,
2,5-dimethylcinnamoylguanidine,
2-(trifluoromethyl)cinnamoylgιιanidine,
6-mefhoxy-2-naphthoylguanidine,
(4-Chlorocinnamoyl)guanidine,
(3-Methoxycinnamoyl)guanidine,
5-bromo-2-fluorocinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
Cinnamoylguanidine,
(2- ethoxycinnamoyl)guanidine,
(a-Methylcinπamoyl)guanidine,
4-phenylcinnamoylguanidine, .
2,6-dichlorocinnamoylguanidine,
(2-Bromocinnamoyl)guanidine,
2,4,6-trimethylcinnamoylguamdme,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
2-(l-naρthyl)acetoylguanidine,
2-ethylcinnamoylguanidine,
2-cyclohexylcinnamoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
2-ethoxycinnamoylguanidine,
3-methylcinnamoylguanidine,
2-mefhylcinnamoylguanidine, 3-fluorocinnamoylguanidine, cinnamoylguanidine hydrochloride,
2,3-dimethylcinnamoylguanidine,
2-fluorocinnamoylguanidine,
4-fluorocinnamoylguanidine,
3,4-difluαrocinnamoylguanidine,
5-tert-butylamiπo-amiloride,
2-napthoylguanidine,
N,N'-Bis(amidino)napthalene-2,6-dicarboxamide,
N,lSP-Bis(3-phenylpropanoyl)guanidine,
4-methylcinnamoylguanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
3-ethoxycinnamoylguaπidine,
NJSI,-bis(3phenylpropanoyl)-N,,-phenylguaπidine,
(4-Methoxycinnamoyl)guanidine,
(2-Chlorocinnamoyl)guanidine,
(3-Nitrocimιamoyl)guanidine,
4-ethoxycinnamoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
2-(2-naρthyl)acetoylguanidine,
N-(3-phenylpropauoyl)-N,-phenylguanidine,
5-(2-bromophenyl)ρenta-2,4- dicnoylguanidine,
(4-Bromocinnamoyl)guanidine,
(2-Nitrocinnamoyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
(4-Methoxycinnamoyl)guanidine,
4-(trifluoromethyl)ciιmamoylguanidine,
[(E)-3-(4-Dimcthylaminophenyl)-2- methylacryloyl]guanidine,
N-Benzoyl-NT-cinnamoylguanidine,
4-ρhenylbenzoylguanidine, trans-3-Furanacryoylguanidine,
N-amidino-3-amino-5-phenyl-6-chloro-2-
Pyrazinecarboxamide,
N-(cinnamoyl)-Nlρhenylguanidine,
Cinnamoylguanidine,
3-methoxy-amiloride,
(3-phenylpropanoyl)guamdme,
3-methoxy-HMA,
Benzyoylguanidjne,
N-amidino-3,5-diamino-6-phynyl-2-
Pyrazinecarboxamide,
(Quinoline-2-carbc∞yl)guanidine,
[3-(3-Pyridyl)acryloyI]guanidine, N-Cinnamoyl-N^lSP-dimethylguanidine, N-(2-napthoyl N'-pheπylguanidine and (Phenylacetyl)guanidine.
44. The method according to claim 42, wherein said compound is selected from the group consisting of
2-t-butylcinnamoylguanidine,
4-isopropylcinnamoylguanidine,
3,4-<fichlorocinnamoylguanidine,
3-(trifluoromethoxy)cinnamoyiguanidine,
2,6-dichlorocinnamoylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
2-cyclohexylcinnamoylguanidine,
5-bro o-2-methoxycinnamoylguanidine,
2-phenylcinnamoylguanidine,
4-t-butylcinnamoylguanidine,
3-phenylciπnamoylguanidine,
(3-Bxomocirmamoyl)guanidine,
5-(NjN-hexamethylene)amiloride, ttans-3-(l-napthyl)acryloylguanidine,
3-(2-napthyl)acryloylguanidine,
2,4-dichlorocinnamolyguanidine,
3-(trifluoromethyl)cinnamoylguaπidine,
5-bromo-2-fiuorocinnamoylguanidine,
4-methylcinnamόylguanidine,
(4-Chlorocinnamoyl)guanidine,
3-fluorooinnamoylguanidine,
3-(cyclohex-l-en-l-yl)cinnamoyIguanidine,
(a-Methylcinnamoyl)guanidine,
2,3,5,6,-tetramethylcmnamoylguanidine,
2-fluorocinnamoylguanidine,
(3-Nitrocinnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
3-t-butylcinnamoylguanidine,
(3-Methoxycinnamoyl)guanidine,
3-methylcinnamoylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
(2-Bromocinnamoyl)guaaidine,
3-ethoxycinnamoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
(2-Chlorocinnamoyl)guanidine,
4-ethoxycπmamoylguanidine,
4-fluorociraιamoyJguanidine,
3,4-difIuorocinnamoyIguanidine,
N-(3-phenyIpropanoyl)-N'-
Phenylguanidine,
2,4,6-trimethiylcinnamoylguanidine, 2-methylcinnamoylguanidine,
(trans-2-Phenylcyclopropanecarbρnyl)- guanidine,
(4-Phenoxybenzoyl)guanidine,
(2-Methoxycinnamoyl)guanidine,
Cinnamoylguanidine,
3,4-(methylenedioxy)cinnamoylguanidme,
N,N,-Bis(amidino)napthalene-2,6-
Dicarboxamide,
2,3-dimethylcinnamoylguanidine,
5-(3Vbromophenyl)ρenta-2,4-dicnoylguanidine,
N,N'-Bis(3-phenylpropanoyl)guanidine,
2,3-difiuorocinnamoylguanidine,
1-naρthoylguanidine,
6-methoxy-2-naphfhoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
2-ethoxycinnamoylguanidine,
2-naρthoylguanidine,
3,4,5-trimethoxyciπnamoylguanidine,
2-(trifluoromethyl)cmnamoylguanidine, cinnamoylguanidine hydrochloride,
(4-Hydroxycinnamoyl)guanidine,
5-(4-fluorophenyl)amilαride,
2-(l-napthyl)acetoylguamdine,
(2-Furanacτyloyl)guanidine,
N-Cinnamoyl-N'jN-dimethylguanidine,
2-(2-napthyl)acetoylguanidine and
NJ\T-bis(3phenylpropanoyl)-N"-
Phenylguanidine.
45. The method according to claim 38, wherein said Coronavirus is human Coronavirus OC43.
46. The method according to claim 45, wherein said compound is selected from the group consisting of
3-methylcinnamoylguanidine, trans-3-(l-naρthyl)acryloylguanidiae,
(3 -Bromociunamoyi)guanidine,
(2-Chlorooinnamoyl)guanidine,
3,4-dichlόrocinnamoylguanidine,
3-(trifluoromethyl)ctnnamoylguanidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine, ■
4-isopropylcύmamoylguaιιidine,
Cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine, 2,4-dichlorocinnamolyguanidine,
(4-Chlorocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
(4-Bromocinnamoyl)guanidine,
2,6-dichlθrθoinnamoyIguanicline,
5-bromo-2-methoxycinnamoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
3-(trifiuoro ethoxy)cinnamoylguanidine and
2-t-butylcinnamoylguanidine.
47. The method according to claim 38, wherein said Coronavirus is porcine respiratory Coronavirus (PRCV).
48. The method according to claim 47, wherein said compound is selected from the group consisting of
5-(N,N-hexamethylene)amiloride,
6-methoxy-2-naphthoylguanidine,
Cinnamoylguanidine,
N-(3-phenylpropanoyl)-N'-phenylguanidine,
3-meώ.ylcinnamoylguanidine,
(3-Bromocinnamoyl)guanic(ine,
(trans-2-Phenylcyclopropaπecarbonyl)guanidine, trans-3-(l-na thyI)acryloylguanidine and
2-(2-napthyl)acetoylguanidine.
49. The method according to claim 38, wherein said Coronavirus is bovine Cαronvirus (BCV)-
50. The method according to claim 49, wherein said compound is selected from the group consisting of
(3-Bromocinnamoyl)guanidine,
3-(trifluoromethyl)cinnamoylguanidine,
6-methoxy-2-naρhthoylguanidine,
5-(N,N-hexamettιylene)amiloride, trans-3-(l-napthyl)acryloyIguanidine,
Cinnamoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
2-(2-napthyl)acetoylguanidine, /
(trans-2-Phenylcyclopropanecarbonyl)guaιιidine,
N-(3-phenylprcφanoyl)- -ρhflmylguanidine and 446-
4-ρhenyIbenzoylguanidine.
51. The method according to claim 38, wherein said Coronavirus is any one of the known Coronavirus isolates listed in Table 1,
52. The method according to claim 51, wherein said compound is selected from the group consisting of
4-isoρropylcinna oylguanidine, 3,4-dichlorooiπnamoylguaπidine, 3-(trifluoromethoxy)cinnamoylguanidine, 4-t-butylcinnamoylguanidine, 3-isopropylcinπamoylguanidine hydrochloride,
53. The method according to claim 32, wherein said virus is the Hepatitis C virus.
54. The method according to claim 53, wherein said compound is selected from the group consisting of
2,3-dimethylcinnamoylguanidine,
2A6-trimethylcinnamoylguanidine,
5-bromo-2-fluαrocinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
3-(trMuoromethyl)cinnamoylguanidine, (trifluoromethyl)cinnamoylguanidine,
6-methoxy-2-naρhthoylguanidine, <
(2-Chlorocinnamoyl)guaπidine,
(4-Chlorocinnamoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
(3-Bromocinnamoyl)gua»idine,
(3-Chlorocinnamoyl)guanidine,
2-(trifluoromethyl)cinnamoyIguanidine,
(4-Phenoxybenzoyl)guanidine,
3,4-dichlorocinnamoylguanidine,
4-isopropylcinnamoylguanidine, trans-3-(l -napthyl)acryloylguanidine,
4-t-butylciπnamoylguanidine,
2-t-butylcinnamoylguanidine,
2-ethylcinnamoylguaπidine,
4-methylcmnamoylguanidine,
5-bromo-2-niethoxycinnamθylguanidine, 3-(trifluoromethoxy)cinnamoylguanidine,
2-cyclohexylcinnamoylguanidine,
1 -napthoylguanidine,
3-t-butylcinnamoylguanidine,
4-phenylbenzoylguamdine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
N-(cinnamoyl)-Nlphenylguanidme,
3-isopropylcinnamoylguanidine hydrochloride,
Benzamil hydrochloride,
N-(3-phenylpropanoyl)-N'-ρhenylgiιaπidine,
N>N,-bis(3ρhenylpropanoyl)-N"-phenylguanidine,
3-(2-napthyl)acryloylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
2'4 DichloroBenzamil HCl,
5-tert-butylammo-amiloride,
5-(N-Ethyl-N-isopropyl)amiloride,
(4-Methoxycinnamoyl)guanidine,
4-fluorocinnamoylguanidine,
(3-Nitrocinnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
(tians-2-Phenylcyclopropanecarbonyl)guanidine,
3-ethoxycinnamoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
4-phenylcinnamoylguamdine, trans-3-Furanacryoylguanidine,
N-(6-Hydroxy-2-napthoyl)-N'-ρhenylguanidine, .
(2-Furanacryloyl)guanidine,
3-(cyclohex-l -en- 1 -yI)cinnamoylguanidine, cinnamoylguanidine hydrochloride,
5-(N,N-hexamethylene)amiloride,
2,3-difluorocinnamoylguanidine,
2-(l -napthyl)acetoylguanidine,
(a-Mefoylcinnamoyl)guanidine,
(2-Nifrocinnamoyl)guanidine,
6-Iodoamiloride,
3,4-(methylene<ϊoxy)cinnamoylguanidine,
2-eth xycinnamoylguanidine,
Cinnamoylguanidine,
2-ρhenylcinnamoylguanidine,
2-(cyclohex-lren-lyl)cinnamoylguanidine,
2-napthoylguanidine,
3-phenylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
5-(4-fluorophenyl)anuloride,
(3-Methoxycinnamoyl)guanidine,
2-fluorocinnamoylguanidme,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine, [(4-Chlorophenoxy-acetyl]guanidine,
(3-ρhenylpropanoyl)guanidine,
2-chloro-6-fluorocinnamoylguanidine,
3-fluorocinnamόylguanidine,
2-methylciιmamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
1 -bromo-2-napthoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
3-methyl πnamoylguanidine,
3-(trans-hept-l-en-l-yl)cinnamoylguanidine,
Phenamil methanesυlfαnate salt ,
2,4-dichlorocinnamolyguanidine,
(4-Nitrociπnamoyl)guamdine,
3,4-difluorocinnamoylguanidine and
[(15)-3-(4-Dimethylaminophenyl)-2- methylacryloyl]guanidine.
55, The method according to claim 32, wherein said virus is the Equine Arteritis virus.
56. The method according to claim 55, wherein said compound is seleoted from the group consisting of
5-(N,N-hexamethylene)amiloride, (3-Bro ocinnamoyl)guanidine, trans-3-(l-napthyl)acryloylguanidine, 2-t-butylcinnamoylguanidine and 2-(cyclohex-l-en-lyl)cinnamoyiguanidine.
57. . The method according to any one of claims 32 to 56, wherein said compound is provided as a pharmaceutical composition according to claim 4 or claim 5.
58. A method for the therapeutic or prophylactic treatment of a subject infected with or exposed to a virus, comprising the administration of a compound accordmg to any one of claims 1 to 3 to a subject in need of said treatment.
59. The method accordmg to claim 58, wherein said virus is a Lentivirus.
60. The method according to claim 59, wherein said Lentivirus is Human . Immunodeficiency Virus (HIV).
61. The method according to claim 60, wherein said compound is selected from the group consisting of
(3-Chlorocinnamoyl)guanidine,
(3-Bromocinnamoyl)guaπidine,
(2-Chlorocinnamoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
3-(trifluoromethyl)cinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
3-methylcinnamoylguaπi hπe,
2-methylcinnamoylguanidine,
2,3-dimethylciιmamoylguanidine,
Cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine, trans-3-(l -napthyl)acιyloylguanidine,
3,4-dichlorocinnamoylguam'dine,
2,6-dichlorocinnamoylguanidine,
4-phenylbenzoylguanidine,
2-ethylcinnamoylguanidine,
(4-Cώorocinnamoyl)guanidine,
2-napthoylguanidine,
2,5-dimethyicinnamoylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
3-phenylcinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(3'-bromopheinyi)penta-2,4-dienoylguanidine,
3-(cyclohex-l-en-l-yl)eύuιamoylguanidine,
3-(trifluoromethoxy)cinnaraoylguanidine,
2-(trifluoromethyl)dnnamoylguaπidine,
N^J'-bis(3phenjflpropanoyl)-N"-phenylguanidine,
2-ethoxycinnamoylguanidine,
N-(3-phenylprϋpanoyl)-N'rphenylguanidine,
4-(trifluorome<hyl)cinnamoylguanidine,
(4-Methoxycinnamoyl)guanidine,
2-t-butylcinnamoylguanidine,
4-methylcinnamoylguanidine,
2-fluorodnnamoylguanidine,
2-phenylciιmamoylguanidine,
N-(6-Hydroxy-2-napthoyl)-N'-phenylguanidine,
3-t-butylciιmamoylguanidine,
3,4-d uorocinnamoylguanidine,
5-(N,N-hexamethyIene)amiloride,
3-fluorocinnamoylguanidine, 5-bromo-2-methoxycinnamoylguanidine,
3-ethoxycinnamoylguanidine,
3,4-(methylenedioxy)cinnamoylguanidine,
(2-Methoxycinnamoyl)guaπidine,
2'4 DichloroBenzamil HCI,
2,3,5,6,-tetramethylcinnamoylguanidine,
3-(2-napthyl)acιyloylguanidine,
2-(l-naρthyl)acetoylguanidine,
2,3-difluorocinnamoylguanidine,
(3-Methoxycinnamoyl)guanidine, ,
4-isopropylcinnamoylguanidine,
2,4,6-t methylcinnamoylguanidme,
N-(cinnamoyl)-N'phettylguanidine,
2-(cyciohex-l-en-lyl)cinnamoylguanidine,
2-(2-napthyl)acetoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
4-phenylcinuamoylguanidine,
4-fluorocinnamoylguanidine,
NJSP-bis-(cinnamoyl)-N"-phenylguanidine,
(2-Furanac yloyl)guamdine, '
Phenamil meth nesulfonate salt ,
Beπzamil hydrochloride,
(3-Nitrocinnamoyl)guanidine,
Benzyoylguanidhie,
(4-PhenDxybenzoyl)guanidiπe,
3-(trans-hepM -en-1 -yl)cinnamoylguanidine,
5-(N-MethyI-N-isobutyl)amiIoride,
2-cyclohexylcinnamoylguanidine,
4-ethoxycinnamoylguaπidine,
2,4-dicMorocinnamolyguanidine,
5-(N-Efhyl-N-isoρropyl)amiloride,
N-amidino-3-amino-5-hexamethyleneimino-6-phenyl-
2-pyrazinecarboxamide,
(a-Methyldnnamoyl)guanidine, cinnamoylguanidine hydrochloride,
[(4-Chlorophenoxy-acetyl]guanidine,
N-amidino-3-amino-5-phenyl-6-chloro-2- pyrazinecarboxamide,
5-(4-fluorophenyl)amiloride,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
(2-Nitrocinnamoyl)guaπidine, trans-3-Furanacryoylguanidinc,
1-napthoylguanidine,
5-tert-butylamino-amiloride,
3-methoxy — HMA,
(3-ρhenylpropanoyl)guanidine,
4-t-butylcinnamoylguanidine,
5-(N,N-Dime l)arailoride hydrochloride, NJSP-Bis(3'phenylpropanoyl)guanidine> N-Benzoyl-N'-cinnamoylguanidine and 1 -bromo-2-napthoylguanidine.
62. The methods according to claim 60, wherein said compound is selected from the group consisting of 4-ρhenylbenzoylguanidine, (3- bromocinnamoyl)guanidine, 3-(trifluoromethyl)cinnamoylguanidine, 5- (N,N-hexamethylene)amilαride, and (5-Fhenyl-ρenta-2,4- dienoyl)guanidine.
63. The method according to any one of claims 60 to 62, wherein said HIV is HIV-1.
64. The method according to claim 58 wherein said virus is a Coronavirus.
65. The method according to claim 64, wherein said Coronavirus is the Severe Acute Respiratory Syndrome virus (SARS).
66. The method according to claim 65, wherein said compound is selected from the group consisting of
2,3-difluorocinnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
4-t-butylcinnamoylguanidine,
3-(2-napthyl)acryloylguanidine,
(3-Chlorociιmamoyl)guanidine,
3-(cyclQhex-l-en-l-yl)cinnamoylguanidine,
2,5-dimethylcinnamoylguanidine, trans-3-(l-naρthyl)acryloylguanidine,
4-isopropylcinnamoyIguanidine,
(3-Bromocdnπamoyl)guaniditιe,
6-mcthoxy-2-naphthoylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
3-ρhenylcinnamoylguanidine,
(2-Chlorocinnamoyl)guanidine,
2'4 DichloroBenzamil HCI,
4-phenylcinnamoylguanidine,
4-(trifluόromethyl)cinnamoylguanidine,
3-(trifluoromethoxy)cinnamoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
2-ethoxycinnamoylguanidine, cinnamoylguanidine hydrochloride,
4-ethoxycinnamoylguanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
5-tert-butylamino-amiloride,
3-t-butylcinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
(4-Chlorocinnamoyl)guaιιidine,
2-t-butylcinnamoylguanidine,
2-cyclohexylcinnamoylguanidine,
6-Iodoamiloride,
Figure imgf000153_0001
(3-phenylpropanoyl)guanidine,
2-(2-napthyl)acetoylguanidine,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
[3-(3-Pyridyl)acryloyl]guanidine,
4-phenylbenzoylguamdine, 453-
2,4-dichlorocinnamolyguanidine,
(3-Methoxycinnamoyl)guanidine,
2-fluorocinnaraoylguanidine,
(4-Phenoxybenzoyl)guanidine,
(a-Methy innamoyl)guanidine,
5-(3'-bromophenyl)peιιta-2, -dienoylguamdine,
(5-Phenyl-ρenta-2,4-dienoyl)guamdine,
(Quinoline-2-carbonyl)guanidine,
(Phenylacetyl)guanidine,
NJ,T'-Bis(amidino)napthalene-2,6-dicarboxamide,
6-bromo-2-napthoylguanidine, l-bromo-2-napthoylguanidine,
2-chloro-6-fluorocinnamoylguanidine,
[(4-Chloroρhenoxyracetyl]guanidine,
Phenamil methanesulfonate salt ,
N-Benzoyi-N'-cinnamoylguanidine and
N-(2-napthoyl)-N'-phenylguanidine.
67. The method according to claim 65, wherein said compound is selected from the group consisting of cinnamoylguanidine, trans-3-(l- naρthyl)acryloylguanidine, and 6-methoxy-2-naphthoylguanidine,
68. The method according to claim 64, wherein said Coronavirus is human Coronavirus 229E.
69. The method according to claim 68, wherein said compound is selected from the group consisting of
4-isoρropylcinnamoylguanidine,
3,4-dichlorocinnamoylguanidinei
3-(trifluoromethoxy)cinnamoylguanidine,
4-t-butylcinnamoylguanidine,
3-isopropylcinnamθylguanidine hydrochloride,
3-t-butylcinnamoylguanidine,
2-t-butylcinnamoylguanidine, trans-3-(l -napthyl)acryloylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
2,3-difiuorocinnamoylguanidine,
3-(2-naρthyl)acryloylguanidine,
2-phenylcinnamoylguanidine,
3-phenylcinnamoylguanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
4-ρhenylbenzoylguaπidine,
3-(trifluoromelhyl)cinnamoylguaπidine, 454-
(4-Phenoxybenzoyl)guanidine,
4-(trifluoromethyl)cinnamoyIguanidine,
2-(cyclohex- 1 -en- lyl)cinnamoylguaπidine,
(4-Bromocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
1-napfhoylgπanidine,
5-(4-fluoroρhenyl)amiloride,
(5-Phenyl-ρenta-2,4-dienoyl)guanidine,
(3-Bromocinnamoyl)guanidine,
2,5-dimethylciπnamoylguanidine,
2-(trifluoromethyl)ciιmamoylguanidine,
6-methoxy-2-naphthoylguaπidine,
(4-Chlorocinnamoyl)gu3nidine,
(3-Methoxydnπamoyl)guanidine,
5-bromo-2-fluorocmuamoylguamdine,
5-(N,N-Dimethyl)amiloride hydrochloride,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guaπidine,
(a-Methylcinnamoyl)guanidine,
4-phenylcinnamoylguanidine,
2,6-dichlorocinnamoylguanidine,
(2-Bromocinnamoyl)guanidine,
2,4,6-trimethylcinnamoylguaπidine,
(1ιans-2-Phenylcyclopropanecarbonyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
2-(l -nϊφthyl)acetoylguanidine,
2-ethylcinnamoylguaπidine,
2-cyclohexylcinnamoylguanidine,
(4-Hydroxycinnamoyl)guanidme,
2-ethoxycinnamoylguanidine, '
3-methylcinnamoylguanidine,
2-mefoylcinnamoylguamdine,
3-fiuorocinnamoylguanidine, cinnamoylguanidine hydrochloride,
2,3-dimethylciπnamoylguaπidine,
2-fiuorocinnamoylguanidine,
4-fluorocinnamoylguanidine,
3,4-difluorocinnamoylguanidine,
5-tert-butylamino-amiloride,
2-napthoylguanidine,
NJ»T-Bis(amidino)naρthalene-2,6-dicarboxamide,
N^'-Bis(3-phenylpropanoyl)guanidine,
4-methylcinnamoylguanidine,
5-(3'-bromophenyl)ρenta-2,4-dienoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
3-ethoxycinnamoylguanidine, ' N,N'-bis(3ρhenylρroρanoyl)-N,,-phenylguanidine, (4-Methoxycitmamoyl)guanidine, (2-Chlorocinπamoyl)guanidine, (3-Nitrocinnamoyl)guanidine, 4-ethoxycinnamoylguanidine, 3,4,5-trimethoxycinnamoylguanidine, 2-(2-napthyl)acetoylguanidine, N-(3-phenylpropanoyl)-lSr-phenylguaιιidine,
5-(2-bromophenyl)penta-2,4- dienoylguanidine,
(4-Bromocinnamoyl)guanid e,
(2-Nitrocinnamoyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
(4-Meihoxycinnamoyl)guamdine,
4^(trifluorome1hyl)cirmamoylguanidine,
[(E)-3-(4-Dimethylaminophenyl)-2- methylacryloyljguanidiπe,
N-Benzoyl-N'-cinnamoylguanidine,
4-phenylbenzoylguanidine, ttans-3-Furanacryoylguanidine,
N-a idino-3-amiήo-5-phenyl-6-chloro-2-
Pyrasanecaiboxamide,
N-(cinnamθyl)-N,pheπylguanidine,
Cinnamoylguanidine,
3-methoxy-amiloride,
(3-ρhenylprqρanoyl)guanidine,
3-methoxy-HMA,
Benzyoylguanidine,
N-amidino-3,5-diamino-6-phynyl-2-
Pyrazinecarboxamide,
(Quinoline-2-carbonyl)guanidine,
[3-(3-Pyridyl)acryloyl]guanidine,
N-Cinnamoyl-N,,N'-dimethylguanidine,
N-(2-napthoyl)-N -phenylguanidine and
(Phenylacetyl)guanidine.
70. The method according to claim 68, wherein said compound is selected from the group consisting of
2-t-butylcinnamoylguanidine,
4-isopropylcinnamoylguanidine,
3 ,4-dichlorocinnamoyiguaπidine,
3-(trifluoromethoxy)cinnamoylguanidine,
2,6-dichloro nnamoylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
2-cyclohexylcinuamoylguanidine, 5-bromo-2-methoxycinnamoylguanidine,
2-phenylcinnamoylguanidinβ,
4-t-butylcinnamoylguanidine,
3-phenylcύmamoylguaπidine,
(3-Bromocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride, trans-3-(l -napthyl)acryloylguanidine,
3-(2-naρthyl)acryloylguanidine,
2,4-dichlorociπnamolyguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidinc,
4-methylcinnamoylguanidine,
(4-Chlorocinnamoyl)guanidine,
3-fluorocinnamoylguanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
(a-Methylc namoyl)guamdine,
2,3,5,6,-tetramethylcmnamoylguanidine,
2-fluorocinπamoylguanidine,
(3-Nitrocinnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
3-t-butylcinnamoylguanidine,
(3-Methoxycinna oyl)guamdine,
3-methylcinnamoylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
(2-Bromocinnamoyl)guamdinfi,
3-ethoxycirraamoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
(2-Chlorocinn8moyl)guanidine,
4-efhoxy nπamoylguanidine,
4-fluorocinnamoylguanidine,
3,4-difluorocinnamoylguanidine,
N-(3-ρhenyϊρrσpanoyl)-N'-
Phenylguanidine,
2,4,6-trimethylcinnamoylguanidine,
2-methylcinnamoylguanidine,
(trans-2-Phenylcyclopropanecarbonyl)- guanidine,
(4-Phenoxybenzoyl)guanidine,
(2-Methoxyoinnamoyl)guanidine,
Cinnamoylguanidine,
3,4-(methylenedioxy)cinnamoylguanidine,
N,NVBis(amidino)nap1halene-2,6-
Dicarboxatnide,
2,3-dimethylciπnamoylguanidine,
5-(3'-bromophenyl)penta-2,4-dienoyϊguanidine,
N,NVBis(3-ph∞ylpropanoyl)guanidine,
2,3-difluorocinnamoylguanidine,
1 -napthoylguanidine, 6-methoxy-2-naphthoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
2-ethoxycinnamoylguanidine,
2-napthoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
2-(trifluoromethyl)cinna oylguanidine, cinnamoylguanidine hydrochloride,
(4-Hydroxycinnamoyl)guaπidhιe,
5-(4-fiuorophenyl)amiloride,
2-(l -napthyl)acetoylguanidine,
(2-Furaπacryloyl)guanidine,
N-Ciιmamoyl-Nl,N'-dimethylguanidine,
2-(2-napthyl)acetoylguaπidine and
N, -bis(3phenylpropanoyl)-NH-
Phenylguanidine.
71. The method according to claim 64, wherein said Coronavirus is human Coronavirus OC43,
72. The method according to claim 71, wherein said compound is selected from the group consisting of
3-methylάιmaraoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
(3-Bromocinnamoyi)guanidine,
(2-Chlorocinnamoyl)guanidine,
3,4-dichlorocinnamoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
(tmns-2-Phenylcyolopropanecarbonyl)guanidine,
4-isofffopylcinnamoylguanidine,
Cinnamoylguanidine,
6-mefhoxy-2-naphthoylguanidine,
2,4-dichlorocianamolyguanidine,
(4-Chlorocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
(4-Bromocinnamoyl)guanidine,
2,6-dichlorooinnamoylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guaπidine,
3-(trifluoromethoxy)cinnamoylguanidine and
2-t-butylcinnamoylguanidine.
73, The method according to claim 64, wherein said Coronavirus is porcine respiratory Coronavirus (PRCV).
74. The method according to claim 73, wherein said compound is selected from the group consisting of
5-(N,N-hexamethylene)amiloride,
6-methoxy-2-naρhthoylguanidine,
Cinnamoylguamdine,
N-(3-phenylpropanoyl)-N,-phenylguanidine,
3-methylcinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
(trans-2-Phenylcyclopropanecarbonyl)guaήidine, teans-3-(l-napthyl)acryloylguanidine and
2-(2-napthyl)acetoylguanidine.
75. The method according to claim 64, wherein said Coronavirus is bovine Coronvirus (BCV).
76. The method accordmg to claim 75, wherein said compound is selected from the group consisting of
(3-Bromocinnamoyl)guanidine,
3-(trifluoromethyl)cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
5-(N*N-hexametiιylene)amilόride, trans-3-(l-n^)thyl)acryloylguanidine,
Cinnamoylguamdine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
2-(2-napthyl)acetoylguanidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
N-(3-phenylpropanoyl)-N'-phenylguaιήdine and
4-phenylbenzoylguanidine. '
77. The method according to claim 64, wherein said Coronavirus is any one of the known Coronavirus isolates listed in Table 1.
78. The method according to claim 77, wherein said compound is selected from the group consisting of
4-isopropylcinnamoylguanidine, 3,4-dicMorociιmamoylguamdine, 3-(trifluoromethoxy)cinnamoylguanidine, 4-t-butylcinnamoylguanidine, 3-isopropylcinnamoylguanidine hydrochloride, 459-
79. The method according to claim 58, wherein said virus is the Hepatitis C virus,
80. The method according to claim 79, wherein said compound is selected from the group consisting of
2,3-dimethylcronamoylguanidine,
2,4,6-trimethylcinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
3 -(trifluoromethyl)cinnamoylguanidine,
4-(trifluoromethyl)cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
(2-Chlorocinnamoyl)guanidine,
(4-Chlorocinnamoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
(3-Bromociιmamoyl)guanidine,
(3-Chlorocinήamoyl)guaπidine,
2-(trifiuoromethyl)cinnamoylguaπidine,
(4-Phenoxybenzoyl)guanidine,
3,4-dicWorocinnamoylguanidine,
4-isopropylcinnamoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
4-t-butylcinnamoylguanidine,
2-t-butylcinnamoylguanidine,
2.-ethylcinnamoyϊguanidiue, -methylcinnamoylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
3-(trifluoromethoxy)ciππamoylguanidine,
2-cyclohexylcinnamoylguanidine,
1-naρthoylguanidine,
3-t-butylciπnamoylguanidine,
4-pheQylbenzoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
N-(cinnamoyl)-N'phenylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
Benzamil hydrochloride,
N-(3-phenylpropanoyl)-N'-ρhenylguanidine,
N*N1-bis(3phenylpropanoyl)-N"-phenylguanidine,
3-(2-naρthyI)acryloylguanidine,
5-(N-Methyl-N-isobutyl)amiIoride,
2'4 DichloroBenzamil HCI,
5-tert-butylamino-amiloride,
5-(N-Ethyl-N-isopropyl)amiloride,
(4-Methoxycinnamoyl)guanidine, 460-
4-fluorocinnamoylguanidine,
(3-Nitrocinnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
(trans-2-Pheπylcycloproρanecarbonyl)guanidine,
3-ethoxycinnamoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
4-phenylcinnamoylguanidine, trans-3-Furanacryoylguanidine,
N-fe-Hydroxy^-napthoy^lSP-phenylguanidine,.
(2-Furanacryloyl)guanidine,
3-(cyclohex-l-en.-l-yl)cinnamoylguanidine, cinnamoylguanidine hydrochloride,
5-(NJvf-hexamethylene)amiloride,
2,3-difluorocinnamoylguanidine,
2-(l-napthyl)acetoylguanidine,
(a-Methylcinnamoyl)guanidine,
(2-Nitrocinnamoyl)guanidine,
6-Iodoamiloride,
3,4-(methylenedioxy)cinnamoylguaniditte,
2-ethoxycinnamoylguaπidine,
Cinnamoylguanidine,
2-ρhenylcinnamoylguanidine,
2-(cyclohex-l-en-lyI)cinnamoylguanidine,
2-napthoylguanidine,
3-phenylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
5-(4-fluorophenyl)amiloride,
(3-Methoxycinnamoyl)guanidine,
2-fluorocinnamoylguanidine,
5-(3'-bromophenyl)penta-294-dienoylguanidine,
[(4-Chlorophenoxy-acetyl]guanidine,
(3-phenylρroρanoyl)guanidine,
2-chloro-6-fluorocinnamoylguanidine,
3-fluorodnnamoyiguaιudine,
2-methylcinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
1 -bro o-2-naρthoylguanidine,
3Λ5-trimethoxycinnamoylguanidine,
3-methylcinnamoylguanidine,
3-(trans-heρt-l-en-l-yl)oiπnamoylguanidine,
Phenamil methanesulfonate salt ,
2,4-dichlorocinnamolyguanidine,
(4-Nitrocinnamoyl)guanidine,
3,4-difluorocinnamoylguanidine and
[(E)-3-(4-Dimethylaminophenyl)-2- methylacryloyl]guanidine.
81. The method according to claim 58, wherein said virus is the Equine Arteritis virus.
82. The method according to claim 81 , wherein said compound is selected from the group consisting of
5-(NJSf-hexamethylene)amiloride, (3-Bromocinnamoyl)guahidine, trans-3-(l-napthyl)acryloylguanidine, 2-t-butylcinnamoylguanidine and 2-(cyclohex-l-en-lyl)cinnamoylguanidine.
83. The method according to any one of claims 58 to 82, wherein said compound is provided as a pharmaceutical composition according to claim
. 4 or claim 5.
84. " A method ofdown regulating a membrane ion channel functional activity in a cell infected with a virus, comprising contacting said cell with a compound according to any one of claims l to 3.
85. The method according to claim 84, wherein said virus is a Lentivirus.
86. The method according to claim 85, wherein said Lentivirus is Human Immunodeficiency Virus (HIV).
87, The method according to claim 86, wherein said membrane ion channel is the HIV Vpu membrane ion channel.
88. The method according to claim 87, wherein said compound is selected from the group consisting of
(3-Chlorocinnamoyl)guanidine, (3-Bromocinnamoyl)guanidine, (2-Chlorocinnamoyl)guanidine, . (2-Bromociπnamoyl)guanidine, 3-(trifluoromethyl)cinnamoylguanidine, 5-bromo-2-fluorocinnamoylguanidine, 3-methylcinnamoylguanidine, 62-
2-methylciιmamoylguamdine,
2,3-dimethylcinnamoylguanidine,
Cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine, trans-3-(l -napthyl)acryloylguanidine,
3,4-dichlorochmamoylguamdine,
2,6-dichlorocinnamoyϊguanidine,
4-phenylbenzoylguanidine,
2-efhylcinnamoyIguanidine,
(4-Chlorocinnamoyl)guanidine„
2-naρthoylguanidine,
2,5-dimethylcinnamoylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
3-phenylcinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(3 -bromophenyl)penta-2,4-dienoylguanidine,
3-(cyclohex- 1 -en- l-yl)cinnamoylguanidine,
3-(trifluorømethoxy)cinnamoylguaπidine,
2-(trifluoromemyl)cinnamoylguanidine,
N,N'-bis(3phenylpϊopa«oyl)-N"-ρhenylguanidine,
2-ethoxycinnamoylguanidine,
N-ζS-phenylpropamoylH'ϊ'-phenylguanidine,
4-(tτi luoromethyl)cinnamoylguanidine,
(4-Methoxycinnamoyl)guanidine,
2-t-butylcinnamoylguanidine,
4-methylcinnamoylguanidine,
2-fIuorocinnamoylguanidine,
2-phenylcinnamoylguanidine,
N-(6-Hydroxy-2-napthoyl)-Nt-phenylguanidine,
3-t-butylcinnamoylguanidine,
3,4-difluorocinnamoylguanidine,
5-(N,N-hexamethylene)amiloride,
3-fluorocianamoylguanidine,
5-bro o-2-methoxycinnamoylguanidine,
3-ethoxycinnamoylguanidme,
3,4-(methylenedioxy)cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
2'4 DichloroBenzamil HCI,
2,3,5,6,-tetramethylcinnamoylguanidine,
3-(2-napthyl)acryloylguanidine,
2-(l-napthyl)acetoylguanidine,
2,3-difluorocinna oylguamdine,
(3-Methoxycinnamoyl)guanidine,
4-isopropylciπnamoylguanidine,
2,4,6-trimcthylcinnamoylguanidine,
N-(ciαnamoyl)-N'phenylgua!nidine,
2-(cyclohex-l-en-lyl)cinnamoylguamdine, 2-(2-napthyl)acetoylguanidine,
(4-Hydroxycinnamoyl)guaπidine,
4-phenylcinnamoylguanidine,
4-fluorocitιnamoylguanidine,
N,N,-bis-(cinnamoyl)-N"-pheuylguanidine,
(2-Furanacayloyl)guanidine,
Phenamil methanesulfonate salt ,
Benzamil hydrochloride,
(3-Nitrocinnamoyl)guanidine,
Benzyoylguanidine,
(4-Phcnoxybenz yl)guanidine,
3-(trans-hg>t-l-en-l-yl)cinnamoylguanidine,
5-(N-Methyl-N-isobutyl)amilori e,
2-cycIohexylcinnamoylguanidine,
4-ethoxyciπnamoylguanidine,
2,4-dichlorocinnamolyguaπidine,
5-(N-Ethyl-N-isopropyl)amiloride,
N-amidino-3-amino-5-hexamethyleneimino-6-phenyl-
2-pyrazinecarboxamide,
(a-Methyicinnamoyl)guanidine, cinnamoylguanidine hydrochloride,
[(4-Chlorophenoxy-ace1yl]guaπicϋne,
N-amidino-3-amino-5-phenyl-6-chloro-2- pyrazinecarboxamide,
5-(4-fluorophenyl)amiloride,
(trans-2-Phenylcyclαpropanecarbonyl)guanidine,
(2-Nitrocinnambyl)guanidine, trans-3-Furanacryoylguanidine,
1-naρthoylguanidine,
5-tert-butylamino-amiloride,
3-methoxy -HMA,
(3-phenylprqpanoyl)guanidine,
4-t-butylcinnamoylguanidine,
5~(N,N-Dimethyl)amilcride hydrochloride,
NJST-Bis(3-phenylpropanoyl)guanidine,
N-Benzoyl~ -cinnamoylguanidine and l-bromo-2-napthoylguanidine.
89. The method according to any one of claims 86 to 88, wherøn said HIV is HIV-1.
90. The method according to claim 84, wherem said virus is a Coronavirus.
91, The method according to claim 90, wherem said membrane ion channel is the Coronavirus E protein.
92. The method according to claύn 91, wherein said Coronavirus is the Severe Acute Respiratory Syndrome virus (SARS).
93. The method according to claim 92, wherein said compound is selected from the group consisting of
2,3-difluorocinnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
4-t-butylcinnamoylguanidine,
3f(2-napthyl)acryioylguamdines
(3-Chlorociαaamoyl)guanidine,
S-^yclohex-l-en-l-y^ nnamoylguaaidine^
2,5-dimethylcinnamoylguanidine, trans-S-fl-napthy^acryloylguanidine, •
4-isoρroρylcinnamoylguanidine,
(3-Bromodnnamoyl)guanidine,
6-methoxy-2-naphthoylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
3-phenylcinnamoylguanidine,
(2-Ofiorøcannamoyl)guamdine,
2'4 DichloroBenzamil HCI,
4-phenylcinnamoylguauidine,
4-(trifluoromerthyl)cinnamoylguanidine,
3-(trifluoromethoxy)cinnamoylguanidine,
3-(trifluoromethyl)cinnamoylguamdine,
2-ethoxycinnamoylguanidine, cinnamoylguanidine hydrochloride,
4-ethoxycύmamoylguanidme,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
5-tert-butylamino-amiloιide,
3-t-butylcinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
(4-Chlαrocinnamoyl)guanidine, .
2-t-butylcinnamoylguanidine,
2-cyclohexylcinnamoylguanidine,
6-Iodoamilαride,
3-(trans-hept-l -en-1 -yl)cinnamoylguanidine,
(4-Bromocmnamoyl)guanidine,
(4-Hydroxyάnnamoyl)guanidine,
N-(3-ρhenylpropanoyl)-N'-phenylguanidine,
(3-Nitrocinnamόyl)guanidine,
3-fluorocinna oylguanidine,
2-(l-naρthyl)acetoylguanidine, 465-
2-ethyIcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
2-napthoylguanidine,
5-(4-fluorophenyl)amiloride,
2-(trifluoromethyl)cinnamoylguanjdine,
N-(6-Hydroxy-2-naρthoyl)-N'-ρhenylguanidine,
(trans-2-Phenylcycloρroρanecarbonyl)guanidine,
N,N'-bis(3phenylpropanoyl)-N"-phenylguamdine,
1 -napthoylguanidine,
Benzamil hydrochloride,
3-mcthoxy-HMA,
4-methylcinnamoylguanidine,
4-fluorøcinnamoylguanidiαe,
3,4-(methylenedioxy)cinnamoylguanidine, .
5-(N,N-hexamethylene)a iloride,
N-(cinnamoyl)-N^)henylguanidine,
5-(N-Ethyl-N-isopropyl)amiloride,
3-methylcinnamoyiguanidine,
2-melhylcinnamoylguanidine,
2,3,5,6,-tettamet yl nnamoylguanidine, trans-3-Fuιanacryoylguanidine,
(4-Methoxycinnamoyl)guanidine,
(2-Furanacryloyl)guanidine,
(3-ph ylpropanoyl)guanidine,
2-(2-napthyl)acetoylguanidine,
Cinnamoylguanidine,
(2-Methoxydnnamoyl)guanidine,
[3-(3-Pyridyl)acryloyl]guanidine,
4-phenylbenzoylguanidine,
2,4-dichlorocinnamolyguanidine,
(3-Methoxycinnamoyl)guanidine,
2-fluorociπnamoylguanidine,
(4-Phenoxybenzoyl)guanidina,
(a-Methylcinnamoyl)guanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guaridkte,
(Quinoline-2-carbonyl)guanidine,
(Phenylacetyl)guanidine,
N,N'-Bis(amidino)napthalene-2,6-dicarboxamide,
6-bromo-2-napthoylguanidine, l-bromo-2-napthoylguanidine,
2-chloro-6-fluorocinnamoylguattidine,
[(4-Chloroρhenoxy-acetyl]guanidine,
Phenamil ethanesulfonate salt ,
N-Benzόyl-N'-cinnamoylguanidine and
N-(2-napthoyl)-N'-phenylguanidine.
94. The method according to claim 91, wherein said Coronavirus is human Coronavirus 229E.
95. The method according to claim 94, wherein said compound is selected from the group consisting of
4-isoρropylcinnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
3-(trifluqromethoxy)cinnamoylguanidine,
4-t-butylchmamoylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
3-t-butylcinnamoylguanidine,
2-t-butylcinnamoylguanidine, trans-3-(l -napthyl)acryloylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
2,3-difluorocinnamoylguanidbιe,
3-(2-napthyl)acryloylguanidine,
2-phenylcinnamoylguanidine,
3-phenylcinnamoylguanidine,
3-(cyclόhex-l -en-1 -yl)cinnamoylguanidine,
4-phenylbenzoylguanidine,
3-(tiifluoromethyl)cinnamoylguanidine,
(4-Phenoxybenzoyl)guanidiαe,
4-(trifluoromethyl) nnamoylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguaπidine,
(4-Bromocinnamoyl)guanidiue,
5-(N,N-hexamethyl e)amiloride, l-n^>1hoylguanidiue,
5-(4-fluorophenyl)amiloήde,
(5-Phenyl-peπta-2,4-dien yl)gU;amdine,
(3-Bromocinnamoyl)guanidine,
2,5-di ethylcinnamoylguaradine,
2-(trifluoromethyl)cinnamoylguanid e,
6-methoxy-2-naphthoylguaπidine,
(4-Chlorocinnamoyl)guamdine,
(3-Methoxycinnamoyl)guanidine,
5-bromo-2-fluoroQinnamoylguanidine,
5-(N^-Dhnethyl)amiloride hydrochloride,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
(a-Meώylcinnamoyl)guaπidine,
4-ρhenylcinnamoylguanidine,
2,6-dichlorocinnamoylguanidtne,
(2-Bromocinnamoyl)guanidine,
2,4,6-trimethylcinnamoylguanidine, (traήs-2-Phenylcyclopropanecarbonyl)guamdine,
(3-Chlorocinnamoyl)guanidine,
2-(l-napthyl)acetoylguanidine,
2-ethylcinnamoylguanidine,
2-cyclohexylcinnamoylguanidine,
(4-Hydroxyc«mamoyl)guanidinje,
2-ethoxycimιamoylguanidine,
3-methylciιmamoylguanidine,
2-methylcinnamoylguanidine,
3-fluorocinnamoylguanidine, cinnamoylguanidine hydrochloride,
2,3-dimethylcinnamoylguanidine,
2-fluorocinnamoylguanidine,
4-fluorocinnamoylguanidine,
3,4-difluorocinnamoylguanidine,
5-tert-butylamino-amiloride,
2-napthoylguanidine,
N*N,-Bis(amidino)napthalene-2,6-dicarboxamide;i
N,N,-Bis(3-phenylρroρanoyl)guanidine,
4-methylciιmamoylguanidine,
S-fS'-bromophenytypenta^^-dienoylguanidine,
2,3,5,6,-tetrarøethylcinnamoylguanidine,
3-ethoxycannamoyIguanidine,
NJSP-bis(3phenylpropanoyl)-N"-phenylguanidine,
(4-Melhoxyciήnamoyl)guamdine,
(2-Chlorocinnamoyl)guanidine,
(3-Nitroc3nnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
3,4,5-lrimefhoxycinnamoylguaiiidine,
2-(2-napthyl)acetoylguanidine and
N^-phenylpropanoy^- -phenylguanidine.
96. The method according to claim 91 , wherein said Coronavirus is any one of the known Coronavirus isolates listed in Table 1.
97. The method according to claim 96, wherein said compound is selected from the group consisting of
4-isopropylcinna oylguanidine, 3,4-dichlorocinnamoylguanidme, 3-(trifluoromethoxy)cinnamoylguanidine, 4-t-butylcinnamoylguanidine, 3-isopropylcmnamoylguanidine hydrochloride, 468-
98. The method according to claim 84, wherein said virus is the Hepatitis C virus.
99. The method according to claim 98, wherein said membrane ion channel is the Hepatitis C virus p7 membrane ion channel.
100. The method according to claim 99, wherein said compound is selected from the group consisting of
2s3-dimethylcinnamoylguanidine,
2,4,6-trimethylcήmamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
3-(trifiucxromethyl)cinnamoylguanidine,.
4"(trifluoromethyl)cinnamoylguamdine,
6-methoxy-2-naphthoylguanidine,
(2-Chlorocinnamoyl)guanidine,
(4-CMoroc namoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguamdine,
(S-Bromocmnamoy^guanidine,
(3-Chloτocmnamoyl)guanidine,
2-(teifluoromethyl)cinnamoylguamdine,
(4-?henoxyberøoyl)guanidine,
3,4-dichlorociπnamoylguaπidine,
4-isopropylcinnamoylguanidine, trans-3-(l -napthyl)acryloylguamdine,
4-t-butylcinnamoylguanidine,
2-t-butyl nnamoylguanidtne,
2-ethylcinnamoyϊguanidine,
4-methylcmnamoylguaπidme,
5-bromo-2-methoxycmπamoylguanidme,
3-(trifluσromethoxy)cinnamoylguanidine,
2-cyclohexyldnnamoylguanjdme,
1-naρthoylguanidine, .
3-t-butylcinnamoylguanidine,
4-phenylbenzoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
N-( nnamoyi)-N^)henylgπanidines l
3-isopropylcinnamoylguanidine hydrochloride,
Benzamil hydrochloride,
N-(3-ρhenylpropanoyl)-Nτ-phenylguanidine,
NJ^-bis(3phenylpropanoyl)-N"-ρhenylguanidines 3-(2-napthyl)acryloylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
2'4 DichloroBenzamil HCI,
5-tert-butylamino-amiloride,
5-(N-Ethyl-N-isopropyl)amiloride,
(4-Methoxycinnamoyl)guanidine,
4-fluorocinnamoylguanidine,
(3-Nitrocinnamoyl)guanidine,
4-ethoxycύmamoylguanidine,
(4-Hydroxycinnamoyl)guanidύιe,
(trans-2-Phenylcycloρropanecarbonyl)guanidine,
3-ethoxyci mamoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
4-phenylcinnamoylguanidine, trans-3-Furanacryoylguanidine,
N-(6-Hydroxy-2-napthoyl)-N'-phenylguanidine,
(2-Furanacryloyl)guanidine,
3-(cyclohex-l-en-l-yl)cidnamoylguanidine, cinnamoylguanidine hydrochloride,
5-(N,N-hexamethylene)amiloride,
2,3-difluorocinnamoylguanidine,
2-(l-naρthyl)acetoylguanidine,
(a-Methylcinnamoyl)guanidine,
(2-Nitrocinnamoyl)guanidine, -Iodoamiloride,
3,4-(methylenedioxy)cinnamoylguanidine,
2-efhoxycinnamoylguanidine,
Cinnamoylguanidine,
2-phenylcinnamoylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
2-napthoylguanidine,
3-phenylcinnamoylguanidine,
5-(N)N-Dimethyl)amiloride hydrochloride,
5-(4-fluorophenyl)amiloride,
(3-Methoxycinnamoyl)guanidine,
2-fiuorocinnamoylguanidine,
5-(3-bromophenyl)ρenta-2,4-dienoylguanidiπe,
[(4-Chlorophenoxy-acetyl]guanidine,
(3-phenylpropanoyl)guanidine,
2-chloro-6-fluorocimιamoylguanidine,
3-fluorocinnamoylguanidine,
2-methyicinnamoylguanidine,
(2-Methoxycinnamoyl)guaπidine}
1 -bromo-2-napthoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
3-methylcinnamoylguanidine,
3 -(trans-hept-1 -en- l-yl)cinnamoylguanidine,
Phenamil ethanesulfonate salt , 470-
2,4-dichlorocinnamolyguanidine, (4-Nitrocinnamoyl)guanidine, 3,4-difluorocinnamoylguanidine and [^)-3-(4-Dimethylaminqphenyl)-2- methylacryloyl]guanidine.
101. The method accordmg to any one of claims 84 to 100, wherein said compound is provided as a pharmaceutical composition according to claim 4 or claim 5.
102, A method of reducing, retarding or otherwise inhibiting growth and/or repHcation of a virus that has infected a cell, said method comprising contacting said infected cell with a compound according to any one of " claims 1 to 3, wherein said compound down regulates functional activity of a membrane ion channel derived from said virus and expressed in said infected cell.
103. The method according to claim 102, wherein said virus is a Lentivirus.
104. The method according to claim 103, wherein said Lentivirus is Human Immunodeficiency Virus (HIV).
105. The method according to claim 104, wherem said membrane ion channel is the HIV Vpu membrane ion channel.
106. The method according to claim 105, wherein said compound is selected from the group consisting of
(3-Chlorocinnamoyl)guanidine,
(3-Bromocinnamoyl)guamdine,
(2-Chlorocinnamoyl)guanidine,
(2-Bromoc.iιxnamoyl)guanidine,
3-(trifluoro ethyl)cinnamoylguanidhιe,
5-bromo-2-fluorocinnamoylguanidine,
3-methylcinnamoylguanidine,
2-methylcinnamoylguanidine,
2^-dimefhylcinnamoylguanidine, 471-
Cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine, trans-3-(l -napthyl)acryloylguanidine,
3,4-dichlorocir amoyIguanidine,
2,6-ώchlorocinnamoylguanidine,
4-phen.ylbeπzoylguanidine,
2-ethylciπnamoylguanidine,
(4-Chlorocinnamoyl)guanidine„
2-naρthoylguanidine,
2,5-dimethylcinnamoylguanidine,
3-isoρroρyl nnamoylguanidine hydrochloride,
(5-Phenyl-penta-2,4-dienoyl)guanidme,
3-phenylcinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
3-(cyclohex-l-en-l-yl)cmnamoylguamdine,
3-(tiMuorαmethoxy)cinnamoylguanidine,
2-(trifluoromethyl)cinnamoylguanidine,
N,N'-bis(3phenylpropanoyl)-N"-phenylguanidine,
2-ethoxycinnamqylguanidine,
^(S-phenylpropanoy^-NP-phenylguanidine,
4-(trifluoromethyl)cύ amoylguanidine,
(4-Methoxycinnamoyl)guanidine,
2-t-butylcmnamoylguanidine,
4-metttyIcinnamoylguanidine,
2-fluorocinnamoylguanidine,
2-ρhcnylcinnamoylguanidine,
N-(6-Hydroxy-2-napthoyl)-l P-phenylguanidine,
3-t-butylcinnamoylguanidina,
3,4-difluorocinnamoylguanidine,
5-(N,N-hexamethylene)amiloride,
3-fluorocinnam.oylguaπidine,
5-bromo-2-methoxycinnamoylguanidine,
3-ethoxycinnamoylguanidine,
3,4-(methylenedioxy)cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
2'4 DichloroBenzamil HCI,
2,3,5,6,-tetramethyl nnamoylguaπidine,
3-(2-napthyl)acryloylguanidine,
2-(l -naρthyl)acetoylguanidine,
2,3-difIuorocinnamoylguanidine,
(3-Methoxycinnamoyl)guanidine,
4-isopropylcinnamoylguanidine,
2,4,6-trimethylcinnamoylguanidine,
N-(cinnamoyl)-N'phenylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguaιύdiαe,
2-(2-napthyl)acetoylguanidine,
(4-Hydroxycinnamoyl)guanidine, 4-phenylcinnamoylguanidine,
4-fiuorocinnamoylguanidine,
NJSP-bis-(cinnamoyl)-N"-phenylguanidine,
(2-Fuxanacryloyl)guanidine,
Phenamil methanesul&nate salt ,
Bβnzamil hydrochloride,
(3-Nitrocinnamoyl)guanidine,
Benzyoylguanidine,
(4-Pheπoxybeπzoyl)guanidine,
3-(trans-hept-l-en-l-yl)cύmamoylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
2-cyclohexylcinnamoylguamdine,
4-ethoxycittuamoylguanidine,
2,4-dichlorocinnamolyguanidine,
5-(N-Ethyl-N-isopropyl)amiloride,
N-anwdino-3-amino-5-hexamethyleneimino-6-phenyl-
2-pyra2 necarboxamide,
(a-Methylcinnamoyl)guanidine, cinnamoylguanidine hydrochloride,
[(4-Chloroρhenoxy-acetyl]guanidine,
N-amidino-3-amino-5-ρhenyl-6-chloro-2- pyrazinecarboxamide,
5-(4-fluorophenyl)amiloride,
(trans-2-Phenylcyclopropanecarbonyϊ)guanidine,
Figure imgf000173_0001
2 h l idi
107. The method according to any one of claims 104 to 106, wherein said HIV ia HIV-1.
108. The method according to claim 102, wherein said virus is a Coronavirus.
109. The method according to claim 108, wherein said membrane ion channel is the Coronavirus E protein.
110. The method according to claim 109, wherein said Coronavirus is the Severe Acute Respiratoiy Syndrome virus (SARS).
111. The method according to claim 110, wherein said compound is selected from the group consisting of
2,3-difluorocinnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
4-t-butylcinnamoylguaπidine,
3-(2-napthyl)acryloylguanidine,
(3-Chlorocinaamoyl)guanidine,
3-(cyclohex-l-en-l-yl)cimιamoylguanidine,
2,5-dύnethylciπnamoylguanidinei, trans-3-(l-napthyl)acιyloylguanidine,
4-isopropylcinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
6-methoxy-2-naρhthoylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
3-phenylcinnamoylguanidine,
(2-Chlorocinnatnoyl)guanidine,
2'4 DichloroBenzamil HCI,
4-phenylcinnamoylguaπidine,
4-(trifluoromethyl)cinnamoylguanidine,
3-(trifluoromethoxy)cinnamoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
2-ethoxycinnamoylguanidine, cinnamoylguanidine hydrochloride,
4-ethoxycinnamoylguaπidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
5-tert-butylamino-amiloride,
3-t-butylcinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
(4-Chlorocinnamoyl)guanidine,
2-t-butylcinnamoylguanJdine,
2-cyclohexylcύmamoylguanidine,
6-Iodoamiloride, _
3-(trans-hept-l-en-l-yl)cinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
(4-Hydroxycinnamoyl)guanidine,
N-(3-phenylρropanoyl)-N1-ρhenylguanidine,
(3-Nitrocinnamoyl)guanidine,
3-fluorocinnamoyiguanidine,
2-(l-napthyl)acetoylguanidine, 474-
2-ethylcinnamoylguanidine,
5-(N,N-Dimethyl)arniloride hydrochloride,
2-napthoylguanidine,
5-(4-fluorophenyl)amiloride,
2-(trifluoromethyl)cinnam yIguanidine,
N-(6-Hydroxy-2-napthoyl)-N'-phenylguanidine,
(trans-2-Phenylcycloproρanecarbonyl)guanidine,
N,N,-bis(3phenylpropanoyl)-N"-phenylguanidine,
1 -napthoylguaπidine,
Benza il hydrochloride,
3-methoxy -HMA,
4-methylcinnamoylguanidine,
4-fluorociraιamoylguanidine,
3,4-(me hylenedioxy)cinnamoylguanidine,
5-(N,N-hexamethylene)amiloride,
N-(cinnamoyl)-N,phenylguaπidine,
5-(N-Ethyl-N-isopropyl)amiloride,
3-methylcinnamoylguanidine,
2-methylcinnamoylguamdine,
2,3,5,6,-tetramethylcinnamoylguanidine, trans-3-Furanacryoylguanidine,
(4-Methoxycinnamoyl)guanidine,
(2-Furanacryloyl)guanidine,
(3-phenyIρropanoyl)guanidine,
2-(2-n^>thyl)acetoylguaπidine,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
[3-(3-Pyridyl)acιyloyl]guanidine,
4-phenylbenzoylguanidine,
2, dichlorocinnamolyguanidine,
(3-Methoxycinnamoyl)guanidme,
2-fluorocinnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
(a-Methylcmnamoyl)guanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
(Quinoline-2-carbonyl)guanidine,
(Phenylacetyl)guanidine,
N^-Bis(amidino)napthaIene-2,6-dicarboxamide,
6-brαmo-2-napthoylguanidine, l-bromo-2-napthoylguanidine,
2-chloro-6-iluorocinnamoylguanidine,
[(4-Chlorophenoxy-acetyl]guanidine,
Phenamil methanesulfonate salt ,
N-Benzoyl-N'-cinnamoylguanidine and
N-(2-napthoyl)-N-phenylguaήidine.
112. The method accordmg to claim 109, wherein said Coronavirus is human Coronavirus 229E.
113. The method according to claim 112, wherein said compound is selected from the group consisting of
4-is propylcinnamoylguanidine,
3,4-<ώchlorocinnamoylguanidine3
3-(trifluoromethoxy)citmamoyϊguamdine,
4-t-butylcinnamoylguanidine,
3-isopτopylcinnamoylguanidine hydrochloride,
3-t-butylcinnamoylguanidine,
2-t-butylcinnamoylguaπidine, trans-3-(l-napthyl)acryloylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
2,3-difluorodnnamoylguanidine,
3-(2-napthyl)acryloylguanidine,
2-ρhenylcinnamoylguanidine,
3-phenyldnnamoylguanidine,
3-(oyclohea:-l-en-l-yl)dnnamoylguanidine,
4-phenylbenzoylguanidine,
3-(trifluoκmethyl)cinnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
4-(trifluo(romethyl)cinnamoylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
1 -napthoylguanidine,
5-(4-fluorophenyl)amiloride,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
(3-Bromo nnamoyl)guanidine,
2,5-dimethyldmιamoylguanidine,
2-(trifluoromethyl)cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
(4-Chlorocinna oyl)guanidine,
(3-MethoxycinnamoyI)guanidine,
5-bromo-2-fkoro rinπamoylguaπidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
(a-Methylcinnamoyl)guanidine,
4-phenylcinnamoylguanidine,
2,6-dichlorociπnamoylguanidine,
(2-Bromocinnamoyl)guanidiπe,
2,4,6-trimethylcinnamoylguanidine, (frans-2-Phenylcyclopropanecarbonyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
2-(l-naρthyl)acetoylguanidine,
2-ethylcitmamoylguanidine,
2-cyclohexylcinnamoylguanidine,
(4-Hydroxycirmamoyl)guanidine,
2-ethoxycinnamoylguanidine,
3-methylciιmamoylguanidine,
2-methylciπnamoylguanidine,
3-fluorocinnamoylguanidine, cinnamoylguanidine hydrochloride,
2,3-dimethylcinnamoylguanidine,
2-fluorocinnamoyϊguanidine,
4-ftuorocinnamoylguanidine,
3,4-difluoϊocinnamoylguanidine,
5-tert-butylammo-amiloride,
2-napthoylguanidine,
N, -Bis(amidino)napthalene-2,6-dicarboxamide,
N.N'-Bis^-phenylpropanoyQguanidine,
4-methylcinnamoylguanidine,
S-tS'-bromophenyl^enta^^-dienoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidme,
3-ethoxycinnamoylguanidine,
N,lSr-bis(3phenylpropanoyl N"-phenylguanidine,
(4-Methoxycinnamoyl)guanidine,
(2-Chlorocinnamoyl)guanidine, .
(3-Nitrocinnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
3,4,5-trimethoxycinnamoylguanidine,
2-(2-napthyl)acetoylguanidine and
N S-phemylpropanoy -N'-phenylguanidine.
114. The method according to claim 109, wherein said Coronavirus is any one of the known Coronavirus isolates listed in Table 1.
115. The method according to claim 114, wherein said compound is selected from the group consisting of
4-isoρroρylcinnamoylguanidine, 3,4-dichlorocinnamoylguanidine, 3-(trifluoromethoxy)cinnamoylguanidine, 4-t-butylcinnamoylguanidine, 3-isopropylcmnamoylguanidine hydrochloride,
116. The method according to claim 102, wherem said virus is the Hepatitis C virus.
117. The method according to claim 116, wherein said membrane ion channel is the Hepatitis C virus p7 membrane ion channel.
118. The method according to claim 117, wherein said compound is selected from the group consisting of
2,3-dimethylcmnamoylguanidine,
2Λ6-t methylcinnamoylguanidine,
5-bromo-2-fluorocimιamoylguanidine,
(4-Bromocinnamoyl)guanidine,
2,5-di ethylcinnamoylguanidinα,
3-(triflu romethyl)cinnamoylguanidine,
4-(trifluoromethyi)cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
(2-Chlorocinnamoyl)guanidine,
(4-Chlorocinnamoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichloτocinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
2-(trifluoromethyl)ciιmamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
3,4-dcMorociπnamoylguanidine,
4-isoρropylcinnamoylguanidine, trans-3-(l -napthyl)acryloylguanidine,
4-t-butylcinnamoylguanidine,
2-t-butylcinnamoylguanidine,
2-ethylcinnamoylguanidine,
4-methylcinnamoylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
3-(trifluoromethoxy)cinnamoylguamdine,
2-cyclohexylcmnamoylguanidine,
1-napthoylguanidine,
3-t-butylcinnamoylguanidiue,
4-phenylbenzoylguanidine,
(5-Phenyl-ρenta-2,4-dienoyl)guanidine,
N-(cinnamoyl)-N^henylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
Benzamil hydrochloride,
N-(3-phenylpropanoyl)-N'-phenylguanidine,
NjN-bisfSphenylpropanoy^-lSr'-phenylguanidine,
3-(2-napthyl)acιyloylguanidme, 5-(N-Methyl-N-isobutyl)amiloride,
2'4 DichloroBenzamil HCI,
5-tert-butylamino-amiloride,
5-(N-Ethyl-N-isopropyl)amiIoride,
(4-Methoxycinnamoyl)guanidine,
4-fl.uorocinnamoylguanidine,
(3-Nitrocinnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
3-ethoxycinnamoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
4-phenylciιmamoylguarddine, trans-3-Furanacryoylguanidine,
N-(6-Hydroxy-2-n^)thoyi)-N-phenylguanidine,
(2-Furanacryloyl)guanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine, cinnamoylguanidine hydrochloride,
5-(N,N-hexamethyleπe)amiloride,
2,3-difluorocinnamoylguanidine,
2-(l-napthyl)acetoylguanidine,
(a-Methylcinnamoyl)guanidine,
(2-Nitrp nnamoyl)guaπidine,
6-Iodoamiloride,
3,4-(mefhylenedioxy)cinnamoylguanidin&,
2-ethoxycinnamoylguanidine,
Cinnamoylguanidine,
2-ρhenylcinnamoylgύanidine,
2-(cyclohex- 1 -en-1 yi)cinnamoylguanidine,
2-napthoylguanidine,
3-ρhenylcinnamoylguanidtne,
5-(N,N-Dimethyl)amilo:tide hydrochloride,
5-(4-fluorophenyl)amiloride,
(3-Methoxycionamoyl)guanidine,
2-fluorocinnamoyiguanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
[(4-Chloroρhenoxy-acetyl}guamdine,
(3-phenylpropanoyl)guanidine,
2-cMoro-6-fluorooinnamoylguanidine,
3-fluorocinnamoylguanidine,
2-methylcinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
1 -bromo-2-naρthoylguanidine,
3A5-trύnethoxycirmamoylguanidine,
3-methylcinnamoylguanidine,
3-(tians-hept-l -en-l-yl)chmamoylguanidine,
Phenamil methanesulfonate salt ,
2,4-dichlorocinnamoiyguanidine, 479-
(4-Nitrocinnamoyl)guanidine, 3,4-difluorocinnamoylguanidine and [(E)-3-(4-Dimethylaminophenyl)-2- methylacryloyljguanidine.
119. The method according to any one of claims 102 to 11 S, wherein said compound is provided as a pharmaceutical composition according to claim 4 or claim 5.
120. A method of reducing, retarding or otherwise inhibiting growth and/or replication of a virus that has infected a cell in a mammal, said method comprising administering to said mammal a compound according to any one of claims 1 to 3, wherein said compound down regulates functional activity of a membrane ion channel expressed in said infected cell.
121. The method according to claim 120, wherein said virus is a Lentivirus.
122. The method according to claim 121, wherein said Lentivirus is Human Immunodeficiency Virus (HIV).
123. The method accordmg to claim 122, wherein said compound is selected from the group consisting of
(3-Chlorocinnamoyl)guamdine,
(3-Bromocinnamoyl)guanidine,
(2-Chlorocinnamoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
3-(tiifluoromethyl)cirøamoylguanidine,
5-bromo-2-fluorociDnamoylguanidine,
3-methylcinnamoylguanidϊne,
2-methylciπnamoylguanidine,
2,3-dimethylcinnamoylguanidine,
Cinnamoylguanidine,
6-melhoχy-2-naphthoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
3,4-dichlorocinnamoylguanidine,
2,6-dichlorocinπamoylguanidine,
4-phenylbenzoylguanidine,
2-ethylcinnamoylguanidάne,
( -Chlorocinnamoyi)guanidine,, 480-
2-napthoylguamdine, 2,5-dimethylcinnamoylguanidine, 3-isopropylcinnamoylguanidine hydrochloride, (5-Phenyl-ρenta-2,4-dienoyl)guanidine, 3-phenylcinnamoylguanidhιe, (4-Bromocinnamoyl)guanidine, 5-(3'-bromophenyl)penta-2,4-dienoylguamdine, 3-(cyclohex-l -en-1 -yl)cinnamoylguanidine, 3-(tπfluoromethoxy)cinnamoylguanidine, 2-(tτifluoromethyl)cinnamoylguamdine, • NJ^-bis(3phenylpropanoyl)-N1,-phenylguanidine, 2-ethoxycinnamoylgua dine, N-(3-phenyl^ropanoyl)-N'-phenyϊguanidine, 4-(trifluoromethyl)cinnamoylguanidine, (4-Mefhoxycinnamoyl)guanidine, 2-t-butylcinuamoylguanidine, 4-methylcinnamoylguanidine, 2-fluorocinnamoylguanidine, 2-ρhenylcinnamoylguanidine, ^(β-Hydroxy^-napthoyl^N -pheαylguanidine, 3-t-butylcinnamoylguanidine, 3,4-difluorocinnamoylguanidine, S-fN.N-hexamethyleneJamiloride, 3-fluorocinnamoylguanidine, 5-bromo-2-methoxycmnamoylguanidine, 3-ethoxycinnamoylguanidine, 3,4- meifhylenedioxy)cinnamoylguanidine, (2-Methoxyciιrøamoyl)guanidine, 2'4 DichloroBenzamil HCI, 2,3,5,6,-tetramethylcinnamoyiguanidine, 3-(2-napthyl)acryloylgπanidine, 2-(l-napthyl)acetoylguanidine, 2,3-difiuorooinnamoylguanidine, (3-Methoxy nnamoyl)guanidine, 4-isopropyloinnamoylguanidine, 2,4,6-trimethylcinnamoylguanidine, N-(cinnamoyl)-N'phenylguanidine, 2-(cyclohex-l-en-lyl)cinnamoylguanidine, 2-(2-napthyl)acetoylguanidine, (4-Hydroxycinnamoyl)guanidine, 4-phenylcirmamoylguanidine, 4-fluorocinnamoylguanidine, NjN-bis-ζcinnamoylJ-N'-phenylguanidine, (2-Furanacryloyl)guaπidine, Phenamil methanesulfonate salt , Benzamil hydrochloride, (3-Nifrocnmamoyl)guanidine, Benzyoylguanidine, (4-Phenoxybenzoyl)guanidine,
3-(trans-hept-l-en-l-yl)cinnamoylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
2-cyclohexylcinnamoylguanidine,
4-ethoxycinnamoylguanidύie,
2,4-dichlorocinnamolyguanidine,
5-(N-Ethyl-N-isoρropyl)amiloride,
N-amidino-3-amino-5-hexamethyleneimino-6-phenyl-
2-ρyrazinecarboxamide,
(a-Methylcmnamoyl)guaπidine, cύma oylguanidine hydrochloride,
[(4-Chloroρhenoxy-acetyl]guanidina,
N-amidino-3-amino-5-ρhenyl-6-chloro-2- pyrazinecarboxamide,
5-(4-fluorophenyl)amiloride,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
(2-Nitrocinnamoyl)guanidine, trans-3-Furanaxryoylguanidine,
1-naρthoylguanidine,
5-tert-butylamino-amiloride,
3-methoxy -HMA,
(3-phenylpropanoyl)guanidine,
4-t-butylcinnamoylguanidine,
5-(N,N-Dύnethyl)amilo:ride hydrochloride,
N,N-Bis(3-ph ylpropanoyl)guanidinft,
N-Benzoyl-N'-cinnamoylguanidine and l-bromo-2-napthoylguanidine.
124. The method according to any one of claims 120 to 123, whereύi said membrane ion channel is the HTV Vpu membrane ion channel.
125. The method according to any one of claims 122 to 124, wherein said HIV is HIV-1.
126. The method according to claim 120, where said virus is a Coronavirus.
127. The method according to claim 126, wherein said Coronavirus is the Severe Acute Respiratory Syndrome virus (SARS). The method according to claim 127, wherein said compound is selected from the group consisting of
Figure imgf000183_0001
S-(N^Sf-Dimethyl)amiloride hydrochloride,
2-napthoylguanidine,
5-(4-£luorophenyl)amiloride,
2-(trifluoromethyl)cinnamoylguanidine,
N-(6-Hydroxy-2-napthoyl)-N,-phenylguanidine, 483-
(trans-2-Phenylcyclopropanecarbonyl)guamdine, ^T,-bis(3phenylpropanoyl)-N"-phenylguanidme,
1 -napthoylguanidine,
Benzamil hydrochloride,
3-methoxy-HMA,
4-methylcύmamoylguanidine,
4-fluorocinnamoylguanidme,
3,4-(methylenedioxy)ciunamoylguanidine,
5-(N,N-hexamethylene)amiloride,
N-(cinnamoyl)-N henylguanidine,
5-(N-Ethyl-N-isopτσpyl)amiloride,
3-methylcinnamoylguanidine,
2-methylcinnamoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine, trans-3-Furanacryoylguanidine,
(4-Methoxycinnamoyl)guaπidine,
(2-Furaαacryloyl)guanidine,
(3-phenylpropanoyl)guanidine,
2-(2-napthyl)acetoylguanidine,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
[3-(3-Pyridyl)acryloyl]guamdine,
4-ρ ylbenzoylguanidine,
2,4-dichlorocinnamolyguanid e,
(3-Meth xycinnamoyl)guanidine,
2-fluorociπnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
(a-Methylcinnamoyl)guanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
(5-Phenyl-pemta-2,4-dienoyl)guamdine,
(Quinoline-2-carbonyl)guanidine,
(Fhenylacetyl)guanidine,
N,l^-Bis(amidino)napthalene'2,6-dicarboxamide,
6-bronio-2-napthoylguanidine,
1 -bromo-2-napthoylguanidine,
2-chloro-6-fluorocmnamoylguanidine,
[(4-Chlorophenoxy-acetyl]guanidine,
Phenamil methanesulfonate salt ,
N-Benzoyl-N'-cinnamoylguanidine and
N-(2-napthoyl)-N1-phenylguani(frπje.
129. The method according to any one of claim 126 or 128, wherein said membrane ion channel is the Coronavirus E protein.
130. The method according to claim 126, wherein said Coronavirus is human Coronavirus 229E.
131. The method according to claim 130, wherein said compound is selected from the group consisting of
132. The method according to claim 131, wherein said compound is selected from the group consisting of
4-isopropylcirniamoylguanidme,
3,4-dichlorocinnamoylguanidine,
3-(trifluoromethoxy)citraamoylguanidine,
4-t-butylcinnamoylguaπidine,
3-isopropylcmnamoylguanidine hydrochloride,
3-t-butylcinnamoylguanidine,
2-t-butylcinna oylguanidine, trans-3-(l-napthyl)acryloylguanidine,
5-bromo-2-methoxycinnamoylguanidine,
2,3-difluorocinnamoylguanidine,
3-(2-napthyl)acryloylguanidine,
2-phenylcύmamoylguanidine,
3-phenylcinnamoylguanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
4-phenylbenzoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
4-(tιiQuoromethyl)cinnamoylguanicline,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(N,N-hexamethylene)amiloride,
1-napthoylguanidine,
5-(4-fluorophenyl)amiloride,
(5-PhenyI-ρenta-2,4-dienoyl)guanidine,
(3-Bromocύmamoyl)guanidine,
2,5-dimethylcύτnamoylguanidine,
2-(trifluoromethyl)cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
(4-Chlorocinnamoyl)guanidine,
(3-Methoxycinnamoyl)guarddine,
5-bromo-2-fluorocinnamoylguanidine,
5-(N N-Dimethyl)amiloride hydrochloride,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
(a-Methylcit∞amoyl)guanidine, 85-
4-ρhenylcinnamoylguanidine,
2,6-dic orocmnamoylguanidine,
(2-Bromocinnamoyl)guanidine,
2,4,6-trimefhylcitmamoylguanidine,
(trans-2-Phenylcyclopropanecarbonyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
2-(l -napthyf)acetoylguanidine,
2-ethylcinnamoylguanidine,
2-cyclohexylcinnamoyIguanidine,
(4-Hydroxycinnamoyl)guanidine,
2-ethoxycinnamoylguanidine,
3-methylcinnamoylguanidine,
2-methylcinnamoylguanidine,
3-fiuorociααamoylguanidine, cinnamoylguanidine hydrochloride,
2,3-dimethylcύmamoylguanidine,
2-fluorocinnamoylguaιιidine,
4-fluorocinna oylguanidine,
3,4-difluorocinnamoylguanidine,
5-tert-butyIamino-amiloride,
2-napthoylguanidine,
N»N'-Bis(amidino)napthalene-2,6-dicarboxamide,
HNVBis(3-phenytøropanoyl)guanidine,
4-methylcinnamoyIguamdine,
5-(3-bromophenyl)penta-2,4-dienoylguanidine,
2,3,5,6,-tetramethylcinnamoylguanidine,
3-ethoxycinnamoylguamdine,
N,N'-bis(3phenylpropanoyl)-N"-phenylguanidine,
(4-Methoxycinnamoyl)guanidύιe,
(2-Chlorocinnamoyl)guanidine,
(3-Mtroc namoyl)guanidine,
4-ethoxyciπnan3θylguanidine,
3,4,5-trimethoxycύmamoyIguanidine,
2-(2-napthyl)acetoylguaπidine and
N-(3-phenylpropanoyl)-N'-phenylguanidine.
133. The method according to any one of claims 130 or 132, whereύi said membrane ion channel is the Coronavirus E protein.
134. The method according to claim 126, wherein said Coronavirus is any one of the known Coronavirus isolates listed in Table 1.
135. The method according to claim 134, wherein said compound is selected from the.group consisting of
4-isoproρylcinnamoylguanidine, 3,4-dichlorocιnnamoylguanidine, 3-(trifluoromέthoxy)cimamoylguanidine, 4-t-butyloύmamoylguanidine, 3-isopropylcinnamoylguanidine hydrochloride,
136. The method according to claim 134 or claim 135, wherein said membrane ion channel is the Coronavirus E protein.
137. The method according to claim 120, wherein said virus is the Hepatitis C virus.
138, The method according to claim 137, wherein said compound is selected from the group consisting of
2,3-dimethylcinnamoylguanidine,
2,4,6-trimethylcannamoylguanidine,
5-bromo-2-fiuorocύmamoylguanidine,
(4-Bromocinnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
3-(trifluoromethyl)cinnamoylguanidine,
4-(trifluoromethyl)cinnamoylguaιudine,
6-methoxy-2-naphthoylguanidine,
(2-Chlorocύmamoyl)guanidineι,
(4-Chlorocinnamoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorocinnamoylguanidine,
(3-Bromocinnamoyl)gύanidine,
(3-Chlorocirmamoyl)guanidine,
2-(trifluoromethyl)cinnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
3,4-dichlorocinnamoylguanidine,
4-isopropylcinnamoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
4-t-butylcinnamoylguanidine,
2-t-butylcinnamoylguanidine,
2-ethylcinnamoylguanidme,
4-methylcinnamoylguanidine,
5-bromo-2-methoxycinnamoylguaπidine,
3-(trifluororøethoxy)cinnamoylguamdine,
2-cyclohexylcinnamoylguanidme,
1 -napthoylguanidine, 3-t-butylcinnamoylguanidine,
4-ρhenylbenzoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
N-(cinnamoyl)-N'phenylguamdine,
3-isopropylcinnamoylguamdine hydrochloride;,
Bemzamil hydrochloride,
N-(3-phenylprcφanoyl)-N-ρhenylguanidine,
N,N'-bis(3phenylpropanoyl)-N1'-phenylguanidine,
3-(2-napthyl)acryloylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
2'4 DichloroBenzamil HCI,
5-tert-butylamino-amiloride,
5-(N-Ethyl-N-isoproρyl)amiloride,
(4-Methoxycinnamoyl)guanidine,
4-fluorocinnamoyIguanidine,
(3-Nitrocinnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
(4-Hydroxy ιmamoyl)guaniditte,
(trans-2-Phenylcyclopropanecarbonyl)guaπidine,
3-ethoxycinnamoylguanidine,
2 ,5,6,-tetramethylcinnamoylguanidine,
4-ρhesαyIdnnamoylguanidine, trans-3-Furanacryoylguanidine,
N-(6-Hydroxy-2-napthoyl)-N'-phenylguanidine,
(2-Furanacryloyl)guanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguamdine, cύmamoylguanidine hydrochloride,
5-(N,N-hexamethylene)amiloride,
2,3-difluorocinnamoylguanidine,
2-(I -napthyl)acetoylguanidine,
(a-Methylcinnamoyl)guanidine,
(2-Nitrocinnamoyl)guanidine,
6-Iodoamiloride,
3,4-(methylenedioxy)cinnamoylguanidine,
2-ethoxycinnamoylguanidine,
Cinnamoylguanidine,
2-phιanylcinnamoylguanidine,
2-(cyclohex-l-ein-lyl)cπmamoylguanidine,
2-napthoylguanidine,
3-phenylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
5-(4-fluorophenyl)amiloride,
(3-Mefhoxycinnamoyl)guanidine,
2-fluorocinnamoylguanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
[(4-Chlorophenoxy-acetyl]guanidine,
(3-phenylpropanoyl)guanidine,
2-chloro-6-fluorocinnamoylguanidine, 3-fluorocinnamoylguanidine,
2-methylcinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine, l-bromo-2-napthoylguanidine,
3A5-trimethoxycinnamoylguanidine,
3-methylcύmamoyIguaπidme,
3-(trans-hept-l-en-l-yl)cinnamoylguanidine,
Phenamil methanesu bnate salt ,
2,4-dichlorocJnnamolyguamdine,
(4-Nitrocinhamoyl)guanidine,
3,4-difluorocinnamoylguanidine and
[(E)-3-(4-Dimeflιylaminophenyl)-2- ethylacryloyljguanidine.
139. The method according to claim 138, wherein, said membrane ion channel is the Hepatitis C virus p7 membrane ion channel.
140. The method according to any one of claims 120 to 133, wherein said mammal is a primate.
141. The method according to any one of claims 137 to 139, wherein said mammal is a primate.
142. The method according to claim 140 or claim 141, wherein said primate is human.
143. The method according to any one of claims 120 to 142, wherein said compound is provided as a pharmaceutical composition according to claim
4 or claim 5.
144. A method for the therapeutic or prophylactic treatment of a subject infected with or exposed to a virus comprising administering to said subject a compound according to any one of claims 1 to 3, wherein said compound down-regulates functional activity of a membrane ion channel derived from said virus.
145. The method according to claim 144, wherein said virus is a Lentivirus.
146. The method according to claim 145, wherein said Lentivirus is Human Immunodeficiency Virus (HTV).
147. The method according to claim 146, wherein said membrane ion channel is the HIV Vpu membrane ion channel.
148. The method according to claim 147, wherein said compound is selected from the group consisting of
(3-Chloroc namoyl)guanidine,
(3-Bromocinnamoyl)guanidine,
(2-Chlorocir amoyl)guanidine,
(2-Bromocinnamoyl)guanidine,
3-(trifluoromethyI)cinnamoylguanidine,
5-bromo-2-fluorocinnamoylguanidine,
3-mefhylciπnamoylguanidine,
2-methylcinnamoylguanidine,
2,3-dimethylcinnamoylguanidine,
Cinnamoylguanidine,
6-melhoxy-2-naphthoylguanidine, trøns-3-(l -napthyljacryloylguanidine,
3 ,4-dichlorocinnamoyiguanidine,
2,6-dichlorocinnamoylguanidine,
4-phenyIbenzoylguanidine,
2-ethylcinnamoylguanidine,
(4-Chloroc namoyl)guanidine„
2-napthoylguanidine,
2,5-dimethylcinnamoylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
(5-Phenyl-penta-2,4-dienoyl)guanidine, •
3-phenylcinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
5-(3'-bromoρhenyl)pε!nta-2,4-dienoylguanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
3-(trifluαromethoxy)cinnamoylguanidine,
2-(trifluoromethyl)cinnamoylguanidine,
NjN'-bisfSphenylpropanoyQ-ls -phenylguanidine,
2-ethoxycinnamoylguamdine,
N-(3-ρhmylpropanoyl)-N^phenylguaιtidine,
4-(trifluoromethyl)cmnamoylguanidine,
(4-Methoxycinnamoyl)guanidine,
2-t-butylcinnamoylguanidine,
4-methylcύmamoylguanidine,
2-fluorocinnamoylguanidine, 490-
2-ρhenylcimιamoylguanidine,
N-(6-Hydroxy-2-napthoyl)-N'-phenylguanidhιe,
3-t-butylciιmamoylguanidine,
3,4-difluorocinnamoylguanidine,
5-C ,N-hexamethylene)amiloride,
3-fluorocinnamoylguamdine,
5-bromo-2-methoxycinnamoylguanidine,
3-ethoxycinnamoylguanidine,
3,4-(melhylenedioxy)cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
2'4 DichloroBenzamil HCI,
2,3,5,6,-tetramethylcinπamoylguanidine,
3-(2-napthyl)acryloylguanidine,
2-(l -napthyl)acetoylguanidine,
2,3-difϊUorocinnamoylguanidine,
(3-Methoxycinnamoyl)guanidine,
4-isc^pylcύmamoyIguanidiae,
2,4,6-tritoelhylciπnamoylguanidine,
N-(cinnamoyl)- phenylguanidine,
2-(cycIohex-l-en-l yl)ctnnamoylguanidine,
2-(2-napthyl)acetoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
4-ρhenyIcinnamoylguanidine,
4-fluorocinnamoylguanidine,
N,N,-bis-(cinna oyl)-N"-ρhenylguanidme,
(2-Fuιanacryloyl)guanidine,
Phenamil methanesulfonate salt ,
Benzamil hydrochloride,
(3-Nitrocinnamoyl)guanidine,
Benzyoylgjuanidine,
(4-Phenoxybenzoyl)guanidine»
3-(trans-hept-l-en-l-yl)cήmamoylguanidine,
5-(N-Methyi-N-isobutyl)aniilorides
2-cyclohexylciιmamoylguanidme,
4-ethoxycinnamoylguanidine,
2,4-dicώotocinnamolyguani(fine,
5-(N-Ethyl-N-isopropyl)amiIoride,
N-aπύdino-3-£Bnino-5-hexanι hyleneiπιino-6-phenyl-
2-pyrazinecarboxarmdej,
(a-Methylcinnamoyl)guanidine, cinnamoylguanidine hydrochloride,
[(4-Chlαroρhenoxy-acetyl]guanidine,
N-amidino-3-amino-5-phenyl-6-chloro-2- pyrazinecarboxamide,
5-(4-fluorophenyl)amiloride,
(trans-2-Phcnylcyclopropanecarbonyl)guanidine,
(2-Nifrocinnamoyl)guanidine, trans-3-Furanacryoylguanidine, 491-
1-napthoylguanidine, 5-tert-butylaminα-amiloride, 3-methoxy -HMA, (3-phanylpropanoyl)guanidine, 4-t-butylcinnamoylguanidine, 5-(N,N-Dimethyl)amiloride hydrochloride, N,N'-Bis(3-phenylpropanoyl)guanidine, N-Benzoyl-N'-cinnamoylguanidine and l-bromo-2-n^lhoylguanidine.
149. The method according to any one of claims 146 to 148, wherein said HIV is HIV-1.
150. The method according to claim 144, wherein said virus is a Coronavirus.
151. The method according to claim 150, wherein said membrane ion channel is the Coronavirus E protein.
152. The method according to claim 151, wherein said Coronavirus is the Severe Acute Respiratory Syndrome virus (SARS).
153. The method according to claim 152, wherein said compound is selected from the group consisting of
2,3-difluorociπnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
4-t-butylcmnamoylguanidine,
3-(2-naρthyl)acryloylguanidine,
(3-Chlorocinnamoyl)guanidine,
3-(cyclohesx-l-en-l-yl)cirmamoylguanidine,
2,5-dύnethyicinnamoylguanidine, trans-3-(l-napthyl)acryloylguanidine,
4-isopropylcinnamoylguanidine,
(3-Bromocinnamoyl)guanidine,
6-methoxy-2-naphthoylguanidine,
5-(N-Methyl-N-isobutyl)amiloride,
3-phenylcinπamoylguanidine,
(2-Chlorocinnamoyl)guanidme,
2*4 DichloroBenzamil HCI,
4-phenylcinnamoylguamdine,
4-(trifluoromethyl)cinnamoylguanidύιe, 492-
3-(trifluoromethoxy)cinnamoylguanidine, 3-(trifluoromethyl)cinπamoylguanidine, 2-ethoxycinnamoylguanidine, cinnamoylguanidine hydrochloride, 4-ethoxycinήamoylguanidine, (2-Bromocinnamoyl)guanidine, 2,6-dichlorocinnamoyIguanidine, 3,4,5-trimethoxycinnamoylguanidine, 5-tert-butylamino-amiloride, 3-t-butylchmamoylguanidine, 5-hromo-2-fluorocinnamoylguanidine, (4-Chlorocinnamoyl)guanidine, 2-t-butylcinnamoylguanidine, 2-cyclohexylcinnamoylguanidine, 6-Iodoamiloride,
3-(trans-hept-l-en-l-yl)cinnamoylguantdine,
Figure imgf000193_0001
(2-Methoxycinnamoyl)guanidine,
[3-(3-Pyridyl)acryloyl]guaιιidinβ,
4-phenylbenzoylguanidine,
2,4i-dichlorocinnamolyguanidine,
(3-Methoxycmnamoyl)guanidme,
2-fluorocinnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
(a-Methylcinnamoyl)guanidine,
5-(3'-brρmcrphenyl)penta-2,4-dienoylguanidine,
(5-Phenyl-penta-2,4-dienoyl)guanidine,
(Quinoline-2-carbonyl)guanidine,
(?henylacetyl)guanidine,
NjN'-Bis^i ino^^thalene^^-dicarboxami e,
6-bromo-2-napthoylguaπidine,
1 -bromo-2-πapmoylguanidine,
2-chloro-6-fluorocinnamoylguanidine,
[(4-ChlorQphenoxy-acetyljguanidine,
Phenamil methanesulfonate salt ,
N-Benzoyl-N'-ciπnamoylguanidine and
N-(2-napthoyl)-N1-phenylguanidine.
154. The method according to claim 151, wherem said Coronavirus is human Coronavirus 229E.
155. The method according to claim 154, wherein said compound is selected from the group consisting of
4-isopropylcinnamoylguanidine,
3,4-dichlorocinnamoylguanidine,
3-(trifluoromethoxy)ciιmamoylguanidine,
4-t-butylcir amoylguanidine,
3-isopropyloinnamoylguanidine hydrochloride,
3-t-butylcinnamoylguanidine,
2-t-butylcinnarøoylguanidine, trans-3-(l-napthyl)acryloylguanidiπe,
5-bromo-2-melhoxycinnamoylguanidine,
2,3-difiuorocinnamoylguani mej
3-(2-napthyl)acryloylguanidine,
2-phenylctnnamoylguanidine,
3-phenylcinnaraoylguanidine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine,
4-phenylbenzoylguaaidine,
3-(trifluoromethyl)cύmamoylguanidine,
(4-Phenoxybenzoyl)guanidine, 4-(trifluorøme!thyl)cinnamoylguanidϊne,
2-(cyclohex-l-en-lyl)citrøamoylguanidine,
(4-Bromocύ amoyl)guanidine,
S-(N,N-hexamethylene)amiloride, l-napthoylguanidine,
5-(4-fluorophenyl)amiloride,
(5-Phaa.yl-penta-2,4-dienoyl)guanidine,
(3-Bromocinnamoyl)guanidine,
2,5-dimethylcinnamoylguanidine,
2-(tritϊuoromethyl)cinnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
(4-CUorocinttamoyl)gπanidine,
(3-Metfaoxycinnamoyl)guanidine,
5-bromo-2-fluorocinnamoyIguanidine9
5-(NJST-Dimethyl)amiloride hydrochloride,
Cinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine,
(a-Methyϊcitmamoyl)guanidine,
4-ρhenylcinnamoylguanidine,
2,6-dichlorocinnamoylguanidine,
(2-Bromocinnamoyl)guanid e,
2,4,6-trimethylcmnamoylguanidine,
(trans-2-Phenylcyclqprc>panecarbonyl)guanidiae,
(3-Chlorocinnamoyl)guanidine,
2-(l-naplhyl)acetoylguanidine,
2-ethylcinnamoylguanidine,
2-cyclohexylc namoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
2-ethoxycinnamoylguanidine,
3-methylcinnamoylguanidine,
2-methylciαnamoylguanidine.
3-fluorocinnamoylguanidine, cinnamoylguanidine hydrochloride,
2,3-dimethylcinnamoylguamdine,
2-fiuotocinnamoylguanidine, -fluorocinnamoylguanidine,
3,4-difluorocinnamoylguanidine,
5-tert-butylamino-amiloride,
2-napthoylguanidine,
NJ^-Bis(amidino)napthalene-2,6-dicarboxamide,
NJ^ -Bis(3-phenylpropanoyl)guanidine,
4-methylcihnamoylguanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
2,3,5,6,-tetramethylcinnamoj guanidine,
3-ethoxycinnamoylguanidine,
N,N'-bis(3phenylpropanoyl)-N"-phenylguanidine, (4-Methoxycinnamoyl)guanidinei,
(2-Chlorocinnamoyl)gu ύdine,
(3-Nitrocinriamoyl)guanidine,
4-ethoxycinnamoylguaniditιe,
3,4,5-tiimethoxycinnamoylguanidine,
2-(2-napthyl)acetoylguamdine and
N-(3-phenylpropanoyl>N'-ρhenylguamdine.
156. The method according to claim 151, wherein said Coronavirus is any one of the known Coronavirus isolates listed in Table 1.
157. The method according to claim 156, wherein said compound is selected from the group consisting of
4-isopropylciπnamoylguanidine, 3,4-dicMorocinnamoylguanidine, 3-(trifiuoιx> ethoxy)c«mamoylguamdine, 4-t-butyldιmamoylguamdine, 3-isopropylcinnamoylguaπidine hydrochloride,
158. The method accordmg to claim 144, wherein said virus is the Hepatitis C virus.
1 9. The method according to claim 158, wherein said membrane ion channel is the Hepatitis C virus p7 membrane ion channel.
160. The method accordmg to claim 159, wherein said compound is selected from the group consisting of
2,3-dimethylcinnamoylguaaidine,
2,4,6-trimethylcinnamoylguanidine,
5-bromcH2-fiυorocinnamoylguanidine,
(4-Bromocinnamoyl)guanidine,
2,5-dimethylcannamoylguanidine,
3-(trifl.uoromethyl)cinnamoylguanidine,
4-(trifluoromethyl)cmnamoylguanidine,
6-methoxy-2-naphthoylguanidine,
(2-Chlorocinnamoyl)guanidine,
(4-Chlorocinnamoyl)guarιidine,
(2-Bromocinnamoyl)guanidine,
2,6-dichlorociπnamoylguamdine,
(3-Bromooinnamoyl)guaπidine, (3-Chlorocinnamoyl)guanidine,
2-(tiifiuoromethyl)ciπnamoylguanidine,
(4-Phenoxybenzoyl)guanidine,
3,4-dichloroc ιnamoylguanidine,
4-isopropylcinnamoylguanidine, trans-3-(l-napthyl)acryIoylguanidine,
4-t-butylcinnamoylguanidine,
2-t-butylcinnamoylguanidine,
2-ethylcinnamoylguanidine,
4-methylcinnamoylguaoidine,
5-bromo-2-methoxycinnamoylguanidine,
3-(trifluoromefhoxy)cinπamoyIguanidin^
2-cyclohexylcinnamoylguaπidine,
I-napthoylguahidine,
3-t-butylcύιnamoylguanidine,
4-phenylbenzoylguanidine,
(5-Phenyl-penta-2,4-dieπoyl)guanid e,
N-(cinnamoyl)-N,phenylguanidine,
3-isopropylcinnamoylguanidine hydrochloride,
Benza il hydrochloride,
N-(3-phenylpropanoyl)-N'-phenylguanidine,
N,N'-bis(3phenylpropanoyl)-N"-phenylguanidine,
3-(2-napthyl)acryloylguanidine,
5-(N-MethyI-N-isobutyl)amiloride,
2'4 DichloroBenzamil HCI,
5 ert-butylamino-amiloride,
5-(N-Ethyl-N-isopκ pyl)amiloride,
(4-Methoxycinnamoyl)guanidine,
4-fluorocinnarøoylguanidine,
(3-Nitrocinnamoyl)guanidine,
4-ethoxycinnamoylguanidine,
(4-Hydroxycinnamoyl)guanidine,
(trans-2-Phenylcycloproρanecarbonyl)guanidine,
3-ethoxycinnamoylguamdine,
2,3,5,6,-tetramethylcinnamoylguanidine,
4-phenylcinnamoylguanidine, trans-3-Furanacιyoylguamdine,
N-(6-Hydroxy-2-napfhoyl)-N-phemylguaιύdine,
(2-Furanacryloyl)guamdine,
3-(cyclohex-l-en-l-yl)cinnamoylguanidine, cinnamoylguanidine hydrochloride,
5-(N,N-hexamefhylene)amiloride,
2,3-difluorocinnamoylguanidine,
2-(l-naρthyl)acetoylguanidine,
(a-Methylcinnamoyl)guanidine,
(2-Nitrocinnamoyl)guanidine,
6-Iodoamiloride,
3,4-(methylenedioxy)cinnamoylguanidine, 497-
2-ethoxycinnamoylguanidine,
Cinnamoylguanidine,
2-phenylc namoylguanidine,
2-(cyclohex-l-en-lyl)cinnamoylguanidine,
2-naptiιoylguanidine,
3-ρhenylcinnamoylguanidine,
5-(N,N-Dimethyl)amiloride hydrochloride,
5-(4-fluorophenyl)amiloride,
(3-Methoxycinnamoyl)guanidine,
2-fl.uorocinnamoylguanidine,
5-(3'-bromophenyl)penta-2,4-dienoylguanidine,
[(4-Chlorophenoxy-acetyl3guanidine,
(3-phenylprppanoyl)guanidύιe,
2-chloro-6-fluαrocinnamoylguanidine,
3-fluorocinnamoylguaniditte,
2-methylcinnamoylguanidine,
(2-Methoxycinnamoyl)guanidine, l-bromo-2-naρthoylguanidine,
3,4,5-trimethoxycinnamoylguamdine,
3-methylcinnamoylguanidine,
3-(trans-hept-l-en-l-yl)chιnamoylguamdine,
Phenamil methanesulfonate salt ,
2,4-dichloτociι∞amolyguanidine,
(4-Nitrocinπamoyl)guaridine,
3,4-difluorocinnamoylguanidine and
[0B)-3-(4-Dimethylaminophenyl)-2- methylacryloyl]guanidine.
161. The method according to any one of claims 144 to 155, wherein said mammal is a primate.
162. The method according to any one of claims 158 to 160, whereύi said mammal is a primate.
163. The method according to olaim 161 or claim 162, wherein said primate is human.
164. An antiviral compound selected from the group consisting of N-(3,5-Diamino-6-chloro-pyrazine-2-carbonyl)-Ns-phenyl-guanidine, N-Benzyl-N'-(3,5-diamino-6-ch ro-pyrazine-2-carbonyl)-guanidine, 3'4 DichloroBenzamil, 498-
2'4 DichloroBenzamil, 5-(N-methyl-N-guanidmocarbonyl-methyl)amiloride,
5-(N-Methyl-N-isobutyl)amiloride,
5-(N-Ethyl-N-isopropyI)amiloride, 5-(N,N-Dimethyl)amiloride hydrochloride,
5-(N,N-hexamethylene)amiloride4
5-(N,N-Diethyl)amiloride hydrochloride,
6-ϊodoamiloride,
Bodipy-FL amiloride, 3-hydroxy-5-hexamethyleneimino-amiloride,
5-(4-fluorophenyl)amiloride,
5-tert-butylamino-amiloride,
N-amidino-3-amino-5-phenyl-6-chloro-2-pyrazinecarboxamide,
3-memoxy-5-(N,N-Hexame1hylene)-an loride, 3-methoxy-amiloride, he« meihyleneimino-6-phenyl-2-pyrazmecarboximide^
N-amidino-3,5-diamn o-6-phenyl-2-ρyrazinecarboxamide,
1 -napthoylguaπidine,
2-napthoylguanidine, N-(2-napthoyl)-N-phenylguamdine,
HN-bis(2-napthoyl)guanidine,
Figure imgf000199_0001
N^Sl,-bis(2-napthoyl)-N"-phenylguanidine,
6-methoxy-2-naphthoylguanidine, 3-quinolinoylguanidine, cinnamoylguanidine,
4-phenylbenzoylguanidine,
N-(ciιmamoyi)-N'phenylguanidine,
(3-phenylρroρanoyl)guanidine, N;JSP-bis-(cύmamoyl)-N"-phenyϊguanidine,
N-(3-phenylpropanoyl)-N'-ρhenylguanidine,
N,N'-bis(3phenylproρanoyl)-N"-phenylguanidine, trans-3-furanacryoylguanidine,
^(β-Hydroxy^-napthoy^-N-phenylguanidine,
(4-Phenoxybenzoyl)guanidine,
N N'-Bis(amidino)napthalene-2,6-dicarboxaιnide, N"-Cinnamoyl-NiJvf,-diphenylguanidine,
(Phenylacetyl)guanidine,
N,N'-Bis(3-phenylpropanoyl)guanidine, benzyoylguanidine,
(4-Chlorophenoxy-acetyl]guanidine, N-benzoyl- -dnnamoylguanidine,
[(E)-3-(4-Dimethylaminophenyl)-2-methylacryloyl]guanidine,
(4-Chlorocinnamoyl)guanidine,
(4"Brcmo nnamoyl)guanidine,
(4-Methoxycinnamoyl)guanidine, (5-Phenyl-ρenta-2,4-dienoyl)guaπidine,
(3-Bromocinnamoyl)guanidine,
(3-Methoxycinnamoyl)guanidine,
(3-Chlorocinnamoyl)guanidine,
(2-Chlorocinnamoyl)guanidine, (2-Bromoc namoyl)guaπidine,
(2-Methoxyriunamoyl)guanidine,
(trans-2-Phenyloyclopropanecarboήyl)guanidine,
[3-(3-PyridyI)acryloyl]guanidme,
(4-Hydroxycinnamoyl)guanidine, (Quinoline-2-carbonyl)guanidine, or pharmaceutically acceptable salts thereof.
A pharmaceutical composition comprising a compound according to claim 164, and optionally one or more pharmaceutical acceptable carriers or derivatives.
166. The pharmaceutical composition according to claim 165, further comprising one or more known antiviral compounds.
167. The method according to any one of claims 6 - 8,12 ,13, 16, 19, 21, 23, 25, ' 27, 29, 31, 32 to 34, 37 to 39, 42, 45, 47, 49, 1, 53, 55, 57, 58 to 60, 3 to
65, 68, 71, 73, 75, 77, 79, 81, 83 to 87, 9 to 92, 4 to 96, 98, 99, 101, 102, 103 to 105, 107 to 110, 112 to 114, 116, 117, 119, 120-122, 124-127, 129-131, 133-134, 136, 137, 139-147, 149-152, 154, 156, 158, ox 159, wherein said compound is selected from the antiviral compounds according to claim 164.
168. The method according to any one of claims 6, 31 ,58,84, 102 or 144, wherein said virus is Dengue vύrus and said compound is selected from the group consisting of cinnamoylguanidine, (2-chlorocinnamoyl)guanidine or trans -3-(l-napthyl)acryloylguamdine.
PCT/AU2004/000866 2003-06-26 2004-06-26 Antiviral acylguanidine compounds and methods WO2004112687A2 (en)

Priority Applications (14)

Application Number Priority Date Filing Date Title
US10/562,296 US20070099968A1 (en) 2004-06-26 2004-06-24 Antiviral compounds and methods
NZ544671A NZ544671A (en) 2003-06-26 2004-06-26 Antiviral compounds and methods
CN2004800240974A CN101111475B (en) 2003-06-26 2004-06-26 Antiviral compounds and methods
CA2529949A CA2529949C (en) 2003-06-26 2004-06-26 Antiviral compounds and methods
AU2004248859A AU2004248859C1 (en) 2003-06-26 2004-06-26 Antiviral acylguanidine compounds and methods
EP13164504.6A EP2617709B8 (en) 2003-06-26 2004-06-26 Guanidine derivatives as antiviral agents
JP2006515560A JP5030587B2 (en) 2003-06-26 2004-06-26 Antiviral acylguanidine compounds and methods
BRPI0411900A BRPI0411900B8 (en) 2003-06-26 2004-06-26 compounds and pharmaceutical compositions comprising them
KR1020057024860A KR101153254B1 (en) 2003-06-26 2004-06-26 Antiviral compounds and methods
EP04737487A EP1646371A4 (en) 2003-06-26 2004-06-26 Antiviral compounds and methods
US13/553,239 US20130035328A1 (en) 2003-06-26 2012-07-19 Antiviral compounds and methods
US14/615,616 US20150313909A1 (en) 2003-06-26 2015-02-06 Antiviral compounds and methods
US15/602,526 US10472332B2 (en) 2003-06-26 2017-05-23 Antiviral compounds and methods
US16/554,990 US11192863B2 (en) 2003-06-26 2019-08-29 Antiviral compounds and methods

Applications Claiming Priority (8)

Application Number Priority Date Filing Date Title
AU2003903251 2003-06-26
AU2003903251A AU2003903251A0 (en) 2003-06-26 2003-06-26 Antiviral compounds and methods
AU2003903850A AU2003903850A0 (en) 2003-07-25 2003-07-25 Anti-coronavirus compounds and methods
AU2003903850 2003-07-25
AU2003904692 2003-08-29
AU2003904692A AU2003904692A0 (en) 2003-08-29 Anti-flavivirus compounds and methods
AU2004902902 2004-05-31
AU2004902902A AU2004902902A0 (en) 2004-05-31 Anti-flavivirus compounds and methods

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US10/562,296 A-371-Of-International US20070099968A1 (en) 2004-06-26 2004-06-24 Antiviral compounds and methods
US13/553,239 Continuation US20130035328A1 (en) 2003-06-26 2012-07-19 Antiviral compounds and methods

Publications (2)

Publication Number Publication Date
WO2004112687A2 true WO2004112687A2 (en) 2004-12-29
WO2004112687A3 WO2004112687A3 (en) 2007-07-12

Family

ID=33545379

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/AU2004/000866 WO2004112687A2 (en) 2003-06-26 2004-06-26 Antiviral acylguanidine compounds and methods

Country Status (8)

Country Link
US (4) US20130035328A1 (en)
EP (2) EP2617709B8 (en)
JP (1) JP5030587B2 (en)
KR (1) KR101153254B1 (en)
BR (1) BRPI0411900B8 (en)
CA (1) CA2529949C (en)
NZ (1) NZ544671A (en)
WO (1) WO2004112687A2 (en)

Cited By (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006135978A1 (en) * 2005-06-24 2006-12-28 Biotron Limited Antiviral compounds and methods
WO2007071400A1 (en) 2005-12-22 2007-06-28 Novartis Ag Pyrazine derivatives as epithelial sodium channel blocker
WO2009018609A1 (en) * 2007-08-03 2009-02-12 Biotrom Limited Hepatitis c antiviral compositions and methods
JP2009520728A (en) * 2005-12-22 2009-05-28 ノバルティス アクチエンゲゼルシャフト Pyrazinoylguanidine compounds useful for the treatment of inflammation or allergic conditions
US7745442B2 (en) * 2003-08-20 2010-06-29 Parion Sciences, Inc. Methods of reducing risk of infection from pathogens
US20110217265A1 (en) * 2008-09-23 2011-09-08 Glenn Jeffrey S Screening for Inhibitors of HCV Amphipathic Helix (AH) Function
AU2013205388B2 (en) * 2005-06-24 2016-05-05 Biotron Limited Antiviral compounds and methods
AU2013219202B2 (en) * 2007-08-03 2016-12-08 Biotron Limited Hepatitis C antiviral compositions and methods
WO2018081863A1 (en) * 2016-11-04 2018-05-11 University Of Wollongong 6-SUBSTITUTED DERIVATIVES OF HEXAMETHYLENE AMILORIDE AS INHIBITORS OF uPA AND USES THEREOF
US11007208B2 (en) 2015-09-16 2021-05-18 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
WO2021154687A1 (en) 2020-01-27 2021-08-05 Gilead Sciences, Inc. Methods for treating sars cov-2 infections
WO2021207049A1 (en) 2020-04-06 2021-10-14 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carbanucleoside analogs
WO2021262826A2 (en) 2020-06-24 2021-12-30 Gilead Sciences, Inc. 1'-cyano nucleoside analogs and uses thereof
US11260070B2 (en) 2017-03-14 2022-03-01 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
EP3960165A1 (en) * 2020-08-25 2022-03-02 Fondation EspeRare Nhe-1 inhibitors for the treatment of coronavirus infections
WO2022046631A1 (en) 2020-08-24 2022-03-03 Gilead Sciences, Inc. Phospholipid compounds and uses thereof
WO2022047065A2 (en) 2020-08-27 2022-03-03 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11266666B2 (en) 2014-10-29 2022-03-08 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US11266681B2 (en) 2017-07-11 2022-03-08 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
WO2022081973A1 (en) 2020-10-16 2022-04-21 Gilead Sciences, Inc. Phospholipid compounds and uses thereof
US11492353B2 (en) 2010-07-22 2022-11-08 Gilead Sciences, Inc. Methods and compounds for treating Paramyxoviridae virus infections
US11491169B2 (en) 2020-05-29 2022-11-08 Gilead Sciences, Inc. Remdesivir treatment methods
WO2022251318A1 (en) 2021-05-26 2022-12-01 Gilead Sciences, Inc. Phospholipid formulations of 1'-cyano substituted carba-nucleoside analogs
WO2023023527A1 (en) 2021-08-18 2023-02-23 Gilead Sciences, Inc. Phospholipid compounds and methods of making and using the same
US11597742B2 (en) 2017-05-01 2023-03-07 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy) (phenoxy) phosphoryl)amino)propanoate
US11613553B2 (en) 2020-03-12 2023-03-28 Gilead Sciences, Inc. Methods of preparing 1′-cyano nucleosides
WO2023168194A1 (en) 2022-03-03 2023-09-07 Gilead Sciences, Inc. Antiviral compounds and methods of making and using the same
WO2023168254A1 (en) 2022-03-03 2023-09-07 Gilead Sciences, Inc. Antiviral compounds and methods of making and using the same
WO2023167938A1 (en) 2022-03-02 2023-09-07 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
WO2023167944A1 (en) 2022-03-02 2023-09-07 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
WO2023239665A1 (en) 2022-06-06 2023-12-14 Gilead Sciences, Inc. Methods for treatment of viral infections including sars-cov-2
WO2024006461A1 (en) 2022-06-30 2024-01-04 Gilead Sciences, Inc. Solid forms of a nucleoside analogue and uses thereof
WO2024006376A1 (en) 2022-06-29 2024-01-04 Gilead Sciences, Inc. Solid forms of a nucleoside analogue and uses thereof
EP4125373A4 (en) * 2020-03-30 2024-04-17 Univ Jefferson Compounds, compositions, and methods for treating, ameliorating, or preventing viral infections

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004112687A2 (en) 2003-06-26 2004-12-29 Biotron Limited Antiviral acylguanidine compounds and methods
WO2022040321A1 (en) * 2020-08-19 2022-02-24 Natreon, Inc. Protection against coronavirus infection by extracts and extract components
WO2023012329A1 (en) 2021-08-06 2023-02-09 Intervet International B.V. Method of treating veterinary viral diseases
WO2023167055A1 (en) * 2022-03-03 2023-09-07 日本曹達株式会社 Antiviral agent

Family Cites Families (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US2734904A (en) * 1956-02-14 Xcxnhxc-nh
NL299931A (en) * 1962-10-30
FR1435379A (en) * 1964-07-30 1966-04-15 American Cyanamid Co Process for stabilizing rigid polyvinyl chloride against the effects of light
US3527758A (en) * 1967-04-13 1970-09-08 Merck & Co Inc Process for the preparation of pyrazinoylguanidines from a pyrazinoic azide and a guanidine
DE2006186A1 (en) 1970-02-11 1971-08-19 Huebner Vamag Method for inserting a seat ring for the shut-off element of a valve
DE2531343A1 (en) * 1975-07-14 1977-02-10 Henkel & Cie Gmbh Antimicrobial compsns. contg. substd. benzoyl phenyl guanidines - prepd. from opt. substd. benzoyl cyanamides and substd. anilines
DD200618A1 (en) * 1981-10-05 1983-05-25 Manfred Augustin PROCESS FOR THE PREPARATION OF NEW N-AROYL AND HETAROYLIMIDES
US4496573A (en) * 1982-08-24 1985-01-29 William H. Rorer, Inc. 1-Pyridylmethyl-3-acyl guanidines
US4894376A (en) * 1988-02-26 1990-01-16 Trustees Of The University Of Pennsylvania Methods of treating diseases characterized by hyperexcitability of neurons
JPH0789859A (en) * 1990-04-05 1995-04-04 Celtrix Pharmaceut Inc Pharmaceutic composition containing pyrazine derivative and curing method for inhibiting neogenesis of new blood vessel
IN177137B (en) * 1992-12-11 1996-11-16 Hoechst India
DE4301739A1 (en) * 1993-01-22 1994-07-28 Sauer Professor Dr Gerhard Treatment of primary tumours and viral diseases
JPH0725768A (en) * 1993-07-09 1995-01-27 Mitsubishi Chem Corp Suppressing agent for hyperplasia of vascular endosporium
DE4325822A1 (en) * 1993-07-31 1995-02-02 Hoechst Ag Substituted benzoylguanidines, process for their preparation, their use as medicament or diagnostic agent, and medicament containing them
TW415937B (en) * 1994-01-25 2000-12-21 Hoechst Ag Phenyl-substituted alkylcarboxylic acid guanidides bearing perfluoroalkyl groups, process for their preparation, their use as a medicament or diagnostic, and medicament containing them
DE4415873A1 (en) * 1994-05-05 1995-11-09 Hoechst Ag Substituted bicyclic heteroaroylguanidines, process for their preparation, their use as medicament or diagnostic agent and medicament containing them
DE4421536A1 (en) * 1994-06-20 1995-12-21 Hoechst Ag Phenyl-substituted alkenylcarboxylic acid guanidines bearing perfluoroalkyl groups, processes for their preparation, their use as medicaments or diagnostic agents, and medicaments containing them
JPH08225513A (en) * 1994-12-21 1996-09-03 Kanebo Ltd Naphthoylguanidine derivative
AT408330B (en) 1995-01-27 2001-10-25 Colop Stempelerzeugung Skopek SELF-COLORING STAMP
ATE183498T1 (en) * 1995-04-18 1999-09-15 Hoechst Ag SUBSTITUTED INDENOYLGUANIDINES WITH ANTIARRHYTHMIC AND CARDIOPROTECTIVE EFFECTS
DE19518796A1 (en) * 1995-05-22 1996-11-28 Hoechst Ag Fluorophenyl-substituted alkenylcarboxylic acid guanidines, processes for their preparation, their use as medicaments or diagnostic agents and medicaments containing them
DE19527305A1 (en) * 1995-07-26 1997-01-30 Hoechst Ag Substituted cinnamic acid guanidides, process for their preparation, their use as a medicament or diagnostic agent and medicament containing them
US5628984A (en) 1995-07-31 1997-05-13 University Of North Carolina At Chapel Hill Method of detecting lung disease
JPH0967332A (en) * 1995-08-29 1997-03-11 Yamanouchi Pharmaceut Co Ltd N-(2-propenoyl)guanidine derivative
DE19621482A1 (en) * 1996-05-29 1997-12-04 Hoechst Ag Substituted 1-naphthoylguanidines, processes for their preparation, their use as medicaments or diagnostic agents and medicaments containing them
DE19622222A1 (en) * 1996-06-03 1997-12-04 Hoechst Ag Use of sodium=proton exchange inhibitor
AUPO545297A0 (en) * 1997-03-04 1997-03-27 Fujisawa Pharmaceutical Co., Ltd. Guanidine derivatives
IL126276A0 (en) * 1997-09-24 1999-05-09 Hoechst Marion Roussel De Gmbh The use of an inhibitor of the na+/H+ exchanger for the production of a medicament for the treatment or prophylaxis of disorders of the central nervous system
US7041702B1 (en) * 1997-10-21 2006-05-09 Scion Pharmaceuticals, Inc. Pharmaceutically active compounds and methods of use
KR20010031065A (en) * 1997-10-21 2001-04-16 케임브리지 뉴로사이언스, 인코포레이티드 Pharmaceutically active compounds and methods of use
JP2002508507A (en) * 1997-12-18 2002-03-19 セプラコア インコーポレーテッド A method for simultaneous identification of novel biological targets and drug discovery lead structures
US6011059A (en) * 1997-12-24 2000-01-04 Bristol-Myers Squibb Company Acyl guanidine sodium/proton exchange inhibitors and method
AUPP646498A0 (en) * 1998-10-12 1998-11-05 Australian National University, The A method of modulating ion channel functional activity
DE19849722A1 (en) * 1998-10-28 2000-05-04 Aventis Pharma Gmbh Substituted phenyl-alkenoylguanidines, processes for their preparation, their use as medicaments or diagnostic agents and medicaments containing them
DE19859727A1 (en) * 1998-12-23 2000-06-29 Aventis Pharma Gmbh The use of inhibitors of the sodium-hydrogen exchanger for the manufacture of a medicament for the prevention of age-related organ dysfunctions, age-related illnesses for the prolongation of life
AUPQ223999A0 (en) * 1999-08-16 1999-09-09 University Of Sydney, The Intracellular feedback controls in the diagnosis and treatment of human disease
AUPS022802A0 (en) 2002-01-31 2002-02-21 Macfarlane Burnet Institute For Medical Research And Public Health Limited, The Anti-viral compounds
WO2004112687A2 (en) 2003-06-26 2004-12-29 Biotron Limited Antiviral acylguanidine compounds and methods
DK1902017T3 (en) * 2005-06-24 2014-06-30 Biotron Ltd Antiviral acylguanidine compounds

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP1646371A4 *

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7745442B2 (en) * 2003-08-20 2010-06-29 Parion Sciences, Inc. Methods of reducing risk of infection from pathogens
AU2013205388B2 (en) * 2005-06-24 2016-05-05 Biotron Limited Antiviral compounds and methods
JP2008543886A (en) * 2005-06-24 2008-12-04 バイオトロン リミテッド Antiviral compounds and methods
US9440926B2 (en) 2005-06-24 2016-09-13 Biotron Limited Antiviral compounds and methods
US10683263B2 (en) 2005-06-24 2020-06-16 Biotron Limited Antiviral compounds and methods
AU2006261593B2 (en) * 2005-06-24 2013-01-24 Biotron Limited Antiviral compounds and methods
JP2013049676A (en) * 2005-06-24 2013-03-14 Biotron Ltd Antiviral compound and method
CN101208297B (en) * 2005-06-24 2013-04-24 拜伊特朗有限公司 Antiviral compounds and methods
WO2006135978A1 (en) * 2005-06-24 2006-12-28 Biotron Limited Antiviral compounds and methods
EP2826770A1 (en) 2005-06-24 2015-01-21 Biotron Limited Acylguanidine compounds with antiviral activity
US8669280B2 (en) 2005-06-24 2014-03-11 Biotron Limited Antiviral compounds and methods
JP2009520728A (en) * 2005-12-22 2009-05-28 ノバルティス アクチエンゲゼルシャフト Pyrazinoylguanidine compounds useful for the treatment of inflammation or allergic conditions
WO2007071400A1 (en) 2005-12-22 2007-06-28 Novartis Ag Pyrazine derivatives as epithelial sodium channel blocker
AU2008286240B2 (en) * 2007-08-03 2013-05-23 Biotron Limited Hepatitis C antiviral compositions and methods
EP2659885A1 (en) * 2007-08-03 2013-11-06 Biotron Limited Hepatitis C Antiviral Compositions and Methods
AU2013219202B2 (en) * 2007-08-03 2016-12-08 Biotron Limited Hepatitis C antiviral compositions and methods
WO2009018609A1 (en) * 2007-08-03 2009-02-12 Biotrom Limited Hepatitis c antiviral compositions and methods
US20110217265A1 (en) * 2008-09-23 2011-09-08 Glenn Jeffrey S Screening for Inhibitors of HCV Amphipathic Helix (AH) Function
US11492353B2 (en) 2010-07-22 2022-11-08 Gilead Sciences, Inc. Methods and compounds for treating Paramyxoviridae virus infections
US11266666B2 (en) 2014-10-29 2022-03-08 Gilead Sciences, Inc. Methods for treating Filoviridae virus infections
US11344565B2 (en) 2014-10-29 2022-05-31 Gilead Sciences, Inc. Methods for the preparation of ribosides
US11382926B2 (en) 2015-09-16 2022-07-12 Gilead Sciences, Inc. Methods for treating Arenaviridae and Coronaviridae virus infections
US11007208B2 (en) 2015-09-16 2021-05-18 Gilead Sciences, Inc. Methods for treating arenaviridae and coronaviridae virus infections
WO2018081863A1 (en) * 2016-11-04 2018-05-11 University Of Wollongong 6-SUBSTITUTED DERIVATIVES OF HEXAMETHYLENE AMILORIDE AS INHIBITORS OF uPA AND USES THEREOF
US11260070B2 (en) 2017-03-14 2022-03-01 Gilead Sciences, Inc. Methods of treating feline coronavirus infections
US11597742B2 (en) 2017-05-01 2023-03-07 Gilead Sciences, Inc. Crystalline forms of (S)-2-ethylbutyl 2-(((S)-(((2R,3S,4R,5R)-5-(4-aminopyrrolo[2,1-f] [1,2,4]triazin-7-yl)-5-cyano-3,4-dihydroxytetrahydrofuran-2-yl)methoxy) (phenoxy) phosphoryl)amino)propanoate
US11266681B2 (en) 2017-07-11 2022-03-08 Gilead Sciences, Inc. Compositions comprising an RNA polymerase inhibitor and cyclodextrin for treating viral infections
US11660307B2 (en) 2020-01-27 2023-05-30 Gilead Sciences, Inc. Methods for treating SARS CoV-2 infections
WO2021154687A1 (en) 2020-01-27 2021-08-05 Gilead Sciences, Inc. Methods for treating sars cov-2 infections
US11613553B2 (en) 2020-03-12 2023-03-28 Gilead Sciences, Inc. Methods of preparing 1′-cyano nucleosides
EP4125373A4 (en) * 2020-03-30 2024-04-17 Univ Jefferson Compounds, compositions, and methods for treating, ameliorating, or preventing viral infections
US11701372B2 (en) 2020-04-06 2023-07-18 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carba-nucleoside analogs
WO2021207049A1 (en) 2020-04-06 2021-10-14 Gilead Sciences, Inc. Inhalation formulations of 1'-cyano substituted carbanucleoside analogs
US11903953B2 (en) 2020-05-29 2024-02-20 Gilead Sciences, Inc. Remdesivir treatment methods
US11491169B2 (en) 2020-05-29 2022-11-08 Gilead Sciences, Inc. Remdesivir treatment methods
WO2021262826A2 (en) 2020-06-24 2021-12-30 Gilead Sciences, Inc. 1'-cyano nucleoside analogs and uses thereof
US11939347B2 (en) 2020-06-24 2024-03-26 Gilead Sciences, Inc. 1′-cyano nucleoside analogs and uses thereof
WO2022046631A1 (en) 2020-08-24 2022-03-03 Gilead Sciences, Inc. Phospholipid compounds and uses thereof
EP3960165A1 (en) * 2020-08-25 2022-03-02 Fondation EspeRare Nhe-1 inhibitors for the treatment of coronavirus infections
WO2022043343A1 (en) * 2020-08-25 2022-03-03 Fondation Esperare Nhe-1 inhibitors for the treatment of coronavirus infections
US11814406B2 (en) 2020-08-27 2023-11-14 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11926645B2 (en) 2020-08-27 2024-03-12 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
WO2022047065A2 (en) 2020-08-27 2022-03-03 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
WO2022081973A1 (en) 2020-10-16 2022-04-21 Gilead Sciences, Inc. Phospholipid compounds and uses thereof
WO2022251318A1 (en) 2021-05-26 2022-12-01 Gilead Sciences, Inc. Phospholipid formulations of 1'-cyano substituted carba-nucleoside analogs
WO2023023527A1 (en) 2021-08-18 2023-02-23 Gilead Sciences, Inc. Phospholipid compounds and methods of making and using the same
WO2023167944A1 (en) 2022-03-02 2023-09-07 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
US11780844B2 (en) 2022-03-02 2023-10-10 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
WO2023167938A1 (en) 2022-03-02 2023-09-07 Gilead Sciences, Inc. Compounds and methods for treatment of viral infections
WO2023168254A1 (en) 2022-03-03 2023-09-07 Gilead Sciences, Inc. Antiviral compounds and methods of making and using the same
WO2023168194A1 (en) 2022-03-03 2023-09-07 Gilead Sciences, Inc. Antiviral compounds and methods of making and using the same
WO2023239665A1 (en) 2022-06-06 2023-12-14 Gilead Sciences, Inc. Methods for treatment of viral infections including sars-cov-2
WO2024006376A1 (en) 2022-06-29 2024-01-04 Gilead Sciences, Inc. Solid forms of a nucleoside analogue and uses thereof
WO2024006461A1 (en) 2022-06-30 2024-01-04 Gilead Sciences, Inc. Solid forms of a nucleoside analogue and uses thereof

Also Published As

Publication number Publication date
US11192863B2 (en) 2021-12-07
US20170260147A1 (en) 2017-09-14
CA2529949A1 (en) 2004-12-29
EP1646371A2 (en) 2006-04-19
KR101153254B1 (en) 2012-07-02
NZ544671A (en) 2009-02-28
BRPI0411900B1 (en) 2018-11-21
EP1646371A4 (en) 2010-09-22
EP2617709A1 (en) 2013-07-24
WO2004112687A3 (en) 2007-07-12
BRPI0411900A (en) 2006-09-19
EP2617709B8 (en) 2022-12-21
US20190389816A1 (en) 2019-12-26
KR20060103086A (en) 2006-09-28
JP5030587B2 (en) 2012-09-19
BRPI0411900B8 (en) 2021-05-25
US20150313909A1 (en) 2015-11-05
JP2007508234A (en) 2007-04-05
EP2617709B1 (en) 2022-11-16
US20130035328A1 (en) 2013-02-07
US10472332B2 (en) 2019-11-12
CA2529949C (en) 2013-08-06

Similar Documents

Publication Publication Date Title
US11192863B2 (en) Antiviral compounds and methods
ES2339306T3 (en) USEFUL GUANILHYDRAZONES IN THE TREATMENT OF DISEASES ASSOCIATED WITH THE ACTIVATION OF CELLS T.
CN102858157A (en) Combination pharmaceutical agents as inhibitors of hcv replication
CN102958525B (en) Antiviral compounds
US11357771B2 (en) Methods of preventing or treating flavivirus virus infections and methods of inhibiting the entry of flvivirus, enterovirus or lentivirus into host cells
KR20190093600A (en) Antibacterial peptide
US20070099968A1 (en) Antiviral compounds and methods
US11135219B2 (en) Methods of treating or preventing Zika virus infection
AU2004248859B2 (en) Antiviral acylguanidine compounds and methods
JP2009242247A (en) Human immunodeficiency virus infection inhibitor and aids-treating or preventing drug
CN101111475B (en) Antiviral compounds and methods
KR20200012894A (en) Peptides and Uses thereof as Antiviral Agents
US20070161550A1 (en) Antiviral compositions which inhibit paramyxovirus infection
EP0684818B1 (en) Method of reversing resistance of hiv-1 strains to zidovudine
US11331371B2 (en) Labyrinthopeptins as anti-viral agents
US20230086360A1 (en) Antimicrobial Peptides
WO2018189566A1 (en) Use of a chemical compound as a therapeutic agent
WO2024040156A2 (en) Novel cyclic gamma aapeptide pan-coronavirus inhibitor and method of treating coronavirus infection
KR20110100759A (en) Peptide compound having an inhibition activity for orientia tsutsugamushi infection or proliferation and pharmaceutical composition for treating scrub typhus using the same
CN115710303A (en) Polypeptide derivative for resisting H1N1 influenza virus and application
MXPA99007077A (en) Quinoxaline in triple combination with protease inhibitors and reverse transcriptase inhibitors as medicines for treating aids
EP0879055A1 (en) Inhibition of viral replication

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 200480024097.4

Country of ref document: CN

AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BW BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE EG 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 NA NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2529949

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 2006515560

Country of ref document: JP

WWE Wipo information: entry into national phase

Ref document number: 2007099968

Country of ref document: US

Ref document number: 10562296

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 1020057024860

Country of ref document: KR

WWE Wipo information: entry into national phase

Ref document number: 2004248859

Country of ref document: AU

Ref document number: 544671

Country of ref document: NZ

WWE Wipo information: entry into national phase

Ref document number: 159/KOLNP/2006

Country of ref document: IN

Ref document number: 00159/KOLNP/2006

Country of ref document: IN

WWE Wipo information: entry into national phase

Ref document number: 2006/00650

Country of ref document: ZA

Ref document number: 200600650

Country of ref document: ZA

WWE Wipo information: entry into national phase

Ref document number: 2004737487

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2004248859

Country of ref document: AU

Date of ref document: 20040626

Kind code of ref document: A

WWP Wipo information: published in national office

Ref document number: 2004248859

Country of ref document: AU

WWP Wipo information: published in national office

Ref document number: 2004737487

Country of ref document: EP

DPEN Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed from 20040101)
ENP Entry into the national phase

Ref document number: PI0411900

Country of ref document: BR

WWP Wipo information: published in national office

Ref document number: 10562296

Country of ref document: US