EP3681508A1 - Verfahren und zusammensetzung zur behandlung von virusinfektionen - Google Patents

Verfahren und zusammensetzung zur behandlung von virusinfektionen

Info

Publication number
EP3681508A1
EP3681508A1 EP18857189.7A EP18857189A EP3681508A1 EP 3681508 A1 EP3681508 A1 EP 3681508A1 EP 18857189 A EP18857189 A EP 18857189A EP 3681508 A1 EP3681508 A1 EP 3681508A1
Authority
EP
European Patent Office
Prior art keywords
infection
acid
subject
antiviral composition
oleandrin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP18857189.7A
Other languages
English (en)
French (fr)
Other versions
EP3681508A4 (de
Inventor
Robert A. Newman
Otis C. Addington
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Phoenix Biotechnology Inc
Original Assignee
Phoenix Biotechnology Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PCT/US2017/051553 external-priority patent/WO2018053123A1/en
Application filed by Phoenix Biotechnology Inc filed Critical Phoenix Biotechnology Inc
Publication of EP3681508A1 publication Critical patent/EP3681508A1/de
Publication of EP3681508A4 publication Critical patent/EP3681508A4/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/56Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids
    • A61K31/58Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin
    • A61K31/585Compounds containing cyclopenta[a]hydrophenanthrene ring systems; Derivatives thereof, e.g. steroids containing heterocyclic rings, e.g. danazol, stanozolol, pancuronium or digitogenin containing lactone rings, e.g. oxandrolone, bufalin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/24Apocynaceae (Dogbane family), e.g. plumeria or periwinkle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/22Heterocyclic compounds, e.g. ascorbic acid, tocopherol or pyrrolidones
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/44Oils, fats or waxes according to two or more groups of A61K47/02-A61K47/42; Natural or modified natural oils, fats or waxes, e.g. castor oil, polyethoxylated castor oil, montan wax, lignite, shellac, rosin, beeswax or lanolin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0014Skin, i.e. galenical aspects of topical compositions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/107Emulsions ; Emulsion preconcentrates; Micelles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2009Inorganic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/2031Organic macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyethylene glycol, polyethylene oxide, poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4816Wall or shell material
    • A61K9/4825Proteins, e.g. gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4858Organic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4841Filling excipients; Inactive ingredients
    • A61K9/4866Organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/4891Coated capsules; Multilayered drug free capsule shells
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2236/00Isolation or extraction methods of medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicine
    • A61K2236/30Extraction of the material
    • A61K2236/37Extraction at elevated pressure or temperature, e.g. pressurized solvent extraction [PSE], supercritical carbon dioxide extraction or subcritical water extraction
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • 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

  • Nerium oleander a member of the Nerium species, is an ornamental plant widely distributed in subtropical Asia, the southeastern United States, and the Mediterranean. Its medical and toxicological properties have long been recognized. It has been proposed for use, for example, in the treatment of hemorrhoids, ulcers, leprosy, snake bites, cancers, tumors, neurological disorders, cell-proliferative diseases.
  • ANVIRZELTM (US 5,135,745 to Ozel), which is commercially available, contains the concentrated form or powdered form of the hot-water extract of Nerium oleander .Muller et al. (Pharmazie. (1991) Sept. 46(9), 657-663) disclose the results regarding the analysis of a water extract of Nerium oleander. They report that the polysaccharide present is primarily galacturonic acid. Other saccharides include rhamnose, arabinose and galactose. Polysaccharide content and individual sugar composition of polysaccharides within the hot water extract of Nerium oleander have also been reported by Newman et al. (J.
  • U.S. Patent No. 5,869,060 to Selvaraj et al. pertains to extracts of Nerium species and methods of production. To prepare the extract, plant material is placed in water and boiled. The crude extract is then separated from the plant matter and sterilized by filtration. The resultant extract can then be lyophilized to produce a powder.
  • U.S. Patent No. 6,565,897 U.S. Pregrant Publication No. 20020114852 and PCT International Publication No. WO 2000/016793 to Selvaraj et al. discloses a hot-water extraction process for the preparation of a substantially sterile extract.
  • Triterpenes are known to possess a wide variety of therapeutic activities. Some of the known triterpenes include oleanolic acid, ursolic acid, betulinic acid, bardoxolone, maslinic acid, and others. The therapeutic activity of the triterpenes has primarily been evaluated individually rather than as combinations of triterpenes.
  • Oleanolic acid is in a class of triterpenoids typified by compounds such as bardoxolone which have been shown to be potent activators of the innate cellular phase 2 detoxifying pathway, in which activation of the transcription factor Nrf2 leads to transcriptional increases in programs of downstream antioxidant genes containing the antioxidant transcriptional response element (ARE).
  • Bardoxolone itself has been extensively investigated in clinical trials in inflammatory conditions; however, a Phase 3 clinical trial in chronic kidney disease was terminated due to adverse events that may have been related to known cellular toxicities of certain triterpenoids including bardoxolone at elevated concentrations.
  • compositions containing triterpenes in combination with other therapeutic components are found as plant extracts.
  • Fumiko et al. (Biol. Pharm. Bull (2002), 25(11), 1485-1487) discloses the evaluation of a methanolic extract of Rosmarimus officinalis L. for treating trypanosomiasis.
  • Addington et al. (US 8481086, US 9220778, US 9358293, US 20160243143 Al) disclose a supercritical fluid extract (SCF; PBI-05204) of Nerium oleander containing oleandrin and triterpenes for the treatment of neurological conditions.
  • SCF supercritical fluid extract
  • Oleanolic acid (O or OA), ursolic acid (U or UA) and betulinic acid (B or BA) are the three major triterpene components found in PBI-05204 (PBI-23; a supercritical fluid extract of Nerium oleander) and PBI-04711 (a triterpene-containing fraction 0-4 of PBI- 05204).
  • PBI-23 a supercritical fluid extract of Nerium oleander
  • PBI-04711 a triterpene-containing fraction 0-4 of PBI- 05204.
  • Extracts of Nerium species are known to contain many different classes of compounds: cardiac glycosides, gly cones, steroids, triterpenes, polysaccharides and others.
  • Specific compounds include oleandrin; neritaloside; odoroside; oleanolic acid; ursolic acid; betulinic acid; oleandrigenin; oleaside A; betulin (urs-12-ene-3p,28-diol); 28-norurs-12-en- 3 ⁇ - ⁇ 1; urs-12-en-3p-ol; 3p,3p-hydroxy-12-oleanen-28-oic acid; 3p,20a-dihydroxyurs-21- en-38-oic acid; 3p,27-dihydroxy-12-ursen-38-oic acid; 3p,13p-dihydroxyurs-l l-en-28-oic acid; 3p,12a-dihydroxyoleanan-28,13p-olide; 3p,
  • Ebolavirus infection i.e. filovirus infections.
  • EBOV Epoval virus
  • MARV Marburg virus infections.
  • Five species of Ebolavirus have been identified: Tai Forest (formerly Ivory Coast), Sudan, Zaire, Reston and Bundibugyo.
  • the Flaviviruses are positive, single-stranded, enveloped RNA viruses. They are found in arthropods, primarily ticks and mosquitoes, and cause widespread morbidity and mortality throughout the world. Some of the mosquito-transmitted viruses include Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, and Zikavirus. Some of the tick-transmitted viral infections include Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Omsk Hemorrhagic Fever. Although not a hemorrhagic infection, Powassan virus is a Flavivirus.
  • Oleandrin has demonstrated anti-HIV activity but has not been evaluated against many viruses.
  • the triterpenes oleanolic acid, betulinic acid and ursolic acid have been reported to exhibit differing levels of antiviral activity but have not been evaluated against many viruses.
  • Betulinic acid has demonstrated some anti-viral activity against HSV-1 strain 1C, influenza A H7N1, ECHO 6, and HIV-1.
  • Oleanolic acid has demonstrated some antiviral activity against HIV-1, HEP C, and HCV H strain NS5B.
  • Ursolic acid has demonstrated some anti-viral activity against HIV-1, HEP C, HCV H strain NS5B, HSV-1, HSV-2, ADV-3, ADV-8, ADV-11, HEP B, ENTV CVB1 and ENTV EV71.
  • the antiviral activity of oleandrin, oleanolic acid, ursolic acid and betulinic acid is unpredictable as far as efficacy against specific viruses.
  • Barrows et al. (“A screen of FDA-approved drugs for inhibitors of Zikavirus infection” in Cell Host Microbe (2016), 20, 259-270) report that digoxin demonstrates antiviral activity against Zikavirus but the doses are too high and likely toxic.
  • Cheung et al. (“Antiviral activity of lanatoside C against dengue virus infection” in Antiviral Res. (2014) 111, 93-99) report that lanatoside C demonstrates antiviral activity against Dengue virus.
  • the viral infection is caused by any of the following virus families: Arenaviridae, Bunyaviridae, Filoviridae, Flaviviridae, Paramyxoviridae, or Togaviridae.
  • Some embodiments of the invention are directed to compositions for and methods of treating viral infections from viruses of the Henipavirus genus, Ebolavirus genus, Flavivirus genus, Marburgvirus genus, or Alphavirus genus.
  • One aspect of the invention provides a method of treating viral infection in a subject by chronic administration to the subject of an antiviral composition.
  • the subject is treated by chronically administering to the subject a therapeutically effective amount (therapeutically relevant dose) of the composition, thereby providing relief of symptoms associated with the viral infection or amelioration of the viral infection.
  • Administration of the composition to the subject can begin immediately after infection or any time within one day to 5 days after infection or at the earliest time after definite diagnosis of infection with virus.
  • the invention also provides a method of treating viral infection in a mammal, the method comprising administering to the mammal one or more therapeutically effective doses of the antiviral composition.
  • One or more doses are administered on a daily, weekly or monthly basis.
  • One or more doses per day can be administered.
  • the invention also provides a method of treating viral infection in a subject in need thereof, the method comprising:
  • the subject's clinical response and/or therapeutic response is adequate, then continuing treatment with antiviral composition as needed until the desired clinical endpoint is achieved; or if the subject's clinical response and/or therapeutic response are inadequate at the initial dose and initial dosing regimen, then escalating or deescalating the dose until the desired clinical response and/or therapeutic response in the subject is achieved.
  • Treatment of the subject with antiviral composition is continued as needed.
  • the dose or dosing regimen can be adjusted as needed until the patient reaches the desired clinical endpoint(s) such as a reduction or alleviation of specific symptoms associated with the viral infecion. Determination of the adequacy of clinical response and/or therapeutic response can be conducted by a clinician familiar with viral infections.
  • the antiviral composition can be administered chronically, i.e. on a recurring basis, such as daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, weekly, every other week, every second week, every third week, monthly, bimonthly, semi-monthly, every other month every second month, quarterly, every other quarter, trimesterly, seasonally, semi-annually and/or annually.
  • the treatment period one or more weeks, one or more months, one or more quarters and/or one or more years.
  • An effective dose of cardiac glycoside is administered one or more times in a day;
  • the subject is administered 140 microg to 315 microg per day of cardiac glycoside.
  • a dose comprises 20 microg to 750 microg, 12 microg to 300 microg, or 12 microg to 120 microg of cardiac glycoside.
  • the daily dose of cardiac glycoside can range from 20 microg to 750 microg, 0.01 microg to 100 mg, or 0.01 microg to 100 microg of cardiac glycoside/day.
  • the recommended daily dose of oleandrin, present in the SCF extract is generally about 0.25 to about 50 microg twice daily or about 0.9 to 5 microg twice daily or about every 12 hours.
  • the dose can be about 0.5 to about 100 microg/day, about 1 to about 80 microg/day, about 1.5 to about 60 microg/day, about 1.8 to about 60 microg/day, about 1.8 to about 40 microg/day.
  • the maximum tolerated dose can be about 100 microg/day, about 80 microg/day, about 60 microg/day, about 40 microg/day, about 38.4 microg/day or about 30 microg/day of oleander extract containing oleandrin and the minimum effective dose can be about 0.5 microg/day, about 1 microg/day, about 1.5 microg/day, about 1.8 microg/day, about 2 microg/day, or about 5 microg/day.
  • Suitable doses comprising cardiac glycoside and triterpene can be about 0.05-0.5 mg/kg/day, 0.05-0.35 mg/kg/day, 0.05-0.22 mg/kg/day, 0.05-0.4 mg/kg/day, 0.05-0.3 mg/kg/day, 0.05-0.5 microg/kg/day, 0.05-0.35 microg/kg/day, 0.05-0.22 microg/kg/day, 0.05-0.4 microg/kg/day, or 0.05-0.3 microg/kg/day.
  • the antiviral composition can be administered systemically. Modes of systemic administration include parenteral, buccal, enteral, intramuscular, subdermal, sublingual, peroral, or oral. The composition can also be administered via injection or intravenously.
  • the cardiac glycoside is preferabley oleandrin but can also include odoroside, neritaloside, or oleandrigenin.
  • the composition further comprises: a) one or more triterpenes; b) one or more steroids; c) one or more triterpene derivatives; d) one or more steroid derivatives; or e) a combination thereof.
  • the composition comprises cardiac glycoside and: a) two or three triterpenes; b) two or three triterpene derivatives; c) two or three triterpene salts; or d) a combination thereof.
  • the triterpene is selected from the group consisting of oleanolic acid, ursolic acid, betulinic acid and salts or derivatives thereof.
  • a pharmaceutical composition comprises at least one pharmaceutical excipient and the antiviral composition.
  • the antiviral composition comprises: a) at least one cardiac glycoside and at least one triterpene; b) at least one cardiac glycoside and at least two triterpenes; c) at least one cardiac glycoside and at least three triterpenes; d) at least two triterpenes and excludes cardiac glycoside; e) at least three triterpenes and excludes cardiac glycoside; or f) at least one cardiac glycoside, e.g. oleandrin.
  • the generic terms triterpene and cardiac glycoside also encompass salts and derivatives thereof, unless otherwise specified.
  • the extract comprises one or more cardiac glycosides and one or more cardiac glycoside precursors (such as cardenolides, cardadienolides and cardatrienolides, all of which are the aglycone constituents of cardiac glycosides, for example, digitoxin, acetyl digitoxins, digitoxigenin, digoxin, acetyl digoxins, digoxigenin, medigoxin, strophanthins, cymarine, ouabain, or strophanthidin).
  • the extract may further comprise one or more glycone constituents of cardiac glycosides (such as glucoside, fructoside, and/or glucuronide) as cardiac glycoside presursors.
  • the antiviral composition may comprise one or more cardiac glycosides and two more cardiac glycoside precursors selected from the group consisting of one or more aglycone constituents, and one or more glycone constituents.
  • a composition containing oleandrin (OL), oleanolic acid (OA), ursolic acid (UA) and betulinic acid (BA) is more efficacious than pure oleandrin, when equivalent doses based upon oleandrin content are compared.
  • the molar ratios of the individual triterpenes to oleandrin range as follows: 2-8 (OA) : 2-8 (UA) : 0.1-1 (BA) : 0.5-1.5 (OL); or 3-6 (OA) : 3-6 (UA) : 0.3-8 (BA) : 0.7-1.2 (OL); or 4-5 (OA) : 4-5 (UA) : 0.4-0.7 (BA) : 0.9-1.1 (OL); or 4.6 (OA) : 4.4 (UA) : 0.6 (BA) : 1 (OL).
  • the other therapeutic agent is not a polysaccharide obtained during preparation of the extract, meaning it is not an acidic homopolygalacturonan or arabinogalaturonan.
  • the extract excludes another therapeutic agent and/or excludes an acidic homopolygalacturonan or arabinogalaturonan obtained during preparation of the extract.
  • the invention also provides use of a cardiac glycoside in the manufacture of a medicament for the treatment of viral infection in a subject.
  • the manufacture of such a medicament comprises: providing one or more antiviral compounds of the invention; including a dose of antiviral compound(s) in a pharmaceutical dosage form; and packaging the pharmaceutical dosage form.
  • the manufacture can be conducted as described in PCT International Application No. PCT/US06/29061.
  • the pharmaceutical composition can further comprise a combination of at least one material selected from the group consisting of a water soluble (miscible) co-solvent, a water insoluble (immiscible) co-solvent, a surfactant, an antioxidant, a chelating agent, and an absorption enhancer.
  • the solubilizer is at least a single surfactant, but it can also be a combination of materials such as a combination of: a) surfactant and water miscible solvent; b) surfactant and water immiscible solvent; c) surfactant, antioxidant; d) surfactant, antioxidant, and water miscible solvent; e) surfactant, antioxidant, and water immiscible solvent; f) surfactant, water miscible solvent, and water immiscible solvent; or g) surfactant, antioxidant, water miscible solvent, and water immiscible solvent.
  • the pharmaceutical composition optionally further comprises: a) at least one liquid carrier; b) at least one emulsifying agent; c) at least one solubilizing agent; d) at least one dispersing agent; e) at least one other excipient; or f) a combination thereof.
  • FIGS. 1-2 depict charts summarizing the in vitro dose response antiviral activity of various compositions against Ebolavirus.
  • FIG. 7 depicts a chart summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against Ebolavirus in Vero E6 cells.
  • FIG. 8 depicts a chart summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against Marburgvirus in Vero E6 cells.
  • FIG. 9 depicts a chart summarizing the in vitro cellular viability of Vero E6 cells in the presence of various compositions (oleandrin, digoxin and PBI-05204).
  • FIGS. 10A and 10B depict charts summarizing the ability of compositions (oleandrin and PBI-05204) to inhibit Ebolavirus in Vero E6 cells shortly after exposure to virus: FIG. 10A- 2 hr post-infection; FIG. 10B- 24 hr post-infection.
  • FIGS. 11A and 11B depict charts summarizing the ability of compositions
  • FIGS. 12A and 12B depict charts summarizing the ability of compositions
  • FIGS. 13A and 13B depict charts summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against consuelen Equine Encephalomyelits virus (FIG. 13 A) and Western Equine Encephalomyelitis virus (FIG. 13B) in Vero E6 cells.
  • the invention provides a method of treating viral infection in a subj ect by chronic administration of an effective dose of antiviral composition (or pharmaceutical composition comprising the antiviral composition and at least one pharmaceutical excipient) to the subject.
  • the pharmaceutical composition is administered according to a dosing regimen best suited for the subject, the suitability of the dose and dosing regimen to be determined clinically according to conventional clinical practices and clinical treatment endpoints for viral infection.
  • the term "subject” is taken to mean warm blooded animals such as mammals, for example, cats, dogs, mice, guinea pigs, horses, bovine cows, sheep, and humans.
  • a subject at risk of viral infection is: a) a subject living in a geographical area within which mosquitos, in particular A edes species (Aedes egypti, Aedes albopictus) mosquitos, live; b) a subject living with or near a person or people having viral infection; c) a subject having sexual relations with a person having a viral infection; d) a subject living in a geographical area within which ticks, in particular Ixodes species (Ixodes marx, Ixodes scapular is, ox Ixodes cooke species) ticks, live; e) a subject living in a geographical area within which fruit bats live; f) subjects living in a tropical region; g) subjects living in Africa; h) subjects in contact with bodily fluids of other subjects having a viral infection; i) a child; or j) a subject with a weakened immune system.
  • the subject is a) a subject living in a geographical area within
  • a subject treated according to the invention will exhibit a therapeutic response.
  • therapeutic response is meant that a subject suffering from the viral infection will enjoy at least one of the following clinical benefits as a result of treatment with a cardiac glycoside: reduction of the active viral titre in the subject's blood or plasma, eradication of active virus from the subject's blood or plasma, amelioration of the infection, reduction in the occurrence of symptoms associated with the infection, partial or full remission of the infection or increased time to progression of the infection.
  • the therapeutic response can be a full or partial therapeutic response.
  • time to progression is the period, length or duration of time after viral infection is diagnosed (or treated) until the infection begins to worsen. It is the period of time during which the level of infection is maintained without further progression of the infection, and the period of time ends when the infection begins to progress again. Progression of a disease is determined by "staging" a subject suffering from the infection prior to or at initiation of therapy. For example, the subject's health is determined prior to or at initiation of therapy. The subject is then treated with cardiac glycoside, and the viral titre is monitored periodically. At some later point in time, the symptoms of the infection may worsen, thus marking progression of the infection and the end of the "time to progression". The period of time during which the infection did not progress or during which the level or severity of the infection did not worsen is the "time to progression".
  • a dosing regimen includes a therapeutically relevant dose (or effective dose) of one or more cardiac glycosides administered according to a dosing schedule.
  • a therapeutically relevant dose therefore, is a therapeutic dose at which a therapeutic response of the viral infection to treatment with antiviral composition is observed and at which a subject can be administered the antiviral composition without an excessive amount of unwanted or deleterious side effects.
  • a therapeutically relevant dose is non-lethal to a subject, even though it may cause some side effects in the patient. It is a dose at which the level of clinical benefit to a subject being administered the antiviral composition exceeds the level of deleterious side effects experienced by the subject due to administration of the antiviral composition or component(s) thereof.
  • a therapeutically relevant dose can be administered according to any dosing regimen typically used in the treatment of viral infection.
  • a therapeutically relevant dose can be administered once, twice, thrice or more daily. It can be administered every other day, every third day, every fourth day, every fifth day, semiweekly, weekly, biweekly, every three weeks, every four weeks, monthly, bimonthly, semimonthly, every three months, every four months, semiannually, annually, or according to a combination of any of the above to arrive at a suitable dosing schedule.
  • a therapeutically relevant dose can be administered one or more times daily for one or more weeks.
  • compositions containing different amounts of oleandrin can be adjusted according to the concentration of oleandrin they contain, and converted that to molarity.
  • FIGS. 1-4 depict the efficacy based on the oleandrin content of the extracts.
  • OL on its own is efficacious.
  • PBI-05204 the SCF extract of Nerium oleander comprising OL, OA, UA and BA, is substantially more efficacious than OL on its own.
  • FIG. 7 a chart summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against Ebolavirus (EBOV) in Vero E6 cells.
  • FIG. 8 depicts a chart summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against Marburgvirus (MARV) in Vero E6 cells.
  • FIG. 9 depicts a chart summarizing the in vitro cellular viability of Vero E6 cells in the presence of various compositions (oleandrin, digoxin and PBI-05204).
  • the host cells were exposed to the compositions prior to infection with virus.
  • the invention provides a method of treating viral infection in a mammal or host cell, the method comprising: administering an antiviral composition to the mammal or host cell prior to contraction of said viral infection, whereby upon viral infection of said mammal or host cell, the antiviral composition reduces the viral titre and ameliorates, reduces or eliminates the viral infection.
  • FIGS. 10A and 10B depict charts summarizing the ability of compositions (oleandrin and PBI-05204) to inhibit Ebolavirus in Vero E6 cells shortly after exposure to virus: FIG. 10A- 2 hr post-infection; FIG. 10B- 24 hr post-infection.
  • the antiviral composition is administered within two hours (or within up to 12 hours) after viral infection, the viral titre antiviral composition provides effective treatment and reduces the EBOV viral titre. Even after 24 hours, the viral composition is effective; however, its efficacy is lower as time after initial viral infection increases. The same evaluations were conducted on MARV.
  • FIGS. 10A and 10B depict charts summarizing the ability of compositions (oleandrin and PBI-05204) to inhibit Ebolavirus in Vero E6 cells shortly after exposure to virus: FIG. 10A- 2 hr post-infection; FIG. 10B- 24 hr post-infection.
  • the viral titre antiviral composition provides effective treatment and reduces the EB
  • FIG. 11A and 11B depict charts summarizing the ability of compositions (oleandrin and PBI-05204) to inhibit Marburgvirus in Vero E6 cells shortly after exposure to virus: FIG. 11 A- 2 hr post-infection; FIG. 1 IB- 24 hr post-infection.
  • the antiviral composition is administered within two hours (or within up to 12 hours) after viral infection, the viral titre antiviral composition provides effective treatment and reduces the MARV viral titre. Even after 24 hours, the viral composition is effective; however, its efficacy is lower as time after initial viral infection increases.
  • the antiviral activity of the composition herein is reduced for a single generation of virus-infected cells, e.g. within 24 hours post-infection.
  • Vero E6 cells were infected with EBOV or MARV in the presence of oleandrin or PBI-05204 and incubated for 48hr. Supernatants from infected cell cultures were passaged onto fresh Vero E6 cells, incubated for lhr, then discarded. Cells containing passaged supernatant were incubated for 48hr. Cells infected with EBOV (B) or MARV (C) were evaluated as described herein. Control infection rates were 66% for EBOV and 67% for MARV.
  • the antiviral composition of the invention inhibited production of infectious progeny.
  • the antiviral composition of the invention a) can be administered prophylactically before viral infection to inhibit viral infection after exposure to virus; b) can be administered after viral infection to inhibit or reduce viral replication and production of infectious progeny; or c) a combination of a) and b).
  • FIGS. 13A and 13B depict charts summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against consuelen Equine Encephalomyelits virus (FIG. 13A) and Western Equine Encephalomyelitis virus (FIG. 13B) in Vero E6 cells.
  • the invention provides a method of treating a viral infection, caused by a Filoviridae virus, Flaviviridae virus or Togaviridae virus, in a subject or host cell, the method comprising administering an effective amount of the antiviral composition, thereby exposing the virus to the antiviral composition and treating said viral infection.
  • Rhinovirus is of the Picornaviridae family and Enterovirus genus. It is not enveloped and is an ss-RNA virus of (+) polarity. Oleandrin was found to be inactive against rhinovirus in the concentrations and assays employed herein.
  • PBI-04711 is a fraction of PBI-05204, but it does not contain cardiac glycoside (OL).
  • the molar ratio of OA: UA:B A in PBI-04711 is about 3:2.2: 1.
  • PBI-04711 also possesses antiviral activity. Accordingly, an antiviral composition comprising OL, OA, UA, and BA is more efficacious than a composition comprising OL as the sole active ingredient based upon an equimolar content of OL.
  • the molar ratios of the individual triterpenes to oleandrin range as follows: 2-8 (OA) : 2-8 (UA) : 0.1-1 (BA) : 0.5-1.5 (OL); or 3-6 (OA) : 3-6 (UA) : 0.3-8 (BA) : 0.7-1.2 (OL); or 4-5 (OA) : 4-5 (UA) : 0.4-0.7 (BA) : 0.9-1.1 (OL); or 4.6 (OA) : 4.4 (UA) : 0.6 (BA) : 1 (OL).
  • Antiviral compositions comprising oleandrin as the sole antiviral agent are within the scope of the invention.
  • Antiviral compositions comprising oleandrin and plural triterpenes as the antiviral agents are within the scope of the invention.
  • the antiviral composition comprises oleandrin, oleanolic acid (free acid, salt, derivative or prodrug thereof), ursolic acid (free acid, salt, derivative or prodrug thereof), and betulinic acid (free acid, salt, derivative or prodrug thereof).
  • the molar ratios of the compounds is as described herein.
  • Antiviral compositions comprising plural triterpenes as the primary active ingredients (meaning excluding steroid, cardiac glycoside and pharmacologically active components) are also within the scope of the invention.
  • PBI-04711 comprises OA, UA and BA as the primary active ingredients, and it exhibits antiviral activity.
  • atriterpene-based antiviral composition comprises OA, UA and BA, each of which is independently selected upon each occurrence from its free acid form, salt form, deuterated form and derivative form.
  • an antiviral composition comprises at least oleanolic acid (free acid, salt, derivative or prodrug thereof) and ursolic acid (free acid, salt, derivative or prodrug thereof) present at a molar ratio of OA to UA as described herein. OA is present in large molar excess over UA.
  • an antiviral composition comprises at least oleanolic acid (free acid, salt, derivative or prodrug thereof) and betulinic acid (free acid, salt, derivative or prodrug thereof) present at a molar ratio of OA to BA as described herein.
  • OA is present in large molar excess over BA.
  • the present method invention comprises: administering to the subject in need thereof a therapeutically relevant dose of antiviral composition and a therapeutically relevant dose of said one or more other therapeutic agents, wherein the antiviral composition is administered according to a first dosing regimen and the one or more other therapeutic agents is administered according to a second dosing regimen.
  • the first and second dosing regimens are the same. In some embodiments, the first and second dosing regimens are different.
  • the antiviral composition(s) of the invention can be administered as primary antiviral therapy, adjunct antiviral therapy, or co-antiviral therapy.
  • Methods of the invention include separate administration or coadministration of the antiviral composition with at least one other known antiviral composition, meaning the antiviral composition of the invention can be administered before, during or after administration of a known antiviral composition (compound(s)) or of a composition for treating symptoms associated with the viral infection.
  • a known antiviral composition compound(s)
  • a composition for treating symptoms associated with the viral infection for example, medications used to treat inflammation, vomiting, nausea, headache, fever, diarrhea, nausea, hives, conjunctivitis, malaise, muscle pain, joint pain, seizure, or paralysis can be administered with or separately from the antiviral composition of the invention.
  • the one or more other therapeutic agents can be administered at doses and according to dosing regimens that are clinician-recognized as being therapeutically effective or at doses that are clinician-recognized as being sub-therapeutically effective.
  • the clinical benefit and/or therapeutic effect provided by administration of a combination of antiviral composition and one or more other therapeutic can be additive or synergistic, such level of benefit or effect being determined by comparison of administration of the combination to administration of the individual antiviral composition component(s) and one or more other therapeutic agents.
  • the one or more other therapeutic agents can be administered at doses and according to dosing regimens as suggested or described by the Food and Drug Administration, World Health Organization, European Medicines Agency (E.M.E.A.), Therapeutic Goods Administration (TGA, Australia), Pan American Health Organization (PAHO), Medicines and Medical Devices Safety Authority (Medsafe, New Zealand) or the various agencies of Health worldwide.
  • Food and Drug Administration World Health Organization, European Medicines Agency (E.M.E.A.), Therapeutic Goods Administration (TGA, Australia), Pan American Health Organization (PAHO), Medicines and Medical Devices Safety Authority (Medsafe, New Zealand) or the various agencies of Health worldwide.
  • oleandrin is taken to mean all known forms of oleandrin unless otherwise specified. Oleandrin can be present in racemic, optically pure or optically enriched form. Nerium oleander plant material can be obtained, for example, from commercial plant suppliers such as Aldridge Nursery, Atascosa, Texas.
  • the supercritical fluid (SCF) extract can be prepared as detailed in US 7,402,325, US 8394434, US 8187644, or PCT International Publication No. WP 2007/016176 A2, the entire disclosures of which are hereby incorporated by reference. Extraction can be conducted with supercritical carbon dioxide in the presence or absence of a modifier (organic solvent) such as ethanol.
  • a modifier organic solvent
  • Organic solvent extracts of Nerium oleander are disclosed by Adome et al. (Afr. Health Sci. (2003) Aug. 3(2), 77-86; ethanolic extract), el-Shazly et al. (J. Egypt Soc. Parasitol. (1996), Aug. 26(2), 461-473; ethanolic extract), Begum et al. (Phytochemistry (1999) Feb. 50(3), 435-438; methanolic extract), Zia et al. (J. Ethnolpharmacol. (1995) Nov. 49(1), 33-39; methanolic extract), and Vlasenko et al. (Farmatsiia. (1972) Sept.-Oct.
  • the SCF extract comprises about 2% by wt. of oleandrin.
  • the SCF extract contains a 3-10 fold higher concentration of oleandrin than the hot-water extract. This was confirmed by both HPLC as well as LC/MS/MS (tandem mass spectrometry) analyses.
  • the SCF extract comprises oleandrin and the triterpenes oleanolic acid, betulinic acid and ursolic acid and optionally other components as described herein.
  • the content of oleandrin and the triterpenes can vary from batch to batch; however, the degree of variation is not excessive.
  • a batch of SCF extract (PBI-05204) was analyzed for these four components and found to contain the following approximate amounts of each.
  • Oleandrin, oleanolic acid, ursolic acid, betulinic acid and derivatives thereof can also be purchased from Sigma- Aldrich (www . si gmaal dnch. com; St. Louis, MO, USA).
  • triterpenes can independently be selected upon each occurrence in their native (unmodified, free acid) form, in their salt form, in derivative form, prodrug form, or a combination thereof.
  • Compositions containing and methods employing deuterated forms of the triterpenes are also within the scope of the invention.
  • Oleanolic acid derivatives, prodrugs and salts are disclosed in US 20150011627 Al to Gribble et al. which published Jan. 8, 2015, US 20140343108 Al to Rong et al which published Nov. 20, 2014, US 20140343064 Al to Xu et al. which published Nov. 20, 2014, US 20140179928 Al to Anderson et al. which published June 26, 2014, US 20140100227 Al to Bender et al. which published April 10, 2014, US 20140088188 Al to Jiang et al. which published Mar. 27, 2014, US 20140088163 Al to Jiang et al. which published Mar. 27, 2014, US 20140066408 Al to Jiang et al. which published Mar.
  • Ursolic acid derivatives, prodrugs and salts are disclosed in US 20150011627 Al to Gribble et al. which published Jan. 8, 2015, US 20130303607 Al to Gribble et al. which published Nov. 14, 2013, US 20150218206 Al to Yoon et al. which published Aug. 6, 2015, US 6824811 to Fritsche et al. which issued Nov. 30, 2004, US 7718635 to Ochiai et al. which issued May 8, 2010, US 8729055 to Lin et al. which issued May 20, 2014, and US 9120839 to Yoon et al. which issued Sep. 1, 2015, the entire disclosures of which are hereby incorporated by reference.
  • the antiviral composition can be formulated in any suitable pharmaceutically acceptable dosage form.
  • Parenteral, otic, ophthalmic, nasal, inhalable, buccal, sublingual, enteral, topical, oral, peroral, and injectable dosage forms are particularly useful.
  • Particular dosage forms include a solid or liquid dosage forms.
  • Exemplary suitable dosage forms include tablet, capsule, pill, caplet, troche, sache, solution, suspension, dispersion, vial, bag, bottle, injectable liquid, i.v. (intravenous), i.m. (intramuscular) or i.p. (intraperitoneal) administrable liquid and other such dosage forms known to the artisan of ordinary skill in the pharmaceutical sciences.
  • Suitable dosage forms containing the antiviral composition can be prepared by mixing the antiviral composition with pharmaceutically acceptable excipients as described herein or as described in Pi et al. ("Ursolic acid nanocrystals for dissolution rate and bioavailability enhancement: influence of different particle size” in Curr. Drug Deliv. (Mar 2016), 13(8), 1358-1366), Yang et al. ("Self-microemulsifying drug delivery system for improved oral bioavailability of oleanolic acid: design and evaluation" in Int. J. Nanomed. (2013), 8(1), 2917-2926), Li et al.
  • Suitable dosage forms can also be made according to US 8187644 B2 to Addington, which issued May 29, 2012, US 7402325 B2 to Addington, which issued July 22, 2008, US 8394434 B2 to Addington et al, which issued Mar. 12, 2013, the entire disclosures of which are hereby incorporated by reference. Suitable dosage forms can also be made as described in Examples 13-15.
  • an effective amount or therapeutically relevant amount of antiviral compound is specifically contemplated.
  • an effective amount it is understood that a pharmaceutically effective amount is contemplated.
  • a pharmaceutically effective amount is the amount or quantity of active ingredient which is enough for the required or desired therapeutic response, or in other words, the amount, which is sufficient to elicit an appreciable biological response when, administered to a patient.
  • the appreciable biological response may occur as a result of administration of single or multiple doses of an active substance.
  • a dose may comprise one or more dosage forms. It will be understood that the specific dose level for any patient will depend upon a variety of factors including the indication being treated, severity of the indication, patient health, age, gender, weight, diet, pharmacological response, the specific dosage form employed, and other such factors.
  • the desired dose for oral administration is up to 5 dosage forms although as few as one and as many as ten dosage forms may be administered as a single dose.
  • Exemplary dosage forms can contain 0.01-100 mg or 0.01-100 microg of the antiviral composition per dosage form, for a total 0.1 to 500 mg (1 to 10 dose levels) per dose.
  • Doses will be administered according to dosing regimens that may be predetermined and/or tailored to achieve specific therapeutic response or clinical benefit in a subject.
  • the antiviral can be included in an oral dosage form. Some embodiments of the dosage form are not enteric coated and release their charge of antiviral composition within a period of 0.5 to 1 hours or less. Some embodiments of the dosage form are enteric coated and release their charge of antiviral composition downstream of the stomach, such as from the jejunum, ileum, small intestine, and/or large intestine (colon). Enterically coated dosage forms will release antiviral composition into the systemic circulation within 1-10 hr after oral administration.
  • the antiviral composition can be included in a rapid release, immediate release, controlled release, sustained release, prolonged release, extended release, burst release, continuous release, slow release, or pulsed release dosage form or in a dosage form that exhibits two or more of those types of release.
  • the release profile of antiviral composition from the dosage form can be a zero order, pseudo-zero, first order, pseudo-first order or sigmoidal release profile.
  • the plasma concentration profile for triterpene in a subject to which the antiviral composition is administered can exhibit one or more maxima.
  • oleandrin Based on human clinical data it is anticipated that 50% to 75% of an administered dose of oleandrin will be orally bioavailable therefore providing 10 to 20 microg, 20 to 40 microg, 30 to 50 microg, 40 to 60 microg, 50 to 75 microg, 75 to 100 microg of oleandrin per dosage form. Given an average blood volume in adult humans of 5 liters, the anticipated oleandrin plasma concentration will be in the range of 0.05 to 2 ng/ml, 0.005 to 10 ng/mL, 0.005 to 8 ng/mL, 0.01 to 7 ng/mL, 0.02 to 7 ng/mL, 0.03 to 6 ng/mL, 0.04 to 5 ng/mL, or 0.05 to 2.5 ng/mL.
  • the recommended daily dose of oleandrin, present in the SCF extract, is generally about 0.2 microg to about 4.5 microg/kg body weight twice daily.
  • the dose of oleandrin can be about 0.2 microg to about 1 microg/kg body weight/day, about 0.5 to about 1.0 microg/kg body weight/day, about 0.75 to about 1.5 microg/kg body weight/day, about 1.5 to about 2.52 microg/kg body weight/day, about 2.5 to about 3.0 microg/kg body weight/day, about 3.0 to 4.0 microg/kg body weight/day or about 3.5 to 4.5 microg oleandrin/kg body weight/day.
  • the maximum tolerated dose of oleandrin can be about about 3.5 microg/kg body weight/day to about 4.0 microg/kg body weight/day.
  • the minimum effective dose can be about 0.5 microg/day, about 1 microg/day, about 1.5 microg/day, about 1.8 microg/day, about 2 microg/day, or about 5 microg/day.
  • the antiviral composition can be administered at low to high dose due to the combination of triterpenes present and the molar ratio at which they are present.
  • a therapeutically effective dose for humans is approximately 100-1000 mg or 100-1000 microg of antiviral composition per Kg of body weight. Such a dose can be administered up to 10 times in a 24-hour period. Other suitable dosing ranges are specified below.
  • a compound herein might possess one or more functions in a composition or formulation of the invention.
  • a compound might serve as both a surfactant and a water miscible solvent or as both a surfactant and a water immiscible solvent.
  • water soluble solvent or “water miscible solvent”, which terms are used interchangeably, refer to an organic liquid which does not form a biphasic mixture with water or is sufficiently soluble in water to provide an aqueous solvent mixture containing at least five percent of solvent without separation of liquid phases.
  • the solvent is suitable for administration to humans or animals.
  • Exemplary water soluble solvents include, by way of example and without limitation, PEG (poly(ethylene glycol)), PEG 400 (poly(ethylene glycol having an approximate molecular weight of about 400), ethanol, acetone, alkanol, alcohol, ether, propylene glycol, glycerin, triacetin, poly(propylene glycol), PVP (poly(vinyl pyrrolidone)), dimethylsulfoxide, N,N- dimethylformamide, formamide, ⁇ , ⁇ -dimethylacetamide, pyridine, propanol, N- methylacetamide, butanol, soluphor (2-pyrrolidone), pharmasolve (N-methyl-2- pyrrolidone).
  • PEG poly(ethylene glycol)
  • PEG 400 poly(ethylene glycol having an approximate molecular weight of about 400
  • ethanol acetone
  • alkanol alcohol
  • ether propylene glycol
  • glycerin triacetin
  • Class 3 solvents include, for example, acetic acid, acetone, anisole, 1 -butanol, 2-butanol, butyl acetate, tert-butlymethyl ether, cumene, ethanol, ethyl ether, ethyl acetate, ethyl formate, formic acid, heptane, isobutyl acetate, isopropyl acetate, methyl acetate, methyl- 1 -butanol, methylethyl ketone, methylisobutyl ketone, 2-methyl-l-propanol, pentane, 1-pentanol, 1- propanol, 2-propanol, or propyl acetate.
  • a surfactant refers to a compound that comprises polar or charged hydrophilic moieties as well as non-polar hydrophobic (lipophilic) moieties; i.e., a surfactant is amphiphilic.
  • the term surfactant may refer to one or a mixture of compounds.
  • a surfactant can be a solubilizing agent, an emulsifying agent or a dispersing agent.
  • a surfactant can be hydrophilic or hydrophobic.
  • the hydrophilic surfactant can be any hydrophilic surfactant suitable for use in pharmaceutical compositions.
  • Such surfactants can be anionic, cationic, zwitterionic or non- ionic, although non-ionic hydrophilic surfactants are presently preferred. As discussed above, these non-ionic hydrophilic surfactants will generally have HLB values greater than about 10. Mixtures of hydrophilic surfactants are also within the scope of the invention.
  • hydrophobic surfactant can be any hydrophobic surfactant suitable for use in pharmaceutical compositions.
  • suitable hydrophobic surfactants will have an HLB value less than about 10. Mixtures of hydrophobic surfactants are also within the scope of the invention.
  • composition or formulation may further comprise one or more chelating agents, one or more preservatives, one or more antioxidants, one or more adsorbents, one or more acidifying agents, one or more alkalizing agents, one or more antifoaming agents, one or more buffering agents, one or more colorants, one or more electrolytes, one or more salts, one or more stabilizers, one or more tonicity modifiers, one or more diluents, or a combination thereof.
  • one or more chelating agents one or more preservatives, one or more antioxidants, one or more adsorbents, one or more acidifying agents, one or more alkalizing agents, one or more antifoaming agents, one or more buffering agents, one or more colorants, one or more electrolytes, one or more salts, one or more stabilizers, one or more tonicity modifiers, one or more diluents, or a combination thereof.
  • the salts prepared from organic acids such as amino acids, acetic, propionic, succinic, glycolic, stearic, lactic, malic, tartaric, citric, ascorbic, pamoic, maleic, hydroxymaleic, phenylacetic, glutamic, benzoic, salicylic, sulfanilic, 2-acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, and others known to those of ordinary skill in the art.
  • a pharmaceutically acceptable base is added to form the pharmaceutically acceptable salt. Lists of other suitable salts are found in Remington ' s Pharmaceutical Sciences, 17 th . ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the relevant disclosure of which is hereby incorporated by reference.
  • phrases "pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with tissues of human beings and animals and without excessive toxicity, irritation, allergic response, or any other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • a dosage form can be made by any conventional means known in the pharmaceutical industry.
  • a liquid dosage form can be prepared by providing at least one liquid carrier and antiviral composition in a container. One or more other excipients can be included in the liquid dosage form.
  • a solid dosage form can be prepared by providing at least one solid carrier and antiviral composition. One or more other excipients can be included in the solid dosage form.
  • a dosage form can be packaged using conventional packaging equipment and materials. It can be included in a pack, bottle, via, bag, syringe, envelope, packet, blister pack, box, ampoule, or other such container.
  • composition of the invention can be included in any dosage form.
  • dosage forms include a solid or liquid dosage forms.
  • exemplary suitable dosage forms include tablet, capsule, pill, caplet, troche, sache, and other such dosage forms known to the artisan of ordinary skill in the pharmaceutical sciences.
  • Powdered oleander leaves were prepared by harvesting, washing, and drying oleander leaf material, then passing the oleander leaf material through a comminuting and dehydrating apparatus such as those described in U.S. Patent Nos. 5,236,132, 5,598,979, 6,517,015, and 6,715,705.
  • the weight of the starting material used was 3.94 kg.
  • the starting material was combined with pure CCh at a pressure of 300 bar (30 MPa, 4351 psi) and a temperature of 50°C (122°F) in an extractor device. A total of 197 kg of CCh was used, to give a solvent to raw material ratio of 50: 1. The mixture of CCh and raw material was then passed through a separator device, which changed the pressure and temperature of the mixture and separated the extract from the carbon dioxide.
  • the extract (65 g) was obtained as a brownish, sticky, viscous material having a nice fragrance. The color was likely caused by chlorophyll and other residual chromophoric compounds.
  • the tubes and separator were rinsed out with acetone and the acetone was evaporated to give an addition 9 g of extract.
  • the total extract amount was 74 g.
  • the yield of the extract was 1.88%.
  • the content of oleandrin in the extract was calculated using high pressure liquid chromatography and mass spectrometry to be 560.1 mg, or a yield of 0.76%.
  • Powdered oleander leaves were prepared by harvesting, washing, and drying oleander leaf material, then passing the oleander leaf material through a comminuting and dehydrating apparatus such as those described in U.S. Patent Nos. 5,236,132, 5,598,979, 6,517,015, and 6,715,705.
  • the weight of the starting material used was 3.85 kg.
  • the starting material was combined with pure CCh and 5% ethanol as a modifier at a pressure of 280 bar (28 MP a, 4061 psi) and a temperature of 50°C (122°F) in an extractor device.
  • a total of 160 kg of CCh and 8 kg ethanol was used, to give a solvent to raw material ratio of 43.6 to 1.
  • the mixture of CCh, ethanol, and raw material was then passed through a separator device, which changed the pressure and temperature of the mixture and separated the extract from the carbon dioxide.
  • the extract (207 g) was obtained after the removal of ethanol as a dark green, sticky, viscous mass obviously containing some chlorophyll. Based on the weight of the starting material, the yield of the extract was 5.38%. The content of oleandrin in the extract was calculated using high pressure liquid chromatography and mass spectrometry to be 1.89 g, or a yield of 0.91%.
  • Hot water extraction is typically used to extract oleandrin and other active components from oleander leaves. Examples of hot water extraction processes can be found in U.S. Patent Nos. 5,135,745 and 5,869,060.
  • a hot water extraction was carried out using 5 g of powdered oleander leaves. Ten volumes of boiling water (by weight of the oleander starting material) were added to the powdered oleander leaves and the mixture was stirred constantly for 6 hours. The mixture was then filtered and the leaf residue was collected and extracted again under the same conditions. The filtrates were combined and lyophilized. The appearance of the extract was brown. The dried extract material weighed about 1.44 g. 34.21 mg of the extract material was dissolved in water and subjected to oleandrin content analysis using high pressure liquid chromatography and mass spectrometry. The amount of oleandrin was determined to be 3.68 mg. The oleandrin yield, based on the amount of extract, was calculated to be 0.26%.
  • a coating dispersion was prepared from the ingredients listed in the table below.
  • Spray nozzle was set such that both the nozzle and spray path were under the flow path of inlet air.
  • Vero E6 cells (aso known as Vero CI 008 cells, ATTC No. CRL-1586; https : //www, atcc. org/Prod ucts/All/CRL- 1 86. aspx) were infected with ZIKV (Zika virus strain PRVABC59; ATCC VR-1843; 3 ⁇ 4 ⁇ 5: ⁇ .8 ⁇ . ⁇ 3 ⁇ 4 ⁇ ⁇ 5/ ⁇ 11/ ⁇ . ⁇ -1843. ⁇ 5 ⁇ ) at an MOI (multiplicity of infection) of 0.2 in the presence of cardiac glycoside. Cells were incubated with virus and compound for 1 hr, after which the inoculum and compound were discarded.
  • ZIKV Zika virus strain PRVABC59
  • ATCC VR-1843 3 ⁇ 4 ⁇ 5: ⁇ .8 ⁇ . ⁇ 3 ⁇ 4 ⁇ ⁇ 5/ ⁇ 11/ ⁇ . ⁇ -1843. ⁇ 5 ⁇
  • MOI multiplicity of infection
  • Method B Compound in Extract Form
  • An extract containing a target compound being tested is evaluated as detailed in Method A, except that the amount of extract is normalized to the amount of target compound in the extract.
  • an extract containing 2% wt of oleandrin contains 20 microg of oleandrin per 1 mg of extract. Accordingly, if the intended amount of oleandrin for evaluation is 20 microg, then 1 mg of extract would be used in the assay.
  • Syloid 244FP is a colloidal silicon dioxide manufactured by Grace Davison. Colloidal silicon dioxide is commonly used to provide several functions, such as an adsorbant, glidant, and tablet disintegrant. Syloid 244FP was chosen for its ability to adsorb 3 times its weight in oil and for its 5.5 micron particle size.
  • the samples were prepared by dissolving the compound or extract in a fixed amount of HPLC solvent to achieve an approximate target concentration of oleandrin.
  • the retention time of oleandrin can be determined by using an intemal standard.
  • the concentration of oleandrin can be determined/ calibrated by developing a signal response curve using the intemal standard.
  • a pharmaceutical composition of the invention can be prepared any of the following methods. Mixing can be done under wet or dry conditions. The pharmaceutical composition can be compacted, dried or both during preparation. The pharmaceutical composition can be portioned into dosage forms.
  • At least one pharmaceutical excipient is mixed with at least one antiviral compound disclosed herein.
  • At least one pharmaceutical excipient is mixed with at least two antiviral compounds disclosed herein.
  • At least one pharmaceutical excipient is mixed with at least two triterpenes disclosed herein.
  • At least one pharmaceutical excipient is mixed with at least one cardiac glycoside disclosed herein and at least two triterpenes disclosed herein.
  • At least one pharmaceutical excipient is mixed with at least three triterpenes disclosed herein.
  • V-C (9) 0.75 0.75 7.5
  • Antiviral compositions can be prepared by mixing the individual triterpene components thereof to form a mixture.
  • the triterpene mixtures prepared above that provided acceptable antiviral activity were formulated into antiviral compositions.
  • Antiviral composition with oleanolic acid and ursolic acid Antiviral composition with oleanolic acid and ursolic acid
  • a pharmaceutically acceptable antiviral composition at least one pharmaceutically acceptable excipient was mixed in with the pharmacologically active agents.
  • An antiviral composition is formulated for administration to a mammal.
  • Antiviral composition with oleanolic acid and betulinic acid with oleanolic acid and betulinic acid
  • oleanolic acid and betulinic acid were mixed according to a predetermined molar ratio of the components as defined herein.
  • the components were mixed in solid form or were mixed in solvent(s), e.g. methanol, ethanol, chloroform, acetone, propanol, dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMAC), N-methylpyrrolidone (NMP), water or mixtures thereof.
  • solvent(s) e.g. methanol, ethanol, chloroform, acetone, propanol, dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMAC), N-methylpyrrolidone (NMP), water or mixtures thereof.
  • DMSO dimethyl sulfoxide
  • DMF dimethylformamide
  • DMAC dimethylacetamide
  • NMP N-methylpyrrolidone
  • oleanolic acid ursolic acid and betulinic acid were mixed according to a predetermined molar ratio of the components as defined herein.
  • the components were mixed in solid form or were mixed in solvent(s), e.g. methanol, ethanol, chloroform, acetone, propanol, dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMAC), N-methylpyrrolidone (NMP), water or mixtures thereof.
  • solvent(s) e.g. methanol, ethanol, chloroform, acetone, propanol, dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMAC), N-methylpyrrolidone (NMP), water or mixtures thereof.
  • DMSO dimethyl sulfoxide
  • DMF dimethylformamide
  • DMAC dimethylacetamide
  • NMP N-methylpyrrolidone
  • a pharmaceutically acceptable antiviral composition at least one pharmaceutically acceptable excipient was mixed in with the pharmacologically active agents.
  • An antiviral composition is formulated for administration to a mammal.
  • oleandrin oleanolic acid ursolic acid and betulinic acid were mixed according to a predetermined molar ratio of the components as defined herein.
  • the components were mixed in solid form or were mixed in solvent(s), e.g. methanol, ethanol, chloroform, acetone, propanol, dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMAC), N-methylpyrrolidone (NMP), water or mixtures thereof.
  • solvent(s) e.g. methanol, ethanol, chloroform, acetone, propanol, dimethyl sulfoxide (DMSO), dimethylformamide (DMF), dimethylacetamide (DMAC), N-methylpyrrolidone (NMP), water or mixtures thereof.
  • DMSO dimethyl sulfoxide
  • DMF dimethylformamide
  • DMAC dimethylacetamide
  • NMP N-methylpyrrolidone
  • Exemplary Filovirus infections include Ebolavirus and Marburgvirus.
  • a subject presenting with Filovirus infection is prescribed antiviral composition, and therapeutically relevant doses are administered to the subject according to a prescribed dosing regimen for a period of time.
  • the subject's level of therapeutic response is determined periodically.
  • the level of therapeutic response can be determined by determining the subject's Filovirus titre in blood or plasma. If the level of therapeutic response is too low at one dose, then the dose is escalated according to a predetermined dose escalation schedule until the desired level of therapeutic response in the subject is achieved. Treatment of the subject with antiviral composition is continued as needed and the dose or dosing regimen can be adjusted as needed until the patient reaches the desired clinical endpoint.
  • Method B Combination therapy: antiviral composition with another agent
  • Method A is followed except that the subject is prescribed and administered one or more other therapeutic agents for the treatment of Filovirus infection or symptoms thereof. Then one or more other therapeutic agents can be administered before, after or with the antiviral composition. Dose escalation (or de-escalation) of the one or more other therapeutic agents can also be done.
  • Exemplary Flavivirus infections include Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, Zikavirus, Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Chikungunya virus, Omsk Hemorrhagic Fever, Powassan virus infection.
  • a subj ect presenting with Flavivirus infection is prescribed antiviral composition, and therapeutically relevant doses are administered to the subject according to a prescribed dosing regimen for a period of time.
  • the subject's level of therapeutic response is determined periodically.
  • the level of therapeutic response can be determined by determining the subject's Flavivirus titre in blood or plasma. If the level of therapeutic response is too low at one dose, then the dose is escalated according to a predetermined dose escalation schedule until the desired level of therapeutic response in the subject is achieved. Treatment of the subject with antiviral composition is continued as needed and the dose or dosing regimen can be adjusted as needed until the patient reaches the desired clinical endpoint.
  • Method B Combination therapy: antiviral composition with another agent
  • Method A is followed except that the subject is prescribed and administered one or more other therapeutic agents for the treatment of Flavivirus infection or symptoms thereof. Then one or more other therapeutic agents can be administered before, after or with the antiviral composition. Dose escalation (or de-escalation) of the one or more other therapeutic agents can also be done.
  • a CPE-based antiviral assay was performed by infecting target cells in the presence or absence of test compositions, at a range of concentrations. Infection of target cells by results in cytopathic effects and cell death. In this type of assay, reduction of CPE in the presence of test composition, and the corresponding increase in cell viability, is used as an indicator of antiviral activity.
  • cell viability was determined with a neutral red readout. Viable cells incorporate neutral red in their lysosomes. Uptake of neutral red relies on the ability of live cells to maintain a lower pH inside their lysosomes than in the cytoplasm, and this active process requires ATP.
  • the neutral red dye becomes charged and is retained intracellularly. After a 3-hour incubation with neutral red (0.033%), the extracellular dye was removed, cells were washed with PBS, and the intracellular neutral red was solubilized with a solution of 50% ethanol + 1% acetic acid. The amount of neutral red in solution was quantified by reading the absorbance (optical density) of each well at 490 nm
  • Adherent cell lines were used to evaluate the antiviral activity of compositions against a panel of viruses. Compositions were pre-incubated with the target cells for 30 min before the addition of virus to the cells. The compositions were present in the cell culture medium for the duration of the infection incubation period. For each infection assay, a viability assay was set up in parallel using the same concentrations of compositions (duplicates) to determine cytotoxicity effects of the compositions in the absence of virus.
  • test compositions were determined by comparing infection levels (for immunostaining-based assay) or viability (for CPE-based assays) of cells under test conditions to the infection level or viability of uninfected cells. Cytotoxic effects were evaluated in uninfected cells by comparing viability in the presence of inhibitors to the viability of mock-treated cells. Cytotoxicity was determined by an XTT viability assay, which was conducted at the same timepoint as the readout for the corresponding infection assay.
  • Test compositions were dissolved in 100% methanol. Eight concentrations of the compositions were generated (in duplicate) by performing 8-fold dilutions, starting with 50 ⁇ as the highest concentration tested. The highest test concentration of composition (50 ⁇ ) resulted in a 0.25% final concentration of methanol (v/v%) in the culture medium. An 8-fold dilution series of methanol vehicle was included in each assay plate, with concentrations mirroring the final concentration of methanol in each composition test condition. When possible, the EC50 and CC50 of the composition was determined for each assay using GraphPad Prism software.
  • Antiviral activity was evaluated by the degree of protection against virus- induced cytopathic effects (CPE).
  • CPE virus- induced cytopathic effects
  • Quality controls for the neutralization assay were performed on every plate to determine: i) signal to background (S/B) values; ii) inhibition by the known inhibitors, and iii) variation of the assay, as measured by the coefficient of variation (C.V.) of all data points.
  • Overall variation in the infection assays ranged from 3.4% to 9.5%, and overall variation in the viability assays ranged from 1.4% to 3.2%, calculated as the average of all C.V. values.
  • the signal-to-background (S/B) for the infection assays ranged from 2.9 to 11.0, while the signal-to-background (S/B) for the viability assays ranged from 6.5 to 29.9.
  • CPE DENV2-induced cytopathic effect
  • Neutral Red readout For the DENV2 antiviral assay, the 08-10381 Montserrat strain was used. Viral stocks were generated in C6/36 insect cells. Vero cells (epithelial kidney cells derived from Cercopithecus aethiops) were maintained in MEM with 5% FBS (MEM5). For both the infection and the viability assays, cells were seeded at 10,000 cells per well in 96-well clear flat bottom plates and maintained in MEM5 at 37°C for 24 hours. The day of infection, samples were diluted 8-fold in U-bottom plates using MEM with 1% bovine serum albumin (BSA).
  • BSA bovine serum albumin
  • Test material dilutions were prepared at 1.25X the final concentration and 40 ⁇ 1 were incubated with the target cells at 37°C for 30 minutes. Following the test material preincubation, ⁇ of virus dilutions prepared in MEM with 1% BSA was added to each well (50 ⁇ 1 final volume per well) and plates were incubated at 37°C in a humidified incubator with 5% C02 for 3 hours. The volume of virus used in the assay was previously determined to produce a signal in the linear range inhibited by Ribavirin and compound A3, known inhibitors of DENV2. After the infection incubation, cells were washed with PBS, then MEM containing 2% FBS (MEM2) to remove unbound virus.
  • MEM2 MEM containing 2% FBS
  • Controls included cells incubated with no virus ("mock-infected"), infected cells incubated with medium alone, or infected cells in the presence of Ribavirin (0.5 ⁇ ) or A3 (0.5 ⁇ ).
  • a full duplicate inhibition curve with methanol vehicle only was included on the same assay plate.
  • Test material dilutions were prepared at 1.25X the final concentration and 40 ⁇ 1 were incubated with the target cells at 37°C for 30 minutes. Following the test material pre-incubation, ⁇ of virus dilutions prepared in MEM with 1% BSA was added to each well (50 ⁇ 1 final volume per well) and plates were incubated at 37°C in a humidified incubator with 5% C02 for 3 hours. After the infection incubation, cells were washed with PBS, then MEM containing 2% FBS (MEM2) to remove unbound virus. Subsequently, 50 ⁇ 1 of medium containing inhibitor dilutions prepared at a IX concentration in MEM2 was added to each well.
  • the plate was incubated at 37°C in the incubator (5% C02) for 6 days. Controls with no virus ("mock-infected'), infected cells incubated with medium alone, infected cells incubated with vehicle alone (methanol), and wells without cells (to determine background) were included in the assay plate. After 6 days of infection, cells were stained with neutral red to monitor cell viability. Test materials were evaluated in duplicates using serial 8-fold dilutions in infection medium. Controls included cells incubated with no virus ("mock-infected"), infected cells incubated with medium alone, or infected cells in the presence of A3 (0.5 ⁇ ). A full duplicate inhibition curve with methanol vehicle only was included on the same assay plate.
  • CPE-based viability data for the neutral red assays, cell viability was determined by monitoring the absorbance at 490 nm. The average signal obtained in wells with no cells was subtracted from all samples. Then, all data points were calculated as a percentage of the average signal observed in the 8 wells of mock (uninfected) cells on the same assay plate. Infected cells treated with medium alone reduced the signal to an average of 4.2% (for HRV), 26.9% (for DENV), and 5.1 % (for ZIKV) of that observed in uninfected cells.
  • the signal-to-background (S/B) for this assay was 2.9 (for DENV), and 7.2 (for ZIKV), determined as the viability percentage in "mock-infected" cells compared to that of infected cells treated with vehicle only.
  • Viability assay to assess compound-induced cytotoxicity: Mock-infected cells were incubated with inhibitor dilutions (or medium only) using the same experimental setup and inhibitor concentrations as was used in the corresponding infection assay. The incubation temperature and duration of the incubation period mirrored the conditions of the corresponding infection assay. Cell viability was evaluated with an XTT method.
  • the XTT assay measures mitochondrial activity and is based on the cleavage of yellow tetrazolium salt (XTT), which forms an orange formazan dye. The reaction only occurs in viable cells with active mitochondria. The formazan dye is directly quantified using a scanning multi- well spectrophotometer.
  • Vero E6 cells were infected with EBOV (A,B) or MARV (C,D). At 2hr postinfection (A,C) or 24hr post-infection (B,D), oleandrin or PBI-05204 was added to cells for lhr, then discarded and cells were returned to culture medium. At 48hr post-infection, infected cells were analyzed as in Figure 1.
  • Vero E6 cells were infected with EBOV or MARV in the presence of oleandrin or PBI-05204 and incubated for 48hr. Supernatants from infected cell cultures were passaged onto fresh Vero E6 cells, incubated for lhr, then discarded (as depicted in A). Cells containing passaged supernatant were incubated for 48hr. Cells infected with EBOV (B) or MARV (C) were detected as described previously. Control infection rates were 66% for EBOV and 67% for MARV.
  • A Venezuelan equine encephalitis virus
  • B Western equine encephalitis virus
  • Exemplary Paramyxoviridae family viral infections include Henipavirus genus infection, Nipah virus infection, or Hendra virus infection.
  • a subject presenting with Paramyxoviridae family infection is prescribed antiviral composition, and therapeutically relevant doses are administered to the subject according to a prescribed dosing regimen for a period of time.
  • the subject's level of therapeutic response is determined periodically.
  • the level of therapeutic response can be determined by determining the subject's virus titre in blood or plasma. If the level of therapeutic response is too low at one dose, then the dose is escalated according to a predetermined dose escalation schedule until the desired level of therapeutic response in the subject is achieved.
  • Treatment of the subject with antiviral composition is continued as needed and the dose or dosing regimen can be adjusted as needed until the patient reaches the desired clinical endpoint.
  • Method B Combination therapy: antiviral composition with another agent
  • the term “about” or “approximately” are taken to mean ⁇ 10%, ⁇ 5%, ⁇ 2.5% or ⁇ 1% of a specified valued. As used herein, the term “substantially” is taken to mean “to a large degree” or "at least a majority of or "more than 50% of

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Epidemiology (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Molecular Biology (AREA)
  • Virology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Botany (AREA)
  • Biotechnology (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Dermatology (AREA)
  • Dispersion Chemistry (AREA)
  • Biophysics (AREA)
  • Inorganic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicinal Preparation (AREA)
  • Steroid Compounds (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
EP18857189.7A 2017-09-14 2018-07-16 Verfahren und zusammensetzung zur behandlung von virusinfektionen Withdrawn EP3681508A4 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
PCT/US2017/051553 WO2018053123A1 (en) 2016-09-14 2017-09-14 Method and compositions for treating viral infection
US201862698365P 2018-07-16 2018-07-16
PCT/US2018/042226 WO2019055119A1 (en) 2017-09-14 2018-07-16 METHOD AND COMPOSITION FOR TREATING VIRAL INFECTION

Publications (2)

Publication Number Publication Date
EP3681508A1 true EP3681508A1 (de) 2020-07-22
EP3681508A4 EP3681508A4 (de) 2021-05-19

Family

ID=65723839

Family Applications (1)

Application Number Title Priority Date Filing Date
EP18857189.7A Withdrawn EP3681508A4 (de) 2017-09-14 2018-07-16 Verfahren und zusammensetzung zur behandlung von virusinfektionen

Country Status (9)

Country Link
EP (1) EP3681508A4 (de)
JP (3) JP6820450B2 (de)
KR (2) KR20210107904A (de)
AU (2) AU2018334386B2 (de)
CA (1) CA3075729A1 (de)
IL (2) IL285232B (de)
MX (1) MX2020002883A (de)
RU (1) RU2020113341A (de)
WO (1) WO2019055119A1 (de)

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11806359B2 (en) 2020-03-31 2023-11-07 Phoenix Biotechnology, Inc. Method and compositions for treating Coronavirus infection
SG11202105728YA (en) 2020-03-31 2021-11-29 Phoenix Biotechnology Inc Method and compositions for treating coronavirus infection
EP4009981B1 (de) 2020-03-31 2023-08-16 Phoenix Biotechnology, Inc. Verfahren und zusammensetzungen zur behandlung von coronavirusinfektionen
ES2965161T3 (es) * 2020-03-31 2024-04-11 Phoenix Biotechnology Inc Método y composiciones para el tratamiento de infección por coronavirus
FR3109297A1 (fr) * 2020-04-19 2021-10-22 Ephyla « Nouvel inhibiteur non protéique de furine »
WO2022094816A1 (en) * 2020-11-04 2022-05-12 Janssen Pharmaceuticals, Inc. Solid formulation
US20230310612A1 (en) * 2022-04-01 2023-10-05 Alexia Reif Anti-viral composition and method for preparing the same

Family Cites Families (62)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT84857B (pt) 1986-05-13 1990-02-08 Huseyin Ziya Ozel Processo para a preparacao de extractos de plantas da especie nerium e de composicoes farmaceuticas que os contem
US5236132A (en) 1992-01-03 1993-08-17 Vortec, Inc. Gradient-force comminuter/dehydrator apparatus and method
US5598979A (en) 1995-04-20 1997-02-04 Vortec, Inc. Closed loop gradient force comminuting and dehydrating system
CA2191923C (en) 1996-12-03 2000-10-24 Ji-Won Yoon Portulaca oleracea and tumor cell growth
CN1307473A (zh) 1998-05-19 2001-08-08 研究发展基金会 三萜组合物和它们的使用方法
US6565897B2 (en) 1998-09-24 2003-05-20 Ozelle Pharmaceuticals, Inc. Extract of nerium species, pharmaceutical composition thereof and methods for preparation thereof
US6517015B2 (en) 2000-03-21 2003-02-11 Frank F. Rowley, Jr. Two-stage comminuting and dehydrating system and method
AU2001284946B2 (en) 2000-08-18 2007-02-01 Advanced Life Sciences Inc. Prodrugs of betulinic acid derivatives for the treatment of cancer and HIV
US9931316B2 (en) * 2004-01-06 2018-04-03 Paul Edward Stamets Antiviral activity from medicinal mushrooms and their active constituents
US6715705B2 (en) 2001-03-16 2004-04-06 Frank F. Rowley, Jr. Two-stage comminuting and dehydrating system and method
ATE295087T1 (de) 2001-04-09 2005-05-15 Loders Croklaan Bv Konzentrat von triterpenen
EP1476031A1 (de) 2001-05-11 2004-11-17 University of Ottawa Angstlösende zusammensetzungen der marcgraviaceae enthaltend betulinsäure, deren derivate und verfahren
US20040082521A1 (en) 2002-03-29 2004-04-29 Azaya Therapeutics Inc. Novel formulations of digitalis glycosides for treating cell-proliferative and other diseases
AU2003240818B2 (en) * 2002-05-28 2006-05-04 Bette Pollard Cardiac glycosides to treat cystic fibrosis and other IL-8 dependent disorders
EP1585504A4 (de) 2002-11-06 2009-07-15 Azaya Therapeutics Inc Proteinstabilisierte liposomale formulierungen pharmazeutischer wirkstoffe
US20050026849A1 (en) 2003-03-28 2005-02-03 Singh Chandra U. Water soluble formulations of digitalis glycosides for treating cell-proliferative and other diseases
US20050239748A1 (en) 2004-03-17 2005-10-27 Panacos Pharmaceuticals, Inc. Pharmaceutical salts of 3-O-(3',3'-dimethylsuccinyl) betulinic acid
US7118508B2 (en) 2004-07-28 2006-10-10 General Motors Corporation Planetary transmissions having three interconnected gear members and clutched input members
AU2005327442B2 (en) 2005-02-09 2011-09-01 Dabur Pharma Limited Novel betulinic acid derivatives a-ring-condensed to a heterocyclic group
CA2603124A1 (en) 2005-03-29 2006-10-05 Regents Of The University Of Minnesota Selective oxidation of triterpenes employing tempo
US20060252733A1 (en) 2005-04-07 2006-11-09 Novelix Pharmaceuticals, Inc. Betulin, betulin derivatives, betulinic acid and betulinic acid derivatives as novel therapeutics in the treatment of disease of lipid and/or glucose metabolism
US20090131714A1 (en) 2005-06-08 2009-05-21 Krasutsky Pavel A Synthesis of betulonic and betulinic aldehydes
CA2611435C (en) 2005-06-10 2013-01-08 Pola Chemical Industries Inc. Novel triterpenic acid derivative and preparation for external application for skin comprising the same
US7402325B2 (en) * 2005-07-28 2008-07-22 Phoenix Biotechnology, Inc. Supercritical carbon dioxide extract of pharmacologically active components from Nerium oleander
CA2633402A1 (en) 2005-12-16 2007-07-12 Panacos Pharmaceuticals, Inc. Preparation of pharmaceutical salts of 3-o-(3',3'-dimethlsuccinyl) betulinic acid
EP1803461A1 (de) * 2005-12-27 2007-07-04 Heinz-Herbert Fiebig Therapeutische Verwendung eines Nerium-oleander Blätterextrakts
GB0604535D0 (en) 2006-03-07 2006-04-12 Sndv Sprl Betulonic acid derivatives
US20090203661A1 (en) 2006-10-12 2009-08-13 Safe Stephen H Betulinic acid, derivatives and analogs thereof and uses therefor
AR063546A1 (es) 2006-11-03 2009-01-28 Panacos Pharmaceuticals Inc DERIVADOS DE TRITERPENO, METODOS PARA SU PREPARACION, COMPOSICIONES FARMACEUTICAS QUE LOS COMPRENDEN Y SU USO EN LA FABRICACION DE MEDICAMENTOS PARA EL TRATAMIENTO DE INFECCIoN POR EL VIRUS VIH.
WO2008091532A1 (en) 2007-01-19 2008-07-31 Panacos Pharmaceuticals, Inc. Salts of 3-o-(3',3'-dimethylsuccinyl)betulinic acid and solid state forms thereof
US8987212B2 (en) 2007-08-07 2015-03-24 Muhammed Majeed Oleanoyl peptide composition and method of treating skin aging
EP2231165B1 (de) 2007-11-13 2013-05-15 Phoenix Biotechnology Inc. Verfahren zur bestimmung der wahrscheinlichkeit einer therapeutischen antwort bei der krebs-chemotherapie mit herzglykosid
BRPI0911422B8 (pt) 2008-04-18 2021-05-25 Reata Pharmaceuticals Inc compostos moduladores inflamatórios antioxidantes, composição farmacêutica e usos dos mesmos
CN102164941B (zh) 2008-04-18 2015-05-27 里亚塔医药公司 抗氧化剂炎症调节剂:具有饱和c环的齐墩果酸衍生物
PL2276493T3 (pl) 2008-04-18 2019-05-31 Reata Pharmaceuticals Inc Antyoksydacyjne modulatory stanu zapalnego: pochodne kwasu oleanolowego z amino- i innymi modyfikacjami przy C-17
TW201004627A (en) 2008-04-18 2010-02-01 Reata Pharmaceuticals Inc Antioxidant inflammation modulators: novel derivatives of oleanolic acid
CA2736568A1 (en) 2008-09-10 2010-03-18 Universite Du Quebec A Chicoutimi Bidesmosidic betulin and betulinic acid derivatives and uses thereof as antitumor agents
WO2010053817A1 (en) 2008-11-04 2010-05-14 Trustees Of Dartmouth College Betulinic acid derivatives and methods of use thereof
US8802727B2 (en) 2009-07-14 2014-08-12 Hetero Research Foundation, Hetero Drugs Limited Pharmaceutically acceptable salts of betulinic acid derivatives
CN101704872B (zh) 2009-11-23 2012-06-27 张南 23-羟基白桦酸衍生物、其制备方法及应用
WO2011064710A1 (en) 2009-11-30 2011-06-03 University Of Kwazulu-Natal In vitro anti-sickling activity of betulinic acid, oleanolic acid and their derivatives
US9011937B2 (en) 2010-11-22 2015-04-21 Phoenix Biotechnology, Inc. Method of treating neurological conditions with extract of Nerium species or Thevetia species
WO2011085307A1 (en) 2010-01-11 2011-07-14 Phoenix Biotechnology, Inc. Method of treating neurological conditions with cardiac glycosides
TWI386415B (zh) 2010-02-02 2013-02-21 Univ Kaohsiung Medical 熊果酸衍生物及其醫藥組合物
TWI410431B (zh) 2010-12-22 2013-10-01 Ind Tech Res Inst 齊敦果酸衍生物在製備預防或治療c型肝炎之藥物的用途
WO2012095705A1 (en) 2011-01-10 2012-07-19 Hetero Research Foundation Pharmaceutically acceptable salts of novel betulinic acid derivatives
PL2670764T3 (pl) 2011-01-31 2016-02-29 Bristol Myers Squibb Co C-28 aminy zmodyfikowanych C-3 pochodnych kwasu betulinowego jako inhibitory dojrzewania HIV
CN102558280B (zh) 2011-11-18 2014-09-17 温州大学 一种30-卤代桦木酸的生产方法
JP6043361B2 (ja) 2011-12-01 2016-12-14 ハンジョウ ベンシェン ファーマシューティカル シーオー., エルティーディー.Hangzhou Bensheng Pharmaceutical Co., Ltd. オレアノール酸アミド化誘導体、その調製方法及び使用
DK2786757T3 (da) 2011-12-01 2017-11-20 Hangzhou Bensheng Pharmaceutical Co Ltd 2-substitueret oleanolsyre-derivat, fremgangsmåde til fremstilling af samme, og anvendelse deraf
US20150119373A1 (en) 2012-04-24 2015-04-30 Hetero Research Foundation Novel betulinic acid derivatives as hiv inhibitors
US8921419B2 (en) 2012-05-08 2014-12-30 Trustees Of Dartmouth College Triterpenoids and compositions containing the same
CN104684883A (zh) 2012-08-14 2015-06-03 内克索利戈有限公司 新型乌索酸衍生物及其制备方法
WO2014040056A1 (en) 2012-09-10 2014-03-13 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US9278912B2 (en) 2012-09-10 2016-03-08 Reata Pharmaceuticals, Inc. C13-hydroxy derivatives of oleanolic acid and methods of use thereof
PE20150627A1 (es) 2012-09-10 2015-06-04 Reata Pharmaceuticals Inc Derivados de c17-alcandiilo y alquendiilo del acido oleanolico y sus metodos de uso
WO2014105926A1 (en) 2012-12-31 2014-07-03 Hetero Research Foundation Novel betulinic acid proline derivatives as hiv inhibitors
BR112015019590A2 (pt) 2013-02-25 2017-07-18 Bristol Myers Squibb Co derivados de ácido betulínico modificado com c-3 alquila e alquenila úteis no tratamento de hiv
WO2015121168A1 (de) * 2014-02-12 2015-08-20 Organobalance Gmbh Hefe-stamm und mikrobielles verfahren zur produktion von pentazyklischen triterpenen und/oder triterpenoiden
WO2015195776A1 (en) 2014-06-19 2015-12-23 Bristol-Myers Squibb Company Betulinic acid derivatives with hiv maturation inhibitory activity
US9492438B2 (en) * 2014-07-25 2016-11-15 Bette Pollard Amphiphilic pyridinum compounds to treat epilepsy and other disorders of the nervous system
MX2017015670A (es) * 2015-06-08 2018-08-15 Dermarc LLC Composicion terapeutica.

Also Published As

Publication number Publication date
JP7037673B2 (ja) 2022-03-16
AU2020204236B2 (en) 2020-12-10
KR102295179B1 (ko) 2021-08-30
CA3075729A1 (en) 2019-03-21
EP3681508A4 (de) 2021-05-19
KR20210107904A (ko) 2021-09-01
WO2019055119A1 (en) 2019-03-21
RU2020113341A3 (de) 2021-10-15
IL285232B (en) 2022-07-01
IL285232A (en) 2021-09-30
AU2018334386A1 (en) 2020-04-02
RU2020113341A (ru) 2021-10-15
JP2021063105A (ja) 2021-04-22
JP6820450B2 (ja) 2021-01-27
IL273162A (en) 2020-04-30
IL273162B (en) 2021-08-31
JP2020534293A (ja) 2020-11-26
MX2020002883A (es) 2020-10-05
JP2022075755A (ja) 2022-05-18
AU2018334386B2 (en) 2021-11-04
AU2020204236A1 (en) 2020-07-16
KR20200081356A (ko) 2020-07-07

Similar Documents

Publication Publication Date Title
EP3512524B1 (de) Verfahren und zusammensetzungen zur behandlung von virusinfektionen
US10596186B2 (en) Method and compositions for treating viral infections
AU2018334386B2 (en) Method and composition for treating viral infection
US10729735B1 (en) Method and compostitions for treating coronavirus infection
US11324789B2 (en) Method and compositions for treating equine encephalitis
IL291727B2 (en) Method and preparations for treating the corona virus infection
US20220047616A1 (en) Method and Compositions for Treating HTLV-1 Virus Infection
CA3091376C (en) Method and compositions for treating coronavirus infection
EP4295854A2 (de) Verfahren und zusammensetzungen zur behandlung von coronavirusinfektionen

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20200219

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Free format text: PREVIOUS MAIN CLASS: A61K0031560000

Ipc: A61K0036240000

A4 Supplementary search report drawn up and despatched

Effective date: 20210416

RIC1 Information provided on ipc code assigned before grant

Ipc: A61K 36/24 20060101AFI20210409BHEP

Ipc: A61K 31/56 20060101ALI20210409BHEP

Ipc: A61K 31/7042 20060101ALI20210409BHEP

Ipc: A61K 31/7048 20060101ALI20210409BHEP

Ipc: A61P 31/12 20060101ALI20210409BHEP

Ipc: A61P 31/14 20060101ALI20210409BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20221004

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20230921

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20240202