WO2019055119A1 - Method and composition for treating viral infection - Google Patents

Method and composition for treating viral infection Download PDF

Info

Publication number
WO2019055119A1
WO2019055119A1 PCT/US2018/042226 US2018042226W WO2019055119A1 WO 2019055119 A1 WO2019055119 A1 WO 2019055119A1 US 2018042226 W US2018042226 W US 2018042226W WO 2019055119 A1 WO2019055119 A1 WO 2019055119A1
Authority
WO
WIPO (PCT)
Prior art keywords
infection
acid
subject
antiviral composition
oleandrin
Prior art date
Application number
PCT/US2018/042226
Other languages
French (fr)
Inventor
Robert A. Newman
Otis C. Addington
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
Priority to CN201880059793.0A priority Critical patent/CN111093672B/en
Priority to EP18857189.7A priority patent/EP3681508A4/en
Priority to CA3075729A priority patent/CA3075729A1/en
Priority to IL285232A priority patent/IL285232B/en
Priority to CN202111022020.5A priority patent/CN114224900A/en
Priority to KR1020217026676A priority patent/KR20210107904A/en
Priority to AU2018334386A priority patent/AU2018334386B2/en
Priority to MX2020002883A priority patent/MX2020002883A/en
Priority to RU2020113341A priority patent/RU2020113341A/en
Priority to JP2020515911A priority patent/JP6820450B2/en
Priority to KR1020207007406A priority patent/KR102295179B1/en
Application filed by Phoenix Biotechnology, Inc. filed Critical Phoenix Biotechnology, Inc.
Priority to US16/276,063 priority patent/US10596186B2/en
Publication of WO2019055119A1 publication Critical patent/WO2019055119A1/en
Priority to US16/424,902 priority patent/US10702567B2/en
Priority to US16/747,637 priority patent/US11007239B2/en
Priority to US16/779,840 priority patent/US10874704B2/en
Priority to US16/803,488 priority patent/US11123387B2/en
Priority to IL273162A priority patent/IL273162B/en
Priority to US16/815,260 priority patent/US10980852B2/en
Priority to US16/825,063 priority patent/US11013776B2/en
Priority to US16/895,920 priority patent/US10729735B1/en
Priority to AU2020204236A priority patent/AU2020204236B2/en
Priority to US17/405,572 priority patent/US11324789B2/en

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

  • the present invention concerns an antiviral composition and its use for treating Flaviviridae infection, Togaviridae infection, or Filoviridae infection in mammals. Some embodiments concern treatment of hemorrhagic viral infection.
  • 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.
  • Erdemoglu et al. J. Ethnopharmacol. (2003) Nov. 89(1), 123-129) discloses results for the comparison of aqueous and ethanolic extracts of plants, including Nerium oleander, based upon their anti-nociceptive and anti-inflammatory activities.
  • Organic solvent extracts of Nerium oleander are also 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.
  • a supercritical fluid extract of Nerium species is known (US 8394434, US 8187644, US 7402325) and has demonstrated efficacy in treating neurological disorders (US 8481086, US 9220778, US 9358293, US 20160243143A1) and cell-proliferative disorders (US 8367363).
  • 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,
  • Viral hemorrhagic fever can be caused by five distinct virus families: Arenaviridae, Bunyaviridae, Filoviridae, Flaviviridae, and Paramyxoviridae.
  • the Filoviruses e.g. Ebolavirus (EBOV) and Marburgvirus (MARV), are among the most pathogenic viruses known to man and the causative agents of viral hemorrhagic fever outbreaks with fatality rates of up to 90%.
  • EBOV Ebolavirus
  • MARV Marburgvirus
  • Each virion contains one molecule of single- stranded, negative-sense RNA.
  • 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 invention provides a pharmaceutical composition and method for treating viral infection in a mammalian subject.
  • the invention also provides a pharmaceutical composition and method for treating viral infection, e.g. Viral hemorrhagic fever (VHF) infection, in a mammalian subject.
  • VHF Viral hemorrhagic fever
  • the invention also provides a method of treating viral infection in mammals by administration of the pharmaceutical composition.
  • the inventors have succeeded in preparing antiviral compositions that exhibit sufficient antiviral activity to justify their use in treating viral infection in humans and animals.
  • the inventors have developed corresponding treatment methods employing particular dosing regimens.
  • 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 Filovirus infection, Flavivirus infection, Henipavirus infection, alphavirus infection, or Togavirus infection.
  • Viral infections that can be treated include, at least, Ebolavirus, Marburgvirus, Alphavirus, Flavivirus, Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, Zikavirus, Venezuelan Equine Encephalomyelitis (encephalitis) (VEE) virus, Chikungunya virus, Western Equine Encephalomyelitis (encephalitis) (WEE) virus, Eastern Equine Encephalomyelitis (encephalitis) (EEE) virus, Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Omsk Hemorrhagic Fever, Hendra virus, Nipah virus, and species thereof.
  • Some embodiments of the invention are directed to compositions for and methods of treating viral infections from viruses of the Filovi
  • 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.
  • the invention provides an antiviral composition comprising (consisting essentially of): a) specific cardiac glycoside(s); b) plural triterpenes; or c) a combination of specific cardiac glycoside(s) and plural triterpenes.
  • 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 cardiac glycoside can be present in a pharmaceutical composition in pure form or as part of an extract containing one or more cardiac glycosides.
  • the triterpene(s) can be present in a pharmaceutical composition in pure form or as part of an extract containing triterpene(s).
  • the cardiac glycoside is present as the primary therapeutic component, meaning the component primarily responsible for antiviral activity, in the pharmaceutical composition.
  • the triterpene(s) is/are present as the primary therapeutic component(s), meaning the component(s) primarily responsible for antiviral activity, in the pharmaceutical composition.
  • an extract comprising the antiviral composition is obtained by extraction of plant material.
  • the extract can comprise a hot-water extract, cold- water extract, supercritical fluid (SCF) extract, organic solvent extract, or combination thereof of the plant material.
  • the plant material is Nerium species or Thevetia species plant mass. Particular species include Nerium oleander or Thevetia nerifolia.
  • the extract comprises at least one other pharmacologically active agent, obtained along with the cardiac glycoside during extraction, that contributes to the therapeutic efficacy of the cardiac glycoside when the extract is administered to a subject.
  • the composition further comprises one or more other non- cardiac glycoside therapeutically effective agents, i.e. one or more agents that are not cardiac glycosides.
  • the composition further comprises one or more antiviral compound(s).
  • the antiviral composition excludes a pharmacologically active polysaccharide.
  • 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 ratio of total triterpene content (OA + UA + BA) to oleandrin ranges from about 15: 1 to about 5: 1, or about 12: 1 to about 8: 1, or about 100: 1 to about 15: 1, or about 100: 1 to about 50: 1, or about 100: 1 to about 75: 1, or about 100: 1 to about 80: 1, or about 100: 1 to about 90: 1, or about 10: 1.
  • 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 manufacture can also include one or more additional steps such as: delivering the packaged dosage form to a vendor (retailer, wholesaler and/or distributor); selling or otherwise providing the packaged dosage form to a subj ect having a viral infection; including with the medicament a label and a package insert, which provides instructions on use, dosing regimen, administration, content and toxicology profile of the dosage form.
  • the treatment of viral infection comprises: determining that a subject has a viral infection; indicating administration of pharmaceutical dosage form to the subject according to a dosing regimen; administering to the subject one or more pharmaceutical dosage forms, wherein the one or more pharmaceutical dosage forms is administered according to the dosing regimen.
  • 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.
  • the water miscible solvent is low molecular weight (less than 6000) PEG, glycol, or alcohol.
  • the surfactant is a pegylated surfactant, meaning a surfactant comprising a poly(ethylene glycol) functional group.
  • the invention includes all combinations of the aspects, embodiments and sub-embodiments of the invention disclosed herein.
  • FIGS. 1-2 depict charts summarizing the in vitro dose response antiviral activity of various compositions against Ebolavirus.
  • FIGS. 3-4 depict charts summarizing the in vitro dose response antiviral activity of various compositions against Marburgvirus.
  • FIG. 5 depicts a chart summarizing the in vitro dose response antiviral activity of oleandrin against Zikavirus (SIKV strain PRVABC59) in Vero E6 cells.
  • FIG. 6 depicts a chart summarizing the in vitro dose response antiviral activity of digoxin against Zikavirus (SIKV strain PRVABC59) in Vero E6 cells.
  • 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 will vary from subject to subject according to a variety of established pharmacologic, pharmacodynamic and pharmacokinetic principles. However, a therapeutically relevant dose (relative, for example, to oleandrin) will typically not exceed 25 micrograms, 100 micrograms, 250 micrograms, 500 micrograms or 750 micrograms of cardiac glycoside/day or it can be in the range of 25-750 micrograms of cardiac glycoside per dose. It is known in the art that the actual amount of antiviral composition required to provide a target therapeutic result in a subject may vary from subject to subject according to the basic principles of pharmacy.
  • 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.
  • Example 15 provides a detailed description of an in vitro assay used to evaluate the efficacy of compositions containing oleandrin (as sole active), Anvirzel and PBI-05204 (supercritical fluid (SCF) extract ofNerium oleander) for the treatment of Ebolavirus (FIGS. 1-2) and Marburgvirus (FIGS. 3-4) infection, both of which are Filoviruses.
  • oleandrin as sole active
  • Anvirzel and PBI-05204 supercritical fluid (SCF) extract ofNerium oleander
  • 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.
  • Anvirzel the hot water extract of Nerium oleander, is more efficacious than OL on its own. Both extracts clearly exhibit efficacy in the nanomolar range.
  • the percentage of oleandrin in the PBI-05204 extract (1.74%) is higher than in Anvirzel (0.459%, 4.59 microg/mg).
  • Anvirzel did not exhibit complete inhibition, because at a dose higher than 20 microg/mL with anvirzel, toxicity is observed.
  • the data demonstrate highest antiviral activity against Ebolavirus and Marburgvirus for PBI-05204.
  • the combination of triterpenes in PBI-05204 increased the antiviral activity of oleandrin.
  • Example 6 provides a detailed description of an in vitro assay used to evaluate the efficacy of the cardiac glycosides for the treatment of Zikavirus (a flavivirus) infection.
  • Vero E6 cells were infected with Zika virus (ZIKV strain PRVABC59) at an MOI of 0.2 in the presence of oleandrin (FIG. 5) or digoxin (FIG. 6).
  • the cells were incubated with virus and the cardiac glycoside for 1 hr, after which the inoculum and non-absorbed cardiac glycoside (if any) was removed.
  • the cells were immersed in fresh medium and incubated for 48 hr, after which they were fixed with formalin and stained for ZIKV infection.
  • the data demonstrate antiviral activity against Zikavirus for both cardiac glycosides; however, oleandrin exhibited higher (almost 8-fold greater) antiviral activity than digoxin.
  • Example 14 provides a detailed description of an assay used to evaluate the antiviral activity of test compositions against Zikavirus and Dengue virus. The data indicate that oleandrin demonstrates efficacy against Zikavirus and Dengue virus.
  • 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.
  • the antiviral composition and method of the invention are also useful in treating viral infection that has occurred prior to administration of the antiviral composition.
  • Vero E6 cells were infected with EBOV (FIGS. 10A, 10B) or MARV (FIGS. 11 A, 1 IB).
  • EBOV EBOV
  • MARV MARV
  • oleandrin or PBI- 05204 was added to cells for lhr, then discarded and cells were returned to culture medium.
  • 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 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).
  • 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.
  • PBI-05204 (as described herein and in 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) comprises cardiac glycoside (oleandrin, OL) and triterpenes (oleanolic acid (OA), ursolic acid (UA) and betulinic acid (BA)) as the primary pharmacologically active components.
  • OL cardiac glycoside
  • OA oleanolic acid
  • U ursolic acid
  • BA betulinic acid
  • the molar ratio of OL to total triterpene is about l :(10-96).
  • the molar ratio of OA:UA:BA is about 7.8:7.4: 1.
  • 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.
  • PBI-01011 is an improved triterpene-based antiviral composition
  • OA, UA and BA wherein the molar ratio of OA:UA:BA is about 9-12 : up to about 2 : up to about 2, or about 10 : about 1 : about 1, or about 9-12 : about 0.1-2 : about 0.1-2, or about 9-11 : about 0.5-1.5 : about 0.5-1.5, or about 9.5-10.5 : about 0.75-1.25 : about 0.75-1.25, or about 9.5-10.5 : about 0.8-1.2 : about 0.8-1.2, or about 9.75-10.5 : about 0.9-1.1 : about 0.9-1.1.
  • 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), ursolic 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 UA to BA as described herein. OA is present in large molar excess over both UA and BA.
  • a triterpene-based antiviral composition excludes cardiac glycoside.
  • 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.
  • Example 5 provides an exemplary procedure for the treatment of Zikavirus infection in a mammal.
  • Example 12 provides an exemplary procedure for the treatment of Filovirus infection (Ebolavirus, Marburgvirus) in a mammal.
  • Example 13 provides an exemplary procedure for the treatment of Flavivirus infection (Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, Zikavirus, Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Omsk Hemorrhagic Fever, Powassan virus infection) in a mammal.
  • Flavivirus infection Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, Zikavirus, Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Omsk Hemorrhagic Fever, Powassan virus infection
  • the antiviral compound(s) (triterpene(s), cardiac glycoside(s), etc.) present in the pharmaceutical composition can be present in their unmodified form, salt form, derivative form or a combination thereof.
  • the term "derivative” is taken to mean: a) a chemical substance that is related structurally to a first chemical substance and theoretically derivable from it; b) a compound that is formed from a similar first compound or a compound that can be imagined to arise from another first compound, if one atom of the first compound is replaced with another atom or group of atoms; c) a compound derived or obtained from a parent compound and containing essential elements of the parent compound; or d) a chemical compound that may be produced from first compound of similar structure in one or more steps.
  • a derivative may include a deuterated form, oxidized form, dehydrated, unsaturated, polymer conjugated or glycosilated form thereof or may include an ester, amide, lactone, homolog, ether, thioether, cyano, amino, alkylamino, sulfhydryl, heterocyclic, heterocyclic ring-fused, polymerized, pegylated, benzylidenyl, triazolyl, piperazinyl or deuterated form thereof.
  • 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
  • Other extracts containing cardiac glycoside, especially oleandrin can be prepared by various different processes.
  • An extract can be prepared according to the process developed by Dr. Huseyin Ziya Ozel (U.S. Patent No. 5,135,745) describes a procedure for the preparation of a hot water extract.
  • the aqueous extract reportedly contains several polysaccharides with molecular weights varying from 2KD to 30KD, oleandrin and oleandrigenin, odoroside and neritaloside.
  • the polysaccharides reportedly include acidic homopolygalacturonans or arabinogalaturonans.
  • 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.
  • U.S. Pregrant Patent Application Publication No. 20040247660 to Singh et al. discloses the preparation of a protein stabilized liposomal formulation of oleandrin for use in the treatment of cancer.
  • U. S. Pregrant Patent Application Publication No. 20050026849 to Singh et al. discloses a water soluble formulation of oleandrin containing a cyclodextrin.
  • U.S. Pregrant Patent Application Publication No. 20040082521 to Singh et al. discloses the preparation of protein stabilized nanoparticle formulations of oleandrin from the hot-water extract.
  • the partial compositions of the SCF CCh extract and hot water extract were determined by DART TOF-MS (Direct Analysis in Real Time Time of Flight Mass Spectrometry) on a JEOL AccuTOF-DART mass spectrometer (JEOL USA, Peabody, MA, USA).
  • the SCF extract of Nerium species or Thevetia species is a mixture of pharmacologically active compounds, such as oleandrin and triterpenes.
  • the extract obtained by the SCF process is a substantially water-insoluble, viscous semi-solid (after solvent is removed) at ambient temperature.
  • the SCF extract comprises many different components possessing a variety of different ranges of water solubility.
  • the extract from a supercritical fluid process contains by weight a theoretical range of 0.9% to 2.5% wt of oleandrin or 1.7% to 2.1% wt of oleandrin or 1.7% to 2.0% wt of oleandrin.
  • SCF extracts comprising varying amount of oleandrin have been obtained.
  • 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.
  • the content of the individual components may vary by ⁇ 25%, ⁇ 20%, ⁇ 15%, ⁇ 10% or ⁇ 5% relative to the values indicated. Accordingly, the content of oleandrin in the SCF extract would be in the range of 20 mg ⁇ 5 mg (which is ⁇ 25% of 20 mg) per mg of SCF extract.
  • 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.
  • Betulinic 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 20120237629 Al to Shode et al. which published Sept. 20, 2012, US 20170204133 Al to Regueiro-Ren et al. which published July 20, 2017, US 20170096446 Al to Nitz et al. which published April 6, 2017, US 20150337004 Al to Parthasaradhi Reddy et al. which published Nov. 26, 2015, US 20150119373 Al to Parthasaradhi Reddy et al.
  • 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 cardiac glycoside can be present in a dosage form in an amount sufficient to provide a subject with an initial dose of oleandrin of 20 to 100 microg, 12 microg to 300 microg, or 12 microg to 120 microg.
  • a dosage form can comprise 20 of oleandrin to 100 microg, 0.01 microg to 100 mg or 0.01 microg to 100 microg oleandrin, oleandrin extract or extract of Nerium oleander containing oleandrin.
  • 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.
  • a liquid composition can comprise one or more pharmaceutically acceptable liquid carriers.
  • the liquid carrier can be an aqueous, non-aqueous, polar, non-polar, and/or organic carrier.
  • Liquid carriers include, by way of example and without limitation, a water miscible solvent, water immiscible solvent, water, buffer and mixtures thereof.
  • 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
  • water insoluble solvent or “water immiscible solvent”, which terms are used interchangeably, refer to an organic liquid which forms a biphasic mixture with water or provides a phase separation when the concentration of solvent in water exceeds five percent.
  • the solvent is suitable for administration to humans or animals.
  • Exemplary water insoluble solvents include, by way of example and without limitation, medium/long chain triglycerides, oil, castor oil, corn oil, vitamin E, vitamin E derivative, oleic acid, fatty acid, olive oil, softisan 645 (Diglyceryl Caprylate / Caprate / Stearate / Hydroxy stearate adipate), miglyol, captex (Captex 350: Glyceryl Tricaprylate/ Caprate/ Laurate triglyceride; Captex 355: Glyceryl Tricaprylate/ Caprate triglyceride; Captex 355 EP / NF: Glyceryl Tricaprylate/ Caprate medium chain triglyceride).
  • Suitable solvents are listed in the "International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) guidance for industry Q3C Impurities: Residual Solvents " (1997), which makes recommendations as to what amounts of residual solvents are considered safe in pharmaceuticals.
  • Exemplary solvents are listed as class 2 or class 3 solvents.
  • 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.
  • Captex 100 Propylene Glycol Dicaprate
  • Captex 200 Propylene Glycol Dicaprylate/ Dicaprate
  • Captex 200 P Propylene Glycol Dicaprylate/ Dicaprate
  • Captex 300 Glyceryl Tricaprylate/ Caprate
  • Captex 300 EP / NF Glyceryl Tricaprylate/ Caprate Medium Chain Triglycerides
  • Captex 350 Glyceryl Tricaprylate/ Caprate/ Laurate
  • Captex 355 Glyceryl Tricaprylate/ Caprate
  • Captex 355 EP / NF Glyceryl Tricaprylate/ Caprate Medium Chain Triglycerides
  • Captex 500 Triacetin
  • Captex 500 P Triacetin (Pharmaceutical Grade)
  • Captex 800 Propylene Glycol Di (2- Ethythexanoate)
  • Captex 810 D Glyceryl Tricaprylate/ Caprate
  • 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.
  • Examples of additional suitable solubilizer include: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether (glycofurol, available commercially from BASF under the trade name Tetraglycol) or methoxy PEG (Union Carbide); amides, such as 2-pyrrolidone, 2-piperidon
  • 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.
  • composition of the invention can also include oils such as fixed oils, peanut oil, sesame oil, cottonseed oil, corn oil and olive oil; fatty acids such as oleic acid, stearic acid and isostearic acid; and fatty acid esters such as ethyl oleate, isopropyl myristate, fatty acid glycerides and acetylated fatty acid glycerides.
  • oils such as fixed oils, peanut oil, sesame oil, cottonseed oil, corn oil and olive oil
  • fatty acids such as oleic acid, stearic acid and isostearic acid
  • fatty acid esters such as ethyl oleate, isopropyl myristate, fatty acid glycerides and acetylated fatty acid glycerides.
  • the composition can also include alcohol such as ethanol, isopropanol, hexadecyl alcohol, glycerol and propylene glycol; glycerol ketals such as 2,2-dimethyl-l,3-dioxolane-4-methanol; ethers such as poly(ethylene glycol) 450; petroleum hydrocarbons such as mineral oil and petrolatum; water; a pharmaceutically suitable surfactant, suspending agent or emulsifying agent; or mixtures thereof.
  • alcohol such as ethanol, isopropanol, hexadecyl alcohol, glycerol and propylene glycol
  • glycerol ketals such as 2,2-dimethyl-l,3-dioxolane-4-methanol
  • ethers such as poly(ethylene glycol) 450
  • petroleum hydrocarbons such as mineral oil and petrolatum
  • water a pharmaceutically suitable surfactant, suspending agent or emulsifying agent; or mixtures thereof.
  • One or more of the components of the formulation can be present in its free base, free acid or pharmaceutically or analytically acceptable salt form.
  • pharmaceutically or analytically acceptable salt refers to a compound that has been modified by reacting it with an acid as needed to form an ionically bound pair.
  • acceptable salts include conventional non-toxic salts formed, for example, from non-toxic inorganic or organic acids. Suitable non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfonic, sulfamic, phosphoric, nitric and others known to those of ordinary skill in the art.
  • 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%.
  • Hard gelatin capsules (50 counts, 00 size) were filled with a liquid composition of Example 3. These capsules were manually filled with 800 mg of the formulation and then sealed by hand with a 50% ethanol/ 50% water solution. The capsules were then banded by hand with 22% gelatin solution containing the following ingredients in the amounts indicated.
  • 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.
  • a subj ect presenting with Zika virus 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 Zika 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
  • Method A is followed except that the subject is prescribed and administered one or more other therapeutic agents for the treatment of Zika virus 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.
  • 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.
  • 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 one cardiac glycosides 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
  • oleanolic acid and ursolic 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.
  • 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
  • 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.
  • 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
  • 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.
  • 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
  • Method A is followed except that the subject is prescribed and administered one or more other therapeutic agents for the treatment of Paramyxoviridae family 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.
  • 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

Abstract

A method of treating viral infection, such as viral infection caused by a virus of the Filoviridae, Flaviviridae or Togaviriade family, is provided. A composition having at least one cardiac glycoside is used to treat viral infection. The composition can futher include at least one triterpene.

Description

METHOD AND COMPOSITION FOR TREATING VIRAL INFECTION
FIELD OF THE INVENTION
[001] The present invention concerns an antiviral composition and its use for treating Flaviviridae infection, Togaviridae infection, or Filoviridae infection in mammals. Some embodiments concern treatment of hemorrhagic viral infection.
BACKGROUND OF THE INVENTION
[002] Nerium oleander, a member of the Nerium species, is an ornamental plant widely distributed in subtropical Asia, the southwestern 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.
[003] Extraction of components from plants of Nerium species has traditionally been carried out using boiling water, cold water, or organic solvent.
[004] ANVIRZEL™ (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. Herbal Pharmacotherapy, (2001) vol 1, pp.1-16). 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.
[005] Erdemoglu et al. (J. Ethnopharmacol. (2003) Nov. 89(1), 123-129) discloses results for the comparison of aqueous and ethanolic extracts of plants, including Nerium oleander, based upon their anti-nociceptive and anti-inflammatory activities. [006] Organic solvent extracts of Nerium oleander are also 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. 21(5), 46-47; alcoholic extract).
[007] A supercritical fluid extract of Nerium species is known (US 8394434, US 8187644, US 7402325) and has demonstrated efficacy in treating neurological disorders (US 8481086, US 9220778, US 9358293, US 20160243143A1) and cell-proliferative disorders (US 8367363).
[008] 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.
[0001] 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.
[0002] 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. Addington et al. (US 9011937, US 20150283191 Al) disclose a triterpene-containing fraction (PBI-04711) of the SCF extract of Nerium oleander containing oleandrin and triterpenes for the treatment of neurological conditions. Jager et al. (Molecules (2009), 14, 2016-2031) disclose various plant extracts containing mixtures of oleanolic acid, ursolic acid, betulinic acid and other components. Mishra et al. (PLoS One 2016 25;l l(7):e0159430. Epub 2016 Jul 25) disclose an extract oiBetula utilis bark containing a mixture of oleanolic acid, ursolic acid, betulinic acid and other components. Wang et al. (Molecules (2016), 21, 139) disclose an extract oiAlstonia scholaris containing a mixture of oleanolic acid, ursolic acid, betulinic acid and other components. L. e Silva et al. (Molecules (2012), 17, 12197) disclose an extract oiEriope blanchetti containing a mixture of oleanolic acid, ursolic acid, betulinic acid and other components. Rui et al. (Int. J. Mol. Sci. (2012), 13, 7648-7662) disclose an extract of Eucaplyptus globulus containing a mixture of oleanolic acid, ursolic acid, betulinic acid and other components. Ayatollahi et al. (Iran. J. Pharm. Res. (2011), 10(2), 287-294) disclose an extract of Euphorbia microsciadia containing a mixture of oleanolic acid, ursolic acid, betulinic acid and other components. Wu et al. (Molecules (2011), 16, 1-15) disclose an extract ofLigustrum species containing a mixture of oleanolic acid, ursolic acid, betulinic acid and other components. Lee et al. (Biol. Pharm. Bull (2010), 33(2), 330) disclose an extract of Forsythia viridissima containing a mixture of oleanolic acid, ursolic acid, betulinic acid and other components.
[009] 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). We (two of the instant inventors) previously reported (Van Kanegan et al, in Nature Scientific Reports (May 2016), 6:25626. doi: 10.1038/srep25626) on the contribution of the triterpenes toward efficacy by comparing their neuroprotective activity in a brain slice oxygen glucose deprivation (OGD) model assay at similar concentrations. We found that PBI-05204 (PBI) and PBI-04711 (Fraction 0-4) provide neuroprotective activity.
[0010] 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- 3β-ο1; 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,27-dihydroxy-12-oleanan-28-oic acid; and other components. [0011] Viral hemorrhagic fever (VHF) can be caused by five distinct virus families: Arenaviridae, Bunyaviridae, Filoviridae, Flaviviridae, and Paramyxoviridae. The Filoviruses, e.g. Ebolavirus (EBOV) and Marburgvirus (MARV), are among the most pathogenic viruses known to man and the causative agents of viral hemorrhagic fever outbreaks with fatality rates of up to 90%. Each virion contains one molecule of single- stranded, negative-sense RNA. Beyond supportive care or symptomatic treatment, there are no commercial therapeutically effective drugs and no prophylactic drugs available to treat EBOV (Eboval virus) and MARV (Marburg virus) infections, i.e. filovirus infections. Five species of Ebolavirus have been identified: Tai Forest (formerly Ivory Coast), Sudan, Zaire, Reston and Bundibugyo.
[0012] 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.
[0013] 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. Viruses exist against which oleandrin, oleanolic acid, ursolic acid and/or betulinic acid have little to no antiviral activity, meaning one cannot predic a priori whether oleandrin, oleanolic acid, ursolic acid and/or betulinic acid will exhibit antiviral activity against particular genuses of viruses.
[0014] 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.
[0015] Even though cardiac glycosides have been demonstrated to exhibit some antiviral activity against a few viruses, the specific compounds exhibit very different levels of antiviral activity against different viruses, meaning that some exhibit very poor antiviral activity and some exhibit better antiviral activity when evaluated against the same virus(es).
[0016] A need remains for improved pharmaceutical compositions containing oleandrin, oleanolic acid, ursolic acid, betulinic acid or any combination thereof that are therapeutically active against specific viral infections.
SUMMARY OF THE INVENTION
[0017] The invention provides a pharmaceutical composition and method for treating viral infection in a mammalian subject. The invention also provides a pharmaceutical composition and method for treating viral infection, e.g. Viral hemorrhagic fever (VHF) infection, in a mammalian subject. The invention also provides a method of treating viral infection in mammals by administration of the pharmaceutical composition. The inventors have succeeded in preparing antiviral compositions that exhibit sufficient antiviral activity to justify their use in treating viral infection in humans and animals. The inventors have developed corresponding treatment methods employing particular dosing regimens.
[0018] In some embodiments, the viral infection is caused by any of the following virus families: Arenaviridae, Bunyaviridae, Filoviridae, Flaviviridae, Paramyxoviridae, or Togaviridae.
[0019] Some embodiments of the invention are directed to compositions for and methods of treating Filovirus infection, Flavivirus infection, Henipavirus infection, alphavirus infection, or Togavirus infection. Viral infections that can be treated include, at least, Ebolavirus, Marburgvirus, Alphavirus, Flavivirus, Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, Zikavirus, Venezuelan Equine Encephalomyelitis (encephalitis) (VEE) virus, Chikungunya virus, Western Equine Encephalomyelitis (encephalitis) (WEE) virus, Eastern Equine Encephalomyelitis (encephalitis) (EEE) virus, Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Omsk Hemorrhagic Fever, Hendra virus, Nipah virus, and species thereof. [0020] Some embodiments of the invention are directed to compositions for and methods of treating viral infections from viruses of the Filoviridae family, Flavivirudae family, Paramyxoviridae family, or Togaviridae family.
[0021] 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.
[0022] In some embodiments, the invention provides an antiviral composition comprising (consisting essentially of): a) specific cardiac glycoside(s); b) plural triterpenes; or c) a combination of specific cardiac glycoside(s) and plural triterpenes.
[0023] 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.
[0024] Accordingly, 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.
[0025] The invention also provides a method of treating viral infection in a subject in need thereof, the method comprising:
determining whether or not the subject has a viral infection;
indicating administration of antiviral composition;
administering an initial dose of antiviral composition to the subject according to a prescribed initial dosing regimen for a period of time;
periodically determining the adequacy of subject's clinical response and/or therapeutic response to treatment with antiviral composition; and
if 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.
[0026] 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.
[0027] The individual steps of the methods of the invention can be conducted at separate facilities or within the same facility.
[0028] 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;
[0029] In some embodiments, the subject is administered 140 microg to 315 microg per day of cardiac glycoside. In some embodiments, 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.
[0030] 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.
[0031] If present in the antiviral composition, the cardiac glycoside is preferabley oleandrin but can also include odoroside, neritaloside, or oleandrigenin. In some embodiments, 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. In some embodiments, 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. In some embodiments, the triterpene is selected from the group consisting of oleanolic acid, ursolic acid, betulinic acid and salts or derivatives thereof.
[0032] Some embodiments of the invention include those wherein a pharmaceutical composition comprises at least one pharmaceutical excipient and the antiviral composition. In some embodiments, 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. As used herein, the generic terms triterpene and cardiac glycoside also encompass salts and derivatives thereof, unless otherwise specified.
[0033] The cardiac glycoside can be present in a pharmaceutical composition in pure form or as part of an extract containing one or more cardiac glycosides. The triterpene(s) can be present in a pharmaceutical composition in pure form or as part of an extract containing triterpene(s). In some embodiments, the cardiac glycoside is present as the primary therapeutic component, meaning the component primarily responsible for antiviral activity, in the pharmaceutical composition. In some embodiments, the triterpene(s) is/are present as the primary therapeutic component(s), meaning the component(s) primarily responsible for antiviral activity, in the pharmaceutical composition.
[0034] In some embodiments, an extract comprising the antiviral composition is obtained by extraction of plant material. The extract can comprise a hot-water extract, cold- water extract, supercritical fluid (SCF) extract, organic solvent extract, or combination thereof of the plant material. In some embodiments, the plant material is Nerium species or Thevetia species plant mass. Particular species include Nerium oleander or Thevetia nerifolia. In some embodiments, the extract comprises at least one other pharmacologically active agent, obtained along with the cardiac glycoside during extraction, that contributes to the therapeutic efficacy of the cardiac glycoside when the extract is administered to a subject. In some embodiments, the composition further comprises one or more other non- cardiac glycoside therapeutically effective agents, i.e. one or more agents that are not cardiac glycosides. In some embodiments, the composition further comprises one or more antiviral compound(s). In some embodiments, the antiviral composition excludes a pharmacologically active polysaccharide.
[0035] In some embodiments, 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. Accordingly, 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.
[0036] In some embodiments, 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.
[0037] In some embodiments, the molar ratio of total triterpene content (OA + UA + BA) to oleandrin ranges from about 15: 1 to about 5: 1, or about 12: 1 to about 8: 1, or about 100: 1 to about 15: 1, or about 100: 1 to about 50: 1, or about 100: 1 to about 75: 1, or about 100: 1 to about 80: 1, or about 100: 1 to about 90: 1, or about 10: 1.
[0038] In some embodiments, 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). [0039] In some embodiments, the other therapeutic agent is not a polysaccharide obtained during preparation of the extract, meaning it is not an acidic homopolygalacturonan or arabinogalaturonan. In some embodiments, the extract excludes another therapeutic agent and/or excludes an acidic homopolygalacturonan or arabinogalaturonan obtained during preparation of the extract.
[0040] The invention also provides use of a cardiac glycoside in the manufacture of a medicament for the treatment of viral infection in a subject. In some embodiments, 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. In some embodiments, the manufacture can be conducted as described in PCT International Application No. PCT/US06/29061. The manufacture can also include one or more additional steps such as: delivering the packaged dosage form to a vendor (retailer, wholesaler and/or distributor); selling or otherwise providing the packaged dosage form to a subj ect having a viral infection; including with the medicament a label and a package insert, which provides instructions on use, dosing regimen, administration, content and toxicology profile of the dosage form. In some embodiments, the treatment of viral infection comprises: determining that a subject has a viral infection; indicating administration of pharmaceutical dosage form to the subject according to a dosing regimen; administering to the subject one or more pharmaceutical dosage forms, wherein the one or more pharmaceutical dosage forms is administered according to the dosing regimen.
[0041] 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.
[0042] 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. [0043] 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.
[0044] In some embodiments, the water miscible solvent is low molecular weight (less than 6000) PEG, glycol, or alcohol. In some embodiments, the surfactant is a pegylated surfactant, meaning a surfactant comprising a poly(ethylene glycol) functional group.
[0045] The invention includes all combinations of the aspects, embodiments and sub-embodiments of the invention disclosed herein.
BRIEF DESCRIPTION OF THE FIGURES
[0046] The following figures form part of the present description and describe exemplary embodiments of the claimed invention. The skilled artisan will, in light of these figures and the description herein, be able to practice the invention without undue experimentation.
[0047] FIGS. 1-2 depict charts summarizing the in vitro dose response antiviral activity of various compositions against Ebolavirus.
[0048] FIGS. 3-4 depict charts summarizing the in vitro dose response antiviral activity of various compositions against Marburgvirus.
[0049] FIG. 5 depicts a chart summarizing the in vitro dose response antiviral activity of oleandrin against Zikavirus (SIKV strain PRVABC59) in Vero E6 cells.
[0050] FIG. 6 depicts a chart summarizing the in vitro dose response antiviral activity of digoxin against Zikavirus (SIKV strain PRVABC59) in Vero E6 cells.
[0051] 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.
[0052] 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.
[0053] 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).
[0054] 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. [0055] FIGS. 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.
[0056] FIGS. 12A and 12B depict charts summarizing the ability of compositions
(oleandrin and PBI-05204) to inhibit the product of infectious progeny by virally infected
Vero E6 cells having been exposed to oleandrin: FIG. 12A- Ebolavirus; FIG. 12B-
Marburgvirus.
[0057] FIGS. 13A and 13B depict charts summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against Venezuelen Equine Encephalomyelits virus (FIG. 13 A) and Western Equine Encephalomyelitis virus (FIG. 13B) in Vero E6 cells.
DETAILED DESCRIPTION OF THE INVENTION
[0058] 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.
[0059] As used herein, 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.
[0060] As used herein, 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. In some embodiments, the subject is a female, a female capable of getting pregnant, or a pregnant female.
[0061] A subject treated according to the invention will exhibit a therapeutic response. By "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.
[0062] As used herein, "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".
[0063] 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 will vary from subject to subject according to a variety of established pharmacologic, pharmacodynamic and pharmacokinetic principles. However, a therapeutically relevant dose (relative, for example, to oleandrin) will typically not exceed 25 micrograms, 100 micrograms, 250 micrograms, 500 micrograms or 750 micrograms of cardiac glycoside/day or it can be in the range of 25-750 micrograms of cardiac glycoside per dose. It is known in the art that the actual amount of antiviral composition required to provide a target therapeutic result in a subject may vary from subject to subject according to the basic principles of pharmacy.
[0064] 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. For example, a therapeutically relevant dose can be administered one or more times daily for one or more weeks.
[0065] Example 15 provides a detailed description of an in vitro assay used to evaluate the efficacy of compositions containing oleandrin (as sole active), Anvirzel and PBI-05204 (supercritical fluid (SCF) extract ofNerium oleander) for the treatment of Ebolavirus (FIGS. 1-2) and Marburgvirus (FIGS. 3-4) infection, both of which are Filoviruses.
[0066] The experiments were set up by adding the compositions to cells at 40 microg/mL, then adding virus and incubating for lhr. Upon addition of the virus to the cells, the final concentration of the compositions is 20 microg/mL. 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. Anvirzel, the hot water extract of Nerium oleander, is more efficacious than OL on its own. Both extracts clearly exhibit efficacy in the nanomolar range. The percentage of oleandrin in the PBI-05204 extract (1.74%) is higher than in Anvirzel (0.459%, 4.59 microg/mg). At the highest dose of PBI-05204, it completely inhibited EBOV and MARV infection, whereas Anvirzel did not exhibit complete inhibition, because at a dose higher than 20 microg/mL with anvirzel, toxicity is observed. The data demonstrate highest antiviral activity against Ebolavirus and Marburgvirus for PBI-05204. The combination of triterpenes in PBI-05204 increased the antiviral activity of oleandrin.
[0067] Example 6 provides a detailed description of an in vitro assay used to evaluate the efficacy of the cardiac glycosides for the treatment of Zikavirus (a flavivirus) infection. Vero E6 cells were infected with Zika virus (ZIKV strain PRVABC59) at an MOI of 0.2 in the presence of oleandrin (FIG. 5) or digoxin (FIG. 6). The cells were incubated with virus and the cardiac glycoside for 1 hr, after which the inoculum and non-absorbed cardiac glycoside (if any) was removed. The cells were immersed in fresh medium and incubated for 48 hr, after which they were fixed with formalin and stained for ZIKV infection. The data demonstrate antiviral activity against Zikavirus for both cardiac glycosides; however, oleandrin exhibited higher (almost 8-fold greater) antiviral activity than digoxin.
[0068] Example 14 provides a detailed description of an assay used to evaluate the antiviral activity of test compositions against Zikavirus and Dengue virus. The data indicate that oleandrin demonstrates efficacy against Zikavirus and Dengue virus.
[0069] 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). For FIGS. 7-8, the host cells were exposed to the compositions prior to infection with virus. Vero E6 cells were infected with EBOV/Kik (FIG. 7, MOI=l) or MARV/Ci67 (FIG. 8, MOI=l) in the presence of oleandrin, digoxin or PBI-05204, an oleandrin-containing plant extract. After lhr, inoculum and compounds were removed and fresh medium added to cells. 48hr later, cells were fixed and immunostained to detect cells infected with EBOV or MARV. Infected cells were enumerated using an Operetta.
[0070] In order to ensure that false positives, in terms of antiviral activity, were not being observed, cellular viability in the presence of the compositions was tested. For the data in FIG. 9, Vero E6 cells were treated with compound as above. ATP levels were measured by CellTiter-Glo as a measurement of cell viability. It was determined that oleandrin, digoxin, and PBI-05204 did not reduce cellular viability, meaning that the antiviral activity detailed in other figures herein is not due to false positives caused by cellular toxicity of the individual compounds. [0071] Accordingly, 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.
[0072] The antiviral composition and method of the invention are also useful in treating viral infection that has occurred prior to administration of the antiviral composition. Vero E6 cells were infected with EBOV (FIGS. 10A, 10B) or MARV (FIGS. 11 A, 1 IB). At 2hr post-infection (FIGS. 10A, 11 A) or 24hr post-infection (FIGS. 10B, 11B), oleandrin or PBI- 05204 was added to cells for lhr, then discarded and cells were returned to culture medium.
[0073] 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. When 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. 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. When 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.
[0074] Given that the antiviral activity of the composition herein is reduced for a single generation of virus-infected cells, e.g. within 24 hours post-infection, we evaluated whether the antiviral composition is capable of inhibiting viral propagation, meaning inhibiting production of infectious progeny. 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.
[0075] Accordingly, 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).
[0076] Antiviral activity of the antiviral composition against Togaviridae alphavirus was evaluated using VEE virus and WEE virus in Vero E6 cells. FIGS. 13A and 13B depict charts summarizing the in vitro dose response antiviral activity of various compositions (oleandrin, digoxin and PBI-05204) against Venezuelen Equine Encephalomyelits virus (FIG. 13A) and Western Equine Encephalomyelitis virus (FIG. 13B) in Vero E6 cells. Vero E6 cells were infected with Venezuelan equine encephalitis virus (FIG. 13A, MOI=0.01) or Western equine encephalitis virus (FIG. 13B, MOI=0.1) for 18hr in the presence or absence of indicated compounds. Infected cells were detected as before and enumerated on an Operetta. The antiviral composition of the invention was found to be efficacious.
[0077] Accordingly, 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.
[0078] Antiviral activity of the compositions herein was evaluated against rhinovirus 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.
[0079] PBI-05204 (as described herein and in 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) comprises cardiac glycoside (oleandrin, OL) and triterpenes (oleanolic acid (OA), ursolic acid (UA) and betulinic acid (BA)) as the primary pharmacologically active components. The molar ratio of OL to total triterpene is about l :(10-96). The molar ratio of OA:UA:BA is about 7.8:7.4: 1. The combination of OA, UA and BA in PBI-05204 increases the antiviral activity of oleandrin when compared on an OL equimolar basis. 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. In some embodiments, 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).
[0080] Antiviral compositions comprising oleandrin as the sole antiviral agent are within the scope of the invention.
[0081] Antiviral compositions comprising oleandrin and plural triterpenes as the antiviral agents are within the scope of the invention. In some embodiments, 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.
[0082] 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. As noted above, PBI-04711 comprises OA, UA and BA as the primary active ingredients, and it exhibits antiviral activity. In some embodiments, 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.
[0083] PBI-01011 is an improved triterpene-based antiviral composition comprising OA, UA and BA, wherein the molar ratio of OA:UA:BA is about 9-12 : up to about 2 : up to about 2, or about 10 : about 1 : about 1, or about 9-12 : about 0.1-2 : about 0.1-2, or about 9-11 : about 0.5-1.5 : about 0.5-1.5, or about 9.5-10.5 : about 0.75-1.25 : about 0.75-1.25, or about 9.5-10.5 : about 0.8-1.2 : about 0.8-1.2, or about 9.75-10.5 : about 0.9-1.1 : about 0.9-1.1.
[0084] In some embodiments, 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.
[0085] In some embodiments, 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.
[0086] In some embodiments, an antiviral composition comprises at least 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) present at a molar ratio of OA to UA to BA as described herein. OA is present in large molar excess over both UA and BA.
[0087] In some embodiments, a triterpene-based antiviral composition excludes cardiac glycoside.
[0088] In general, a subject having Filoviridae infection, Flaviviridae infection or Togaviridae infection is treated as follows. The subject is evaluated to determine whether said subject is infected with said virus. Administration of antiviral composition is indicated. Initial doses of antiviral composition are administered to the subject according to a prescribed dosing regimen for a period of time (a treatment period). The subject's clinical response and level of therapeutic response are determined periodically. If the level of therapeutic response is too low at one dose, then the dose is escalated according to a predetermine 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. The dose or dosing regimen can be adjusted as needed until the patient reaches the desired clinical endpoint(s) such as cessation of the infection itself, reduction in infection- associated symptoms, and/or a reduction in the progression of the infection.
[0089] If a clinician intends to treat a subject having viral infection with a combination of a antiviral composition and one or more other therapeutic agents, and it is known that the viral infection, which the subject has, is at least partially therapeutically responsive to treatment with said one or more other therapeutic agents, then 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. In some embodiments, the first and second dosing regimens are the same. In some embodiments, the first and second dosing regimens are different.
[0090] 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. 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.
[0091] 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 Ministries of Health worldwide.
[0092] Example 5 provides an exemplary procedure for the treatment of Zikavirus infection in a mammal. Example 12 provides an exemplary procedure for the treatment of Filovirus infection (Ebolavirus, Marburgvirus) in a mammal. Example 13 provides an exemplary procedure for the treatment of Flavivirus infection (Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, Zikavirus, Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Omsk Hemorrhagic Fever, Powassan virus infection) in a mammal.
[0093] The antiviral compound(s) (triterpene(s), cardiac glycoside(s), etc.) present in the pharmaceutical composition can be present in their unmodified form, salt form, derivative form or a combination thereof. As used herein, the term "derivative" is taken to mean: a) a chemical substance that is related structurally to a first chemical substance and theoretically derivable from it; b) a compound that is formed from a similar first compound or a compound that can be imagined to arise from another first compound, if one atom of the first compound is replaced with another atom or group of atoms; c) a compound derived or obtained from a parent compound and containing essential elements of the parent compound; or d) a chemical compound that may be produced from first compound of similar structure in one or more steps. For example, a derivative may include a deuterated form, oxidized form, dehydrated, unsaturated, polymer conjugated or glycosilated form thereof or may include an ester, amide, lactone, homolog, ether, thioether, cyano, amino, alkylamino, sulfhydryl, heterocyclic, heterocyclic ring-fused, polymerized, pegylated, benzylidenyl, triazolyl, piperazinyl or deuterated form thereof.
[0094] As used herein, the term "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.
[0095] 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.
[0096] Other extracts containing cardiac glycoside, especially oleandrin, can be prepared by various different processes. An extract can be prepared according to the process developed by Dr. Huseyin Ziya Ozel (U.S. Patent No. 5,135,745) describes a procedure for the preparation of a hot water extract. The aqueous extract reportedly contains several polysaccharides with molecular weights varying from 2KD to 30KD, oleandrin and oleandrigenin, odoroside and neritaloside. The polysaccharides reportedly include acidic homopolygalacturonans or arabinogalaturonans. U.S. Patent No. 5,869,060 to Selvaraj et al. discloses hot water extracts of Nerium species and methods of production thereof, e.g. Example 2. 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. Erdemoglu et al. (J. Ethnopharmacol. (2003) Nov. 89(1), 123-129) discloses results for the comparison of aqueous and ethanolic extracts of plants, including Nerium oleander, based upon their anti-nociceptive and antiinflammatory activities. 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. 21(5), 46-47; alcoholic extract). U.S. Pregrant Patent Application Publication No. 20040247660 to Singh et al. discloses the preparation of a protein stabilized liposomal formulation of oleandrin for use in the treatment of cancer. U. S. Pregrant Patent Application Publication No. 20050026849 to Singh et al. discloses a water soluble formulation of oleandrin containing a cyclodextrin. U.S. Pregrant Patent Application Publication No. 20040082521 to Singh et al. discloses the preparation of protein stabilized nanoparticle formulations of oleandrin from the hot-water extract.
[0097] The extracts also differ in their polysaccharide and carbohydrate content. The hot water extract contains 407.3 glucose equivalent units of carbohydrate relative to a standard curve prepared with glucose while analysis of the SCF CCh extract found carbohydrate levels that were found in very low levels that were below the limit of quantitation. The amount of carbohydrate in the hot water extract of Nerium oleander was, however, at least 100-fold greater than that in the SCF CCh extract. The polysaccharide content of the SCF extract can be 0%, <0.5%, <0.1%, <0.05%, or <0.01% wt. In some embodiments, the SCF extract excludes polysaccharide obtained during extraction of the plant mass.
Figure imgf000024_0001
[0098] The partial compositions of the SCF CCh extract and hot water extract were determined by DART TOF-MS (Direct Analysis in Real Time Time of Flight Mass Spectrometry) on a JEOL AccuTOF-DART mass spectrometer (JEOL USA, Peabody, MA, USA).
[0099] The SCF extract of Nerium species or Thevetia species is a mixture of pharmacologically active compounds, such as oleandrin and triterpenes. The extract obtained by the SCF process is a substantially water-insoluble, viscous semi-solid (after solvent is removed) at ambient temperature. The SCF extract comprises many different components possessing a variety of different ranges of water solubility. The extract from a supercritical fluid process contains by weight a theoretical range of 0.9% to 2.5% wt of oleandrin or 1.7% to 2.1% wt of oleandrin or 1.7% to 2.0% wt of oleandrin. SCF extracts comprising varying amount of oleandrin have been obtained. In one embodiment, 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.
[00100] 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. For example, a batch of SCF extract (PBI-05204) was analyzed for these four components and found to contain the following approximate amounts of each.
Figure imgf000025_0001
WRT denotes "with respect to".
[00101] The content of the individual components may vary by ±25%, ±20%, ±15%, ±10% or ±5% relative to the values indicated. Accordingly, the content of oleandrin in the SCF extract would be in the range of 20 mg ± 5 mg (which is ±25% of 20 mg) per mg of SCF extract.
[00102] 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).
[00103] As used herein, the individually named 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.
[00104] 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. 6, 2014, US 20130317007 Al to Anderson et al. which published Nov. 28, 2013, US 20130303607 Al to Gribble et al. which published Nov. 14, 2013, US 20120245374 to Anderson et al. which published Sep. 27, 2012, US 20120238767 Al to Jiang et al. which published Sep. 20, 2012, US 20120237629 Al to Shode et al. which published Sept. 20, 2012, US 20120214814 Al to Anderson et al. which published Aug. 23, 2012, US 20120165279 Al to Lee et al. which published June 28, 2012, US 20110294752 Al to Amtzen et al. which published Dec. 1, 2011, US 20110091398 Al to Majeed et al. which published April 21, 2011, US 20100189824 Al to Amtzen et al. which published July 29, 2010, US 20100048911 Al to Jiang et al. which published Feb. 25, 2010, and US 20060073222 Al to Arntzen et al. which published April 6, 2006, the entire disclosures of which are hereby incorporated by reference.
[00105] 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.
[00106] Betulinic 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 20120237629 Al to Shode et al. which published Sept. 20, 2012, US 20170204133 Al to Regueiro-Ren et al. which published July 20, 2017, US 20170096446 Al to Nitz et al. which published April 6, 2017, US 20150337004 Al to Parthasaradhi Reddy et al. which published Nov. 26, 2015, US 20150119373 Al to Parthasaradhi Reddy et al. which published April 30, 2015, US 20140296546 Al to Yan et al. which published Oct. 2, 2014, US 20140243298 Al to Swidorski et al. which published Aug. 28, 2014, US 20140221328 Al to Parthasaradhi Reddy et al. which published Aug. 7, 2014, US 20140066416 Al tp Leunis et al. which published March 6, 2014, US 20130065868 Al to Durst et al. which published March 14, 2013, US 20130029954 Al to Regueiro-Ren et al. which published Jan. 31, 2013, US 20120302530 Al to Zhang et al. which published Nov. 29, 2012, US 20120214775 Al to Power et al. which published Aug. 23, 2012, US 20120101149 Al to Honda et al. which published April 26, 2012, US 20110224182 to Bullock et al. which published Sep. 15, 2011, US 20110313191 Al to Hemp et al. which published Dec. 22, 2011, US 20110224159 Al to Pichette et al. which published Sep. 15, 2011, US 20110218204 to Parthasaradhi Reddy et al. which published Sep. 8, 2011, US 20090203661 Al to Safe et al. which published Aug. 13, 2009, US 20090131714 Al to Krasutsky et al. which published May 21, 2009, US 20090076290 to Krasutsky et al. which published March 19, 2009, US 20090068257 Al to Leunis et al. which published March 12, 2009, US 20080293682 to Mukherjee et al. which published Nov. 27, 2008, US 20070072835 Al to Pezzuto et al. which published March 29, 2007, US 20060252733 Al to Jansen et al. which published Nov. 9, 2006, and US 2006025274 Al to O'Neill et al. which published Nov. 9, 2006, the entire disclosures of which are hereby incorporated by reference.
[00107] 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.
[00108] 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. (Development and evaluation of optimized sucrose ester stabilized oleanolic acid nanosuspensions prepared by wet ball milling with design of experiments" in Biol. Pharm. Bull. (2014), 37(6), 926-937), Zhang et al. ("Enhancement of oral bioavailability of triterpene through lipid nanospheres: preparation, characterization, and absorption evaluation" in J. Pharm. Sci. (June 2014), 103(6), 1711-1719), Godugu et al. ("Approaches to improve the oral bioavailability and effects of novel anticancer drugs berberine and betulinic acid" in PLoS One (Mar 2014), 9(3):e89919), Zhao et al. ("Preparation and characterization of betulin nanoparticles for oral hypoglycemic drug by antisolvent precipitation" in Drug Deliv. (Sep 2014), 21(6), 467-479), Yang et al. ("Physicochemical properties and oral bioavailability of ursolic acid nanoparticles using supercritical anti-solvent (SAS) process" in Food Chem. (May 2012), 132(1), 319-325), Cao et al. ("Ethylene glycol-linked amino acid diester prodrugs of oleanolic acid for PEPT1- mediated transport: synthesis, intestinal permeability and pharmacokinetics" in Mol. Pharm. (Aug. 2012), 9(8), 2127-2135), Li et al. ("Formulation, biological and pharmacokinetic studies of sucrose ester-stabilized nanosuspensions of oleanolic acid" in Pharm. Res. (Aug 2011), 28(8), 2020-2033), Tong et al. ("Spray freeze drying with polyvinylpyrrolidone and sodium caprate for improved dissolution and oral bioavailablity of oleanolic acid, a BCS Class IV compound" in Int. J. Pharm. (Feb 2011), 404(1-2), 148-158), Xi et al. (Formulation development and bioavailability evaluation of a self-nanoemulsified drug delivery system of oleanolic acid" in AAPS PharmSciTech (2009), 10(1), 172-182), Chen et al. ("Oleanolic acid nanosuspensions: preparation, in-vitro characterization and enhanced hepatoprotective effect" in J. Pharm. Pharmacol. (Feb 2005), 57(2), 259-264), the entire disclosures of which are hereby incorporated by reference.
[00109] 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.
[00110] An effective amount or therapeutically relevant amount of antiviral compound (cardiac glycoside, triterpene or combinations thereof) is specifically contemplated. By the term "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.
[00111] 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.
[00112] The cardiac glycoside can be present in a dosage form in an amount sufficient to provide a subject with an initial dose of oleandrin of 20 to 100 microg, 12 microg to 300 microg, or 12 microg to 120 microg. A dosage form can comprise 20 of oleandrin to 100 microg, 0.01 microg to 100 mg or 0.01 microg to 100 microg oleandrin, oleandrin extract or extract of Nerium oleander containing oleandrin.
[00113] 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. [00114] 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.
[00115] 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.
[00116] 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. Composition Oleandrin Oleanolic Ursolic Betulinic Suitable (moles) acid acid acid dose
(moles) (moles) (moles)
A 0.5-1.5 4-6 0.05 to 0.5
mg/kg/day
B 0.5-1.5 4-6 4-6 0.05 to 0.35 mg/kg/day
C 0.5-1.5 4-6 4-6 0.1-1 0.05 to 0.22
(PBI-05204) mg/kg/day
D 0.5-1.5 4-6 0.05 to 0.4
mg/kg/day
E 0.5-1.5 0.1-1 0.05 to 0.4
mg/kg/day
AA About 1 0.3-0.7 0.05 to 0.4
mg/kg/day
AB About 1 About 4.7 0.05 to 0.5
mg/kg/day
AC About 1 About 4.7 About 4.5 0.05 to 0.4
mg/kg/day
AD About 1 About 4.7 About 4.5 About 0.6 0.05 to 0.22
(PBI-05204) mg/kg/day
AE About 1 About 4.5 0.05 to 0.4
mg/kg/day
AF About 1 About 0.6 0.05 to 0.3
mg/kg/day
All values are approximate, meaning "about" the specifiec value.
[00117] It should be noted that a compound herein might possess one or more functions in a composition or formulation of the invention. For example, a compound might serve as both a surfactant and a water miscible solvent or as both a surfactant and a water immiscible solvent.
[00118] A liquid composition can comprise one or more pharmaceutically acceptable liquid carriers. The liquid carrier can be an aqueous, non-aqueous, polar, non-polar, and/or organic carrier. Liquid carriers include, by way of example and without limitation, a water miscible solvent, water immiscible solvent, water, buffer and mixtures thereof.
[00119] As used herein, the terms "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, Ν,Ν-dimethylacetamide, pyridine, propanol, N- methylacetamide, butanol, soluphor (2-pyrrolidone), pharmasolve (N-methyl-2- pyrrolidone).
[00120] As used herein, the terms "water insoluble solvent" or "water immiscible solvent", which terms are used interchangeably, refer to an organic liquid which forms a biphasic mixture with water or provides a phase separation when the concentration of solvent in water exceeds five percent. The solvent is suitable for administration to humans or animals. Exemplary water insoluble solvents include, by way of example and without limitation, medium/long chain triglycerides, oil, castor oil, corn oil, vitamin E, vitamin E derivative, oleic acid, fatty acid, olive oil, softisan 645 (Diglyceryl Caprylate / Caprate / Stearate / Hydroxy stearate adipate), miglyol, captex (Captex 350: Glyceryl Tricaprylate/ Caprate/ Laurate triglyceride; Captex 355: Glyceryl Tricaprylate/ Caprate triglyceride; Captex 355 EP / NF: Glyceryl Tricaprylate/ Caprate medium chain triglyceride).
[00121] Suitable solvents are listed in the "International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH) guidance for industry Q3C Impurities: Residual Solvents " (1997), which makes recommendations as to what amounts of residual solvents are considered safe in pharmaceuticals. Exemplary solvents are listed as class 2 or class 3 solvents. 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.
[00122] Other materials that can be used as water immiscible solvents in the invention include: Captex 100: Propylene Glycol Dicaprate; Captex 200: Propylene Glycol Dicaprylate/ Dicaprate; Captex 200 P: Propylene Glycol Dicaprylate/ Dicaprate; Propylene Glycol Dicaprylocaprate; Captex 300: Glyceryl Tricaprylate/ Caprate; Captex 300 EP / NF: Glyceryl Tricaprylate/ Caprate Medium Chain Triglycerides; Captex 350: Glyceryl Tricaprylate/ Caprate/ Laurate; Captex 355: Glyceryl Tricaprylate/ Caprate; Captex 355 EP / NF: Glyceryl Tricaprylate/ Caprate Medium Chain Triglycerides; Captex 500: Triacetin; Captex 500 P: Triacetin (Pharmaceutical Grade); Captex 800: Propylene Glycol Di (2- Ethythexanoate); Captex 810 D: Glyceryl Tricaprylate/ Caprate/ Linoleate; Captex 1000: Glyceryl Tricaprate; Captex CA: Medium Chain Triglycerides; Captex MCT-170: Medium Chain Triglycerides; Capmul GMO: Glyceryl Monooleate; Capmul GMO-50 EP/NF: Glyceryl Monooleate; Capmul MCM: Medium Chain Mono- & Diglycerides; Capmul MCM C8: Glyceryl Monocaprylate; Capmul MCM CIO: Glyceryl Monocaprate; Capmul PG-8: Propylene Glycol Monocaprylate; Capmul PG-12: Propylene Glycol Monolaurate; Caprol 10G10O: Decaglycerol Decaoleate; Caprol 3GO: Triglycerol Monooleate; Caprol ET: Polyglycerol Ester of Mixed Fatty Acids; Caprol MPGO: Hexaglycerol Dioleate; Caprol PGE 860: Decaglycerol Mono-, Dioleate.
[00123] As used herein, 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.
[00124] 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.
[00125] Similarly, the hydrophobic surfactant can be any hydrophobic surfactant suitable for use in pharmaceutical compositions. In general, suitable hydrophobic surfactants will have an HLB value less than about 10. Mixtures of hydrophobic surfactants are also within the scope of the invention. [00126] Examples of additional suitable solubilizer include: alcohols and polyols, such as ethanol, isopropanol, butanol, benzyl alcohol, ethylene glycol, propylene glycol, butanediols and isomers thereof, glycerol, pentaerythritol, sorbitol, mannitol, transcutol, dimethyl isosorbide, polyethylene glycol, polypropylene glycol, polyvinylalcohol, hydroxypropyl methylcellulose and other cellulose derivatives, cyclodextrins and cyclodextrin derivatives; ethers of polyethylene glycols having an average molecular weight of about 200 to about 6000, such as tetrahydrofurfuryl alcohol PEG ether (glycofurol, available commercially from BASF under the trade name Tetraglycol) or methoxy PEG (Union Carbide); amides, such as 2-pyrrolidone, 2-piperidone, caprolactam, N- alkylpyrrolidone, N-hydroxyalkylpyrrolidone, N-alkylpiperidone, N-alkylcaprolactam, dimethylacetamide, and polyvinypyrrolidone; esters, such as ethyl propionate, tributylcitrate,acetyl triethylcitrate, acetyl tributyl citrate, triethylcitrate, ethyl oleate, ethyl caprylate, ethyl butyrate, triacetin, propylene glycol monoacetate, propylene glycol diacetate, caprolactone and isomers thereof, valerolactone and isomers thereof, butyrolactone and isomers thereof; and other solubilizers known in the art, such as dimethyl acetamide, dimethyl isosorbide (Arlasolve DMI (ICI)), N-methyl pyrrolidones (Pharmasolve (ISP)), monooctanoin, diethylene glycol nonoethyl ether (available from Gattefosse under the trade name Transcutol), and water. Mixtures of solubilizers are also within the scope of the invention.
[00127] Except as indicated, compounds mentioned herein are readily available from standard commercial sources.
[00128] Although not necessary, the 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.
[00129] The composition of the invention can also include oils such as fixed oils, peanut oil, sesame oil, cottonseed oil, corn oil and olive oil; fatty acids such as oleic acid, stearic acid and isostearic acid; and fatty acid esters such as ethyl oleate, isopropyl myristate, fatty acid glycerides and acetylated fatty acid glycerides. The composition can also include alcohol such as ethanol, isopropanol, hexadecyl alcohol, glycerol and propylene glycol; glycerol ketals such as 2,2-dimethyl-l,3-dioxolane-4-methanol; ethers such as poly(ethylene glycol) 450; petroleum hydrocarbons such as mineral oil and petrolatum; water; a pharmaceutically suitable surfactant, suspending agent or emulsifying agent; or mixtures thereof.
[00130] It should be understood that the compounds used in the art of pharmaceutical formulation generally serve a variety of functions or purposes. Thus, if a compound named herein is mentioned only once or is used to define more than one term herein, its purpose or function should not be construed as being limited solely to that named purpose(s) or function(s).
[00131] One or more of the components of the formulation can be present in its free base, free acid or pharmaceutically or analytically acceptable salt form. As used herein, "pharmaceutically or analytically acceptable salt" refers to a compound that has been modified by reacting it with an acid as needed to form an ionically bound pair. Examples of acceptable salts include conventional non-toxic salts formed, for example, from non-toxic inorganic or organic acids. Suitable non-toxic salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfonic, sulfamic, phosphoric, nitric and others known to those of ordinary skill in the art. 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. On the other hand, where the pharmacologically active ingredient possesses an acid functional group, a pharmaceutically acceptable base is added to form the pharmaceutically acceptable salt. Lists of other suitable salts are found in Remington 's Pharmaceutical Sciences, 17th. ed., Mack Publishing Company, Easton, PA, 1985, p. 1418, the relevant disclosure of which is hereby incorporated by reference.
[00132] The phrase "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.
[00133] 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.
[00134] 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.
[00135] The composition of the invention can be included in any dosage form. Particular 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.
[00136] In view of the above description and the examples below, one of ordinary skill in the art will be able to practice the invention as claimed without undue experimentation. The foregoing will be better understood with reference to the following examples that detail certain procedures for the preparation of embodiments of the present invention. All references made to these examples are for the purposes of illustration. The following examples should not be considered exhaustive, but merely illustrative of only a few of the many embodiments contemplated by the present invention.
Example 1
Supercritical fluid extraction of powdered oleander leaves
Method A. With carbon dioxide.
[00137] 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.
[00138] 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.
[00139] 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. For an exact yield determination, 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. Based on the weight of the starting material, 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%.
Method B. With mixture of carbon dioxide and ethanol
[00140] 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.
[00141] 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.
[00142] 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%.
Example 2
Hot-water extraction of powdered oleander leaves,
(comparative example)
[00143] 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.
[00144] 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%.
Example 3
Preparation of pharmaceutical compositions.
Method A. Cremopho -based drug delivery system
[00145] The following ingredients were provided in the amounts indicated.
Figure imgf000038_0001
[00146] The excipients were dispensed into ajar and shook in a New Brunswick Scientific C24KC Refrigerated Incubator shaker for 24 hours at 60°C to ensure homogeneity. The samples were then pulled and visually inspected for solubilization. Both the excipients and antiviral composition were totally dissolved for all formulations after 24 hours.
Method B. GMO /Cremophor -based drug delivery system
[00147] The following ingredients were provided in the amounts indicated.
Reagent Percent of Formulation
Name Function (% w/w)
antiviral composition Active agent 4.7
Vitamin E Antioxidant 0.1
Labrasol Surfactant 8.5
Ethanol Co-solvent 7.6
Cremophor EL Surfactant 56.1
Glycerol Monooleate Surfactant 23.2 [00148] The procedure of Method A was followed.
Method C. Labrasol-based drug delivery system
[00149] The following ingredients were provided in the amounts indicated.
Figure imgf000039_0001
[00150] The procedure of Method A was followed.
Method D. Vitamin E-TPGS based micelle forming system
[00151] The following ingredients were provided in the amounts indicated.
Figure imgf000039_0002
[00152] The procedure of Method A was followed.
Method E. Multi-component drug delivery system
[00153] The following ingredients were provided in the amounts indicated.
Component Weight (g) Weight % (w/w)
Vitamin E 10.0 1.0
Cremophor ELP 580.4 55.9
Labrasol 89.0 8.6
Glycerol Monooleate 241.0 23.2
Ethanol 80.0 7.7
antiviral composition 38.5 3.7
Total 1038.9 100 [00154] The procedure of Method A was followed.
Method F. Multi-component drug delivery system
[00155] The following ingredients were provided in the amounts indicated an included in a capsule.
Figure imgf000040_0001
[00156] The procedure of Method A was followed.
Example 4
Preparation of enteric coated capsules
Step I: Preparation of liquid-filled capsule
[00157] Hard gelatin capsules (50 counts, 00 size) were filled with a liquid composition of Example 3. These capsules were manually filled with 800 mg of the formulation and then sealed by hand with a 50% ethanol/ 50% water solution. The capsules were then banded by hand with 22% gelatin solution containing the following ingredients in the amounts indicated.
Figure imgf000040_0002
[00158] The gelatin solution mixed thoroughly and allowed to swell for 1-2 hours. After the swelling period, the solution was covered tightly and placed in a 55°C oven and allowed to liquefy. Once the entire gelatin solution was liquid, the banding was performed [00159] Using a pointed round 3/0 artist brush, the gelatin solution was painted onto the capsules. Banding kit provided by Shionogi was used. After the banding, the capsules were kept at ambient conditions for 12 hours to allow the band to cure.
Step II: Coating of liquid-filled capsule
[00160] A coating dispersion was prepared from the ingredients listed in the table below.
Figure imgf000041_0001
[00161] If banded capsules according to Step I were used, the dispersion was applied to the capsules to a 20.0 mg/cm2 coating level. The following conditions were used to coat the capsules.
Figure imgf000041_0002
* Spray nozzle was set such that both the nozzle and spray path were under the flow path of inlet air. Example 5
Treatment of Zika virus infection in a subject
Method A. Antiviral Composition therapy
[00162] A subj ect presenting with Zika virus 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 Zika 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
[00163] Method A, above, is followed except that the subject is prescribed and administered one or more other therapeutic agents for the treatment of Zika virus 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.
Example 6
In vitro Evaluation of Antiviral Activity Against Zika Virus Infection
Method A. Pure compound
[00164] 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; ¾ί 5:^^.8ΐ∞.θΓ¾ ·ΡΓθάϋθί5/Α11/ν.Κ-1843.β5 χ) 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. Cells were given fresh medium and incubated for 48hr, after which they were fixed with formalin and stained for ZIKV infection. Representative infection rates for oleandrin (FIG. 1A) and digoxin (FIG. IB) as determined by scintigraphy are depicted. Other compounds are evaluated under the same conditions and exhibit very varying levels of antiviral activity against Zika virus.
Method B. Compound in Extract Form [00165] 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. For example, 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.
Example 7
Preparation of a tablet comprising antiviral composition
[00166] An initial tabletting mixture of 3% Syloid 244FP and 97% microcrystalline cellulose (MCC) was mixed. Then, an existing batch of composition prepared according to Example 3 was incorporated into the Syloid/MCC mixture via wet granulation. This mixture is labeled "Initial Tabletting Mixture) in the table below. Additional MCC was added extra- granularly to increase compressibility. This addition to the Initial Tabletting Mixture was labeled as "Extra-granular Addition. " The resultant mixture from the extra-granular addition was the same composition as the "Final Tabletting Mixture."
Component Weight (g) Weight % (w/w)
Initial Tabletting Mixture
Microcrystalline cellulose 48.5 74.2
Colloidal Silicon Dioxide/Syloid
244FP 1.5 2.3
Formulation from Ex. 3 15.351 23.5
Total 65.351 100.0
Extr agranular addition
Component Weight (g) Weight % (w/w)
Initial Tabulating Mixture 2.5 50.0
Microcrystalline cellulose 2.5 50.0
Total 5 100.0 Final Tabletting Mixture:
Abbreviated
Figure imgf000044_0001
[00167] 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.
Example 8
HPLC analysis of solutions containing oleandrin
[00168] Samples (oleandrin standard, SCF extract and hot-water extract) were analyzed on HPLC (Waters) using the following conditions: Symmetry C18 column (5.0 μιτι, 150 x4.6 mm I.D.; Waters); Mobile phase of MeOH:water = 54: 46 (v/v) and flow rate at 1.0 ml/min. Detection wavelength was set at 217 nm. 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.
Example 9
Preparation of Pharmaceutical Composition
[00169] 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.
Method A.
[00170] At least one pharmaceutical excipient is mixed with at least one antiviral compound disclosed herein.
Method B.
[00171] At least one pharmaceutical excipient is mixed with at least two antiviral compounds disclosed herein.
Method C.
[00172] At least one pharmaceutical excipient is mixed with at least one cardiac glycosides disclosed herein.
Method D.
[00173] At least one pharmaceutical excipient is mixed with at least two triterpenes disclosed herein.
Method E.
[00174] At least one pharmaceutical excipient is mixed with at least one cardiac glycoside disclosed herein and at least two triterpenes disclosed herein.
Method D.
[00175] At least one pharmaceutical excipient is mixed with at least three triterpenes disclosed herein.
Example 10
Preparation of Triterpene Mixtures
[0003] The following compositions were made by mixing the specified triterpenes in the approximate molar ratios indicated. Triterpene (Approximate Relative Molar Content)
Composition Oleanolic acid (0) Ursolic acid (U) Betulinic acid (B)
I (A-C) 3 2.2 1
II (A-C) 7.8 7.4 1
III (A-C) 10 1 1
IV (A-C) 1 10 1
V (A-C) 1 1 10
VI (A-C) 1 1 0
VII (A-C) 1 1 1
VIII (A-C) 10 1 0
IX (A-C) 1 10 0
[0004] For each composition, three different respective solutions were made, whereby the total concentration of triterpenes in each solution was approximately 9 μΜ, 18 μΜ, or 36 μΜ.
Composition Triterpene (Approximate Content of Each, μΜ)
(total triterpene Oleanolic acid (O) Ursolic acid (U) Betulinic acid (B) content, μΜ)
I- A (36) 17.4 12.8 5.8
I-B (18) 8.7 6.4 2.9
I-C (9) 4.4 3.2 1.5
II-A (36) 17.3 16.4 2.2
II-B (18) 8.7 8.2 1.1
II-C (9) 4.3 4.1 0.6
III-A (36) 30 3 3
III-B (18) 15 1.5 1.5
III-C (9) 7.5 0.75 0.75
IV- A (36) 3 30 3
IV-B (18) 1.5 15 1.5
IV-C (9) 0.75 7.5 0.75
V-A (36) 3 3 30
V-B (18) 1.5 1.5 15 Composition Triterpene (Approximate Content of Each, μΜ)
(total triterpene Oleanolic acid (0) Ursolic acid (U) Betulinic acid (B) content, μΜ)
V-C (9) 0.75 0.75 7.5
VI- A (36) 18 18 0
VI-B (18) 9 9 0
VI-C (9) 4.5 4.5 0
VII-A (36) 12 12 12
VII-B (18) 6 6 6
VII-C (9) 3 3 3
VIII-A (36) 32.7 3.3 0
VIII-B (18) 16.35 1.65 0
VIII-C (9) 8.2 0.8 0
IX- A (36) 3.3 32.7 0
IX-B (18) 1.65 16.35 0
IX-C (9) 0.8 8.2 0
Example 11
Preparation of Antiviral Compositions
[0005] 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
[0006] Known amounts of oleanolic acid and ursolic 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. The resultant mixture contained the components in the relative molar ratios as described herein.
[0007] For 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
[0008] Known amounts of 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. The resultant mixture contained the components in the relative molar ratios as described herein.
[0009] For 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, ursolic acid, and betulinic acid
[0010] Known amounts of 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. The resultant mixture contained the components in the relative molar ratios as described herein.
[0011] For 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 oleadrin, oleanolic acid, ursolic acid, and betulinic acid
[0012] Known amounts of 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. The resultant mixture contained the components in the relative molar ratios as described herein.
[0013] For 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. Example 12
Treatment of Filovirus infection in a subject
[00176] Exemplary Filovirus infections include Ebolavirus and Marburgvirus.
Method A. Antiviral Composition therapy
[00177] 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
[00178] Method A, above, 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.
Example 13
Treatment of Flavivirus infection in a subject
[00179] 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.
Method A. Antiviral Composition therapy
[00180] 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
[00181] Method A, above, 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.
Example 14
Evaluation of antiviral activity againt Zikavirus and Dengue virus
[00182] 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. For CPE-based assays, 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. Once inside the lysosome, 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
[00183] 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.
[00184] The antiviral activity of test compositions was 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.
[00185] Test compositions were dissolved in 100% methanol. Eight concentrations of the compositions were generated (in duplicate) by performing 8-fold dilutions, starting with 50 μΜ as the highest concentration tested. The highest test concentration of composition (50 μΜ) 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.
[00186] Antiviral activity was evaluated by the degree of protection against virus- induced cytopathic effects (CPE). Cells were challenged with virus in the presence of different concentrations of control or compositions. The extent of protection against CPE was monitored after 6 days (ZIKV, Zikavirus) or 7 days (DENV, Dengue virus) post infection by quantifying cell viability in different test conditions and comparing values with that of untreated cells and cells treated with vehicle alone (infection medium).
[00187] 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.
[00188] Protection of DENV2-induced cytopathic effect (CPE) with 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). Test material dilutions were prepared at 1.25X the final concentration and 40μ1 were incubated with the target cells at 37°C for 30 minutes. Following the test material preincubation, ΙΟμΙ of virus dilutions prepared in MEM with 1% BSA was added to each well (50μ1 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. Subsequently, 50μ1 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 7 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. Control wells containing 50μΜ Ribavirin and 0.5μΜ compound A3 were also included on the assay plate. After 7 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 Ribavirin (0.5μΜ) or A3 (0.5μΜ). A full duplicate inhibition curve with methanol vehicle only was included on the same assay plate.
[00189] Protection of ZIKV-induced cytopathic effect (CPE) with Neutral Red readout: For the ZIKV antiviral assay, the PLCal ZV strain was used. 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). Test material dilutions were prepared at 1.25X the final concentration and 40μ1 were incubated with the target cells at 37°C for 30 minutes. Following the test material pre-incubation, ΙΟμΙ of virus dilutions prepared in MEM with 1% BSA was added to each well (50μ1 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, 50μ1 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 (0.5μΜ). A full duplicate inhibition curve with methanol vehicle only was included on the same assay plate.
[00190] Analysis of 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.
[00191] Viability assay (XTT) 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. Background levels obtained from wells with no cells were subtracted from all data-points. Controls with methanol vehicle alone (at 7 concentrations mirroring the final percent methanol of each Oleandrin test wells) were included in the viability assay plate. The extent of viability was monitored by measuring absorbance at 490 nm.
[00192] Analysis of cytotoxicity data: For the XTT 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. The signal-to-background (S/B) for this assay was 29.9 (for IV A), 8.7 (for HRV), 6.5 (for DENV), and 6.7 (for ZIKV), determined as the viability percentage in "mock-infected" cells compared to the signal observed for wells without cells.
Example 15
Evaluation of antiviral activity againt Filovirus (Ebolavirus and Marburgvirus)
Method A.
[00193] Vero E6 cells were infected with EBOV/Kik (A, MOI=l) or MARV/Ci67 (B, MOI=l) in the presence of oleandrin, digoxin or PBI-05204, an oleandrin-containing plant extract. After lhr, inoculum and compounds were removed and fresh medium added to cells. 48hr later, cells were fixed and immunostained to detect cells infected with EBOV or MARV. Infected cells were enumerated using an Operetta. C) Vero E6 were treated with compound as above. ATP levels were measured by CellTiter-Glo as a measurement of cell viability.
Method B.
[00194] 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.
Method C.
[00195] 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.
Example 16
Evaluation of antiviral activity againt Togaviridae virus
(Alphavirus: VEEV and WEEV)
[00196] Vero E6 cells were infected with Venezuelan equine encephalitis virus (A, MOI=0.01) or Western equine encephalitis virus (B, MOI=0.1) for 18hr in the presence or absence of indicated compounds. Infected cells were detected as described herein and enumerated on an Operetta. Example 17
Treatment of Paramyxoviridae infection in a subject
[00197] Exemplary Paramyxoviridae family viral infections include Henipavirus genus infection, Nipah virus infection, or Hendra virus infection.
Method A. Antiviral Composition therapy
[00198] 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
[00199] Method A, above, is followed except that the subject is prescribed and administered one or more other therapeutic agents for the treatment of Paramyxoviridae family 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.
[00200] As used herein, 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
[00201] The above is a detailed description of particular embodiments of the invention. It will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without departing from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims. All of the embodiments disclosed and claimed herein can be made and executed without undue experimentation in light of the present disclosure.

Claims

1) A method of treating viral infection, caused by a virus of the Flaviviridae family, Filoviridae family, Paramyxoviridae family, or Togaviridae family, in a subject in need thereof, the method comprising administering to the subject one or more doses of an antiviral composition comprising: a) cardiac glycoside selected from oleandrin or digoxin; b) plural triterpenes; or c) a combination of cardiac glycoside(s) and plural triterpenes.
2) A prophylactic method of treating a subject at risk of contracting a viral infection caused by a virus of the Flaviviridae family, Filoviridae family, Paramyxoviridae family, or Togaviridae family, the method comprising chronically administering to the subject one or more doses of an antiviral composition on a recurring basis over an extended treatment period prior to the subject contracting the viral infection, thereby preventing the subject from contracting the viral infection; wherein the antiviral composition comprises: a) cardiac glycoside selected from oleandrin or digoxin; b) plural triterpenes; or c) a combination of cardiac glycoside(s) and plural triterpenes.
3) A method of treating viral infection, caused by a virus of the Flaviviridae family, Filoviridae family, Paramyxoviridae family, or Togaviridae family, in a subject in need thereof, the method comprising:
determining whether or not the subject has a viral infection viral infection caused by a virus of the Flaviviridae family, Filoviridae family, Paramyxoviridae family, or Togaviridae family;
indicating administration of an antiviral composition comprising: a) cardiac glycoside; b) plural triterpenes; or c) a combination of cardiac glycoside(s) and plural triterpenes; administering an initial dose of antiviral composition to the subject according to a prescribed initial dosing regimen for a period of time;
periodically determining the adequacy of subject's clinical response and/or therapeutic response to treatment with the antiviral composition; and
if the subject's clinical response and/or therapeutic response is adequate, then continuing treatment with the 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. 4) The method of claim 1, wherein the cardiac glycoside is oleandrin.
5) The method of claim 1 , wherein the plural triterpenes are at least two triterpenes selected from the group consisting of oleanolic acid (free acid, salt, prodrug or derivative thereof), ursolic acid (free acid, salt, prodrug or derivative thereof), betulinic acid (free acid, salt, prodrug or derivative thereof).
6) The method of any one of claims 1 -5, wherein the antiviral composition comprises oleandrin, oleanolic acid (free acid, salt, prodrug or derivative thereof) and ursolic acid (free acid, salt, prodrug or derivative thereof).
7) The method of any one of claims 1 -5, wherein the antiviral composition comprises oleandrin, oleanolic acid (free acid, salt, prodrug or derivative thereof) and betulinic acid (free acid, salt, prodrug or derivative thereof).
8) The method of any one claims 1-5, wherein the antiviral composition comprises oleandrin, oleanolic acid (free acid, salt prodrug or derivative thereof), ursolic acid (free acid, salt, prodrug or derivative thereof), and betulinic acid (free acid, salt, prodrug or derivative thereof).
9) The method of any one of claims 1 -3, wherein the antiviral composition comprises oleanolic acid (free acid, salt, prodrug or derivative thereof), ursolic acid (free acid, salt, prodrug or derivative thereof), and betulinic acid (free acid, salt, prodrug or derivative thereof).
10) The method of any one of claims 1 -3, wherein the antiviral composition comprises oleanolic acid (free acid, salt, prodrug or derivative thereof) and ursolic acid (free acid, salt, prodrug or derivative thereof).
1 1) The method of any one of claims 1 -3, wherein the antiviral composition comprises oleanolic acid (free acid, salt, prodrug or derivative thereof) and betulinic acid (free acid, salt, prodrug or derivative thereof).
12) The method of any one of the above claims, wherein the viral infection is selected from the group consisting of Filovirus infection, Ebolavirus infection and Marburgvirus infection.
13) The method of any one of claims 1-1 1, wherein the viral infection is selected from the group consisting of Flavivirus infection, Yellow Fever, Dengue Fever, Japanese Enchephalitis, West Nile Viruses, Zikavirus, Tick-borne Encephalitis, Kyasanur Forest Disease, Alkhurma Disease, Omsk Hemorrhagic Fever, and Powassan virus infection. 14) The method of any one of claims 1-1 1, wherein the viral infection is selected from the group consisting of Alpahavirus infection, Venezuelan Equine Encephalits virus infection, Western Equine Encephalitis virus infection, Chikungunya virus infection, and Eastern Equine Encephalitis virus.
15) The method of any one of claims 1-1 1, wherein the viral infection is selected from the group consisting of Henipavirus infection, Hendra virus infection, and Nipah virus infection.
16) The method according to any one of the above claims, wherein the viral titre in the subject's blood or plasma is reduced or does not increase as a result of the treatment.
17) The method according to any one of the above claims, wherein one or more doses are administered on a daily, weekly or monthly basis.
18) The method according to any one of the above claims, wherein the administration is parenteral, buccal, enteral, intramuscular, subdermal, sublingual, peroral, oral administration or a combination thereof.
19) The method according to any one of the above claims, wherein the one or more cardiac glycoside is present in pure form or as part of an extract containing the cardiac glycoside.
20) The method according to any one of the above claims, wherein the antiviral composition further comprises at least one pharmaceutical excipient selected from the group consisting of water soluble (or water miscible) co-solvent, water insoluble (water immiscible) co-solvent, antioxidant, and surfactant.
21) The method of claim 20, wherein the at least one water miscible solvent, at least one antioxidant, and at least one solubilizing agent are present.
22) Use of one or more antiviral compositions as described herein in the manufacture of a medicament for the treatment of Filovirus infection or Flavivirus infection in a subject.
23) Use of one or more antiviral compositions as described herein in the treatment of viral infection.
PCT/US2018/042226 2016-09-14 2018-07-16 Method and composition for treating viral infection WO2019055119A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
KR1020217026676A KR20210107904A (en) 2017-09-14 2018-07-16 Composition for treating viral infection
EP18857189.7A EP3681508A4 (en) 2017-09-14 2018-07-16 Method and composition for treating viral infection
CA3075729A CA3075729A1 (en) 2017-09-14 2018-07-16 Use of oleandrin to treatn viral infection
IL285232A IL285232B (en) 2017-09-14 2018-07-16 Compositions comprising oleandrin for treating viral infection
CN202111022020.5A CN114224900A (en) 2016-09-14 2018-07-16 Methods and compositions for treating viral infections
CN201880059793.0A CN111093672B (en) 2016-09-14 2018-07-16 Methods and compositions for treating viral infections
AU2018334386A AU2018334386B2 (en) 2017-09-14 2018-07-16 Method and composition for treating viral infection
MX2020002883A MX2020002883A (en) 2017-09-14 2018-07-16 Method and composition for treating viral infection.
RU2020113341A RU2020113341A (en) 2017-09-14 2018-07-16 METHOD AND COMPOSITION FOR VIRAL INFECTION TREATMENT
JP2020515911A JP6820450B2 (en) 2017-09-14 2018-07-16 Methods and compositions for treating viral infections
KR1020207007406A KR102295179B1 (en) 2017-09-14 2018-07-16 Methods and compositions for the treatment of viral infections
US16/276,063 US10596186B2 (en) 2016-09-14 2019-02-14 Method and compositions for treating viral infections
US16/424,902 US10702567B2 (en) 2016-09-14 2019-05-29 Method and compositions for treating viral infection
US16/747,637 US11007239B2 (en) 2016-09-14 2020-01-21 Method and compositions for treating viral infection
US16/779,840 US10874704B2 (en) 2016-09-14 2020-02-03 Method and compositions for treating viral infection
US16/803,488 US11123387B2 (en) 2016-09-14 2020-02-27 Method and compositions for treating viral infection
IL273162A IL273162B (en) 2017-09-14 2020-03-09 Compositions comprising oleandrin for treating viral infection
US16/815,260 US10980852B2 (en) 2016-09-14 2020-03-11 Method and compositions for treating viral infection
US16/825,063 US11013776B2 (en) 2016-09-14 2020-03-20 Method and compositions for treating viral infection
US16/895,920 US10729735B1 (en) 2016-09-14 2020-06-08 Method and compostitions for treating coronavirus infection
AU2020204236A AU2020204236B2 (en) 2017-09-14 2020-06-25 Method and composition for treating viral infection
US17/405,572 US11324789B2 (en) 2016-09-14 2021-08-18 Method and compositions for treating equine encephalitis

Applications Claiming Priority (4)

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
USPCT/US2017/051553 2017-09-14
US201862698365P 2018-07-16 2018-07-16
US62/698,365 2018-07-16

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2017/051553 Continuation-In-Part WO2018053123A1 (en) 2016-09-14 2017-09-14 Method and compositions for treating viral infection
US16/276,063 Continuation-In-Part US10596186B2 (en) 2016-09-14 2019-02-14 Method and compositions for treating viral infections

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/276,063 Continuation US10596186B2 (en) 2016-09-14 2019-02-14 Method and compositions for treating viral infections

Publications (1)

Publication Number Publication Date
WO2019055119A1 true WO2019055119A1 (en) 2019-03-21

Family

ID=65723839

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2018/042226 WO2019055119A1 (en) 2016-09-14 2018-07-16 Method and composition for treating viral infection

Country Status (9)

Country Link
EP (1) EP3681508A4 (en)
JP (3) JP6820450B2 (en)
KR (2) KR102295179B1 (en)
AU (2) AU2018334386B2 (en)
CA (1) CA3075729A1 (en)
IL (2) IL285232B (en)
MX (1) MX2020002883A (en)
RU (1) RU2020113341A (en)
WO (1) WO2019055119A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112689510A (en) * 2020-03-31 2021-04-20 菲尼克斯生物技术公司 Methods and compositions for treating coronavirus infection
WO2021201903A1 (en) 2020-03-31 2021-10-07 Phoenix Biotechnology, Inc. Method and compositions for treating coronavirus infection
FR3109297A1 (en) * 2020-04-19 2021-10-22 Ephyla "New non-protein furin inhibitor"
WO2022094816A1 (en) * 2020-11-04 2022-05-12 Janssen Pharmaceuticals, Inc. Solid formulation
US11806359B2 (en) 2020-03-31 2023-11-07 Phoenix Biotechnology, Inc. Method and compositions for treating Coronavirus infection
EP4295854A2 (en) 2020-03-31 2023-12-27 Phoenix Biotechnology, Inc. Method and compositions for treating coronavirus infection

Citations (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5135745A (en) 1986-05-13 1992-08-04 Ozel Huseyin Z Extracts of nerium species, methods of preparation, and use therefore
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
US5869060A (en) 1996-12-03 1999-02-09 Eastwood Biomedical Research, Inc. Portulaca oleracea and tumor cell growth
WO2000016793A1 (en) 1998-09-24 2000-03-30 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
US6715705B2 (en) 2001-03-16 2004-04-06 Frank F. Rowley, Jr. Two-stage comminuting and dehydrating system and method
US20040082521A1 (en) 2002-03-29 2004-04-29 Azaya Therapeutics Inc. Novel formulations of digitalis glycosides for treating cell-proliferative and other diseases
US6824811B2 (en) 2001-04-09 2004-11-30 Loders Croklaan Usa Llc Concentrate of triterpenes
US20040247660A1 (en) 2002-11-06 2004-12-09 Singh Chandra U. Protein-stabilized liposomal formulations of pharmaceutical agents
US20050026849A1 (en) 2003-03-28 2005-02-03 Singh Chandra U. Water soluble formulations of digitalis glycosides for treating cell-proliferative and other diseases
US20060025274A1 (en) 2004-07-28 2006-02-02 Usoro Patrick B Planetary transmissions having three interconnected gear members and clutched input members
US20060073222A1 (en) 1998-05-19 2006-04-06 Arntzen Charles J Triterpene compositions and methods for use thereof
US20060234955A1 (en) * 2002-05-28 2006-10-19 Bette Pollard Cardiac glycosides to treat cystic fibrosis and other il-8 dependent disorders
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
WO2007016176A2 (en) 2005-07-28 2007-02-08 Phoenix Biotechnology Inc. Scf extract containing cardiac glycoside
US20070072835A1 (en) 2000-08-18 2007-03-29 The Board Of Trustees Of The University Of Illinois Method of preparing and use of prodrugs of betulinic acid derivatives
US20080293682A1 (en) 2005-02-09 2008-11-27 Rama Mukherjee Novel Betulinic Acid Derivatives
US20090068257A1 (en) 2006-03-07 2009-03-12 Jean-Claude Leunis Betulonic and betulinic acid derivatives
US20090076290A1 (en) 2005-03-29 2009-03-19 Regents Of The University Of Minnesota Selective oxidation of triterpenes employing tempo
US20090131714A1 (en) 2005-06-08 2009-05-21 Krasutsky Pavel A Synthesis of betulonic and betulinic aldehydes
US20090203661A1 (en) 2006-10-12 2009-08-13 Safe Stephen H Betulinic acid, derivatives and analogs thereof and uses therefor
US20100048911A1 (en) 2008-04-18 2010-02-25 Xin Jiang Antioxidant inflammation modulators: novel derivatives of oleanolic acid
US7718635B2 (en) 2005-06-10 2010-05-18 Pola Chemical Industries Inc. Triterpenic acid derivative and preparation for external application for skin comprising the same
US20110091398A1 (en) 2007-08-07 2011-04-21 Muhammed Majeed Oleanoyl peptide composition and a method of treating skin aging
US20110218204A1 (en) 2009-07-14 2011-09-08 Hetero Research Foundation, Hetero Drugs Limited Pharmaceutically acceptable salts of novel betulinic acid derivatives
US20110224182A1 (en) 2007-01-19 2011-09-15 Myriad Pharmaceuticals, Incorporated Salts of (3-0-(3',3'-dimethylsuccinyl) betulinic acid and solid state forms thereof
US20110224159A1 (en) 2008-09-10 2011-09-15 Universite du Quebec a Chicoutime Bidesmosidic betulin and betulinic acid derivatives and uses thereof as antitumor agents
US20110313191A1 (en) 2005-12-16 2011-12-22 Myrexis, Inc. Preparation of Pharmaceutical Salts of 3-0-(3',3'-Dimethylsuccinyl) Betulinic Acid
US20120101149A1 (en) 2008-11-04 2012-04-26 Tadashi Honda Betulinic Acid Derivatives and Methods of Use Thereof
US20120165279A1 (en) 2010-12-22 2012-06-28 Industrial Technology Research Institute Pharmaceutical composition and method for preventing or treating hepatitis c
US20120214814A1 (en) 2008-04-18 2012-08-23 Eric Anderson Antioxidant inflammation modulators: oleanolic acid derivatives with amino acid and other modifications at c-17
US20120214775A1 (en) 2004-03-17 2012-08-23 Myrexis, Inc. Pharmaceutical Salts of 3-O-(3',3'-Dimethylsuccinyl) Betulinic Acid
US20120237629A1 (en) 2009-11-30 2012-09-20 University of Zwazulu-Natal In vitro anti-sickling activity of betulinic acid, oleanolic acid and their derivatives
US20120245374A1 (en) 2008-04-18 2012-09-27 Reata Pharmaceuticals, Inc. Antioxidant Inflammation Modulators: Oleanolic Acid Derivatives with Saturation in the C-Ring
US20120302530A1 (en) 2009-11-23 2012-11-29 Suzhou Maidixian Pharmaceutical Inc. 23-hydroxy-betulinic acid derivatives, preparation methods and uses thereof
US20130029954A1 (en) 2011-01-31 2013-01-31 Bristol-Myers Squibb Company C-28 amines of c-3 modified betulinic acid derivatives as hiv maturation inhibitors
US8367363B2 (en) 2007-11-13 2013-02-05 Phoenix Biotechnology, Inc. Method of determining the probability of a therapeutic response in cancer chemotherapy with cardiac glycoside
US20130065868A1 (en) 2001-05-11 2013-03-14 University Of Ottawa Anxioytic marcgraviaceae compositions containing betulinic acid, betulinic acid derivatives, and methods
US8481086B2 (en) 2010-01-11 2013-07-09 Phoenix Biotechnology, Inc. Method of treating neurological conditions with cardiac glycoside
US20130303607A1 (en) 2012-05-08 2013-11-14 Trustees Of Dartmouth College Triterpenoids and Compositions Containing the Same
US20140066408A1 (en) 2008-04-18 2014-03-06 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: c-17 homologated oleanolic acid derivatives
US20140088163A1 (en) 2012-09-10 2014-03-27 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US20140088188A1 (en) 2012-09-10 2014-03-27 Reata Pharmaceuticals, Inc. C13-hydroxy derivatives of oleanolic acid and methods of use thereof
US20140100227A1 (en) 2012-09-10 2014-04-10 Reata Pharmaceuticals, Inc. C17-alkanediyl and alkenediyl derivatives of oleanolic acid and methods of use thereof
US8729055B2 (en) 2010-02-02 2014-05-20 Kaohsiung Medical University Ursolic acid derivative and pharmaceutical composition thereof
US20140221328A1 (en) 2011-01-10 2014-08-07 Bandi Parthasaradhi Reddy Pharmaceutically acceptable salts of novel betulinic acid derivatives
US20140243298A1 (en) 2013-02-25 2014-08-28 Bristol-Myers Squibb Company C-3 alkyl and alkenyl modified betulinic acid derivatives
US20140296546A1 (en) 2011-11-18 2014-10-02 Wenzhou University Method for producing 30-halogenated betulinic acid
US20140343064A1 (en) 2011-12-01 2014-11-20 Hanghzhou Bensheng Pharmaceutical Co., Ltd 2-substituted oleanolic acid derivative, method preparing for same, and application thereof
US20140343108A1 (en) 2011-12-01 2014-11-20 Hangzhou Bensheng Pharmaceutical Co., Ltd. Oleanolic acid amidate derivatives, preparation methods and uses thereof
US9011937B2 (en) 2010-11-22 2015-04-21 Phoenix Biotechnology, Inc. Method of treating neurological conditions with extract of Nerium species or Thevetia species
US20150119373A1 (en) 2012-04-24 2015-04-30 Hetero Research Foundation Novel betulinic acid derivatives as hiv inhibitors
US20150218206A1 (en) 2012-08-14 2015-08-06 Nexoligo Co., Ltd. Novel ursolic acid derivative and method for preparing same
US20150337004A1 (en) 2012-12-31 2015-11-26 Hetero Research Foundation Novel betulinic acid proline derivatives as hiv inhibitors
US20160000754A1 (en) * 2004-01-06 2016-01-07 Paul Edward Stamets Antiviral Activity from Medicinal Mushrooms and their Active Constituents
US20160354396A1 (en) * 2015-06-08 2016-12-08 Cutanea Life Sciences, Inc. Therapeutic composition
US20170096446A1 (en) 2006-11-03 2017-04-06 Myrexis, Inc. Extended Triterpene Derivatives
US20170130233A1 (en) * 2014-02-12 2017-05-11 Organobalance Gmbh Yeast strain and microbial method for production of pentacyclic triterpenes and/or triterpenoids
US20170204133A1 (en) 2014-06-19 2017-07-20 VIIV Healthcare UK (No.5) Limited Betulinic acid derivatives with hiv maturation inhibitory activity

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1803461A1 (en) * 2005-12-27 2007-07-04 Heinz-Herbert Fiebig Therapeutic use of an extract from the leaves of Nerium oleander
US9492438B2 (en) * 2014-07-25 2016-11-15 Bette Pollard Amphiphilic pyridinum compounds to treat epilepsy and other disorders of the nervous system

Patent Citations (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5135745A (en) 1986-05-13 1992-08-04 Ozel Huseyin Z Extracts of nerium species, methods of preparation, and use therefore
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
US5869060A (en) 1996-12-03 1999-02-09 Eastwood Biomedical Research, Inc. Portulaca oleracea and tumor cell growth
US20110294752A1 (en) 1998-05-19 2011-12-01 Research Development Foundation Triterpene compositions and methods for use thereof
US20060073222A1 (en) 1998-05-19 2006-04-06 Arntzen Charles J Triterpene compositions and methods for use thereof
US20100189824A1 (en) 1998-05-19 2010-07-29 Arntzen Charles J Triterpene compositions and methods for use thereof
US20020114852A1 (en) 1998-09-24 2002-08-22 Ulagaraj Selvaraj Extract of nerium species, pharmaceutical composition thereof and methods for preparation thereof
US6565897B2 (en) 1998-09-24 2003-05-20 Ozelle Pharmaceuticals, Inc. Extract of nerium species, pharmaceutical composition thereof and methods for preparation thereof
WO2000016793A1 (en) 1998-09-24 2000-03-30 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
US20070072835A1 (en) 2000-08-18 2007-03-29 The Board Of Trustees Of The University Of Illinois Method of preparing and use of prodrugs of betulinic acid derivatives
US6715705B2 (en) 2001-03-16 2004-04-06 Frank F. Rowley, Jr. Two-stage comminuting and dehydrating system and method
US6824811B2 (en) 2001-04-09 2004-11-30 Loders Croklaan Usa Llc Concentrate of triterpenes
US20130065868A1 (en) 2001-05-11 2013-03-14 University Of Ottawa Anxioytic marcgraviaceae compositions containing betulinic acid, betulinic acid derivatives, and methods
US20040082521A1 (en) 2002-03-29 2004-04-29 Azaya Therapeutics Inc. Novel formulations of digitalis glycosides for treating cell-proliferative and other diseases
US20060234955A1 (en) * 2002-05-28 2006-10-19 Bette Pollard Cardiac glycosides to treat cystic fibrosis and other il-8 dependent disorders
US20040247660A1 (en) 2002-11-06 2004-12-09 Singh Chandra U. Protein-stabilized liposomal formulations of pharmaceutical agents
US20050026849A1 (en) 2003-03-28 2005-02-03 Singh Chandra U. Water soluble formulations of digitalis glycosides for treating cell-proliferative and other diseases
US20160000754A1 (en) * 2004-01-06 2016-01-07 Paul Edward Stamets Antiviral Activity from Medicinal Mushrooms and their Active Constituents
US20120214775A1 (en) 2004-03-17 2012-08-23 Myrexis, Inc. Pharmaceutical Salts of 3-O-(3',3'-Dimethylsuccinyl) Betulinic Acid
US20060025274A1 (en) 2004-07-28 2006-02-02 Usoro Patrick B Planetary transmissions having three interconnected gear members and clutched input members
US20080293682A1 (en) 2005-02-09 2008-11-27 Rama Mukherjee Novel Betulinic Acid Derivatives
US20090076290A1 (en) 2005-03-29 2009-03-19 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
US7718635B2 (en) 2005-06-10 2010-05-18 Pola Chemical Industries Inc. Triterpenic acid derivative and preparation for external application for skin comprising the same
US8187644B2 (en) 2005-07-28 2012-05-29 Phoenix Biotechnology Inc. SCF extract containing cardiac glycoside
US7402325B2 (en) 2005-07-28 2008-07-22 Phoenix Biotechnology, Inc. Supercritical carbon dioxide extract of pharmacologically active components from Nerium oleander
WO2007016176A2 (en) 2005-07-28 2007-02-08 Phoenix Biotechnology Inc. Scf extract containing cardiac glycoside
US8394434B2 (en) 2005-07-28 2013-03-12 Phoenix Biotechnology, Inc. SCF extract containing cardiac glycoside
US20110313191A1 (en) 2005-12-16 2011-12-22 Myrexis, Inc. Preparation of Pharmaceutical Salts of 3-0-(3',3'-Dimethylsuccinyl) Betulinic Acid
US9303058B2 (en) * 2006-03-07 2016-04-05 Jean-Claude Leunis Betulonic and betulinic acid derivatives
US20090068257A1 (en) 2006-03-07 2009-03-12 Jean-Claude Leunis Betulonic and betulinic acid derivatives
US20140066416A1 (en) 2006-03-07 2014-03-06 Jean-Claude Leunis Betulonic and betulinic acid derivatives
US20090203661A1 (en) 2006-10-12 2009-08-13 Safe Stephen H Betulinic acid, derivatives and analogs thereof and uses therefor
US20170096446A1 (en) 2006-11-03 2017-04-06 Myrexis, Inc. Extended Triterpene Derivatives
US20110224182A1 (en) 2007-01-19 2011-09-15 Myriad Pharmaceuticals, Incorporated Salts of (3-0-(3',3'-dimethylsuccinyl) betulinic acid and solid state forms thereof
US20110091398A1 (en) 2007-08-07 2011-04-21 Muhammed Majeed Oleanoyl peptide composition and a method of treating skin aging
US8367363B2 (en) 2007-11-13 2013-02-05 Phoenix Biotechnology, Inc. Method of determining the probability of a therapeutic response in cancer chemotherapy with cardiac glycoside
US20120214814A1 (en) 2008-04-18 2012-08-23 Eric Anderson Antioxidant inflammation modulators: oleanolic acid derivatives with amino acid and other modifications at c-17
US20140179928A1 (en) 2008-04-18 2014-06-26 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with saturation in the c-ring
US20120245374A1 (en) 2008-04-18 2012-09-27 Reata Pharmaceuticals, Inc. Antioxidant Inflammation Modulators: Oleanolic Acid Derivatives with Saturation in the C-Ring
US20140066408A1 (en) 2008-04-18 2014-03-06 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: c-17 homologated oleanolic acid derivatives
US20130317007A1 (en) 2008-04-18 2013-11-28 Reata Pharmaceuticals, Inc. Antioxidant inflammation modulators: oleanolic acid derivatives with amino and other modifications at c-17
US20120238767A1 (en) 2008-04-18 2012-09-20 Xin Jiang Antioxidant inflammation modulators: novel derivatives of oleanolic acid
US20100048911A1 (en) 2008-04-18 2010-02-25 Xin Jiang Antioxidant inflammation modulators: novel derivatives of oleanolic acid
US20110224159A1 (en) 2008-09-10 2011-09-15 Universite du Quebec a Chicoutime Bidesmosidic betulin and betulinic acid derivatives and uses thereof as antitumor agents
US20120101149A1 (en) 2008-11-04 2012-04-26 Tadashi Honda Betulinic Acid Derivatives and Methods of Use Thereof
US20110218204A1 (en) 2009-07-14 2011-09-08 Hetero Research Foundation, Hetero Drugs Limited Pharmaceutically acceptable salts of novel betulinic acid derivatives
US20120302530A1 (en) 2009-11-23 2012-11-29 Suzhou Maidixian Pharmaceutical Inc. 23-hydroxy-betulinic acid derivatives, preparation methods and uses thereof
US20120237629A1 (en) 2009-11-30 2012-09-20 University of Zwazulu-Natal In vitro anti-sickling activity of betulinic acid, oleanolic acid and their derivatives
US9358293B2 (en) 2010-01-11 2016-06-07 Phoenix Biotechnology, Inc. Method of treating neurological conditions with oleandrin
US8481086B2 (en) 2010-01-11 2013-07-09 Phoenix Biotechnology, Inc. Method of treating neurological conditions with cardiac glycoside
US20160243143A1 (en) 2010-01-11 2016-08-25 Phoenix Biotechnology, Inc. Method of Treating Neurological Conditions with Oleandrin
US9220778B2 (en) 2010-01-11 2015-12-29 Phoenix Biotechnology, Inc. Method of treating neurological conditions with oleandrin-containing extract
US20150283191A1 (en) 2010-01-11 2015-10-08 Phoenix Biotechnology, Inc. Method of Treating Neurological Conditions with Extract of Nerium Species or Thevetia Species
US8729055B2 (en) 2010-02-02 2014-05-20 Kaohsiung Medical University Ursolic acid derivative and pharmaceutical composition thereof
US9011937B2 (en) 2010-11-22 2015-04-21 Phoenix Biotechnology, Inc. Method of treating neurological conditions with extract of Nerium species or Thevetia species
US20120165279A1 (en) 2010-12-22 2012-06-28 Industrial Technology Research Institute Pharmaceutical composition and method for preventing or treating hepatitis c
US20140221328A1 (en) 2011-01-10 2014-08-07 Bandi Parthasaradhi Reddy Pharmaceutically acceptable salts of novel betulinic acid derivatives
US20130029954A1 (en) 2011-01-31 2013-01-31 Bristol-Myers Squibb Company C-28 amines of c-3 modified betulinic acid derivatives as hiv maturation inhibitors
US20140296546A1 (en) 2011-11-18 2014-10-02 Wenzhou University Method for producing 30-halogenated betulinic acid
US20140343064A1 (en) 2011-12-01 2014-11-20 Hanghzhou Bensheng Pharmaceutical Co., Ltd 2-substituted oleanolic acid derivative, method preparing for same, and application thereof
US20140343108A1 (en) 2011-12-01 2014-11-20 Hangzhou Bensheng Pharmaceutical Co., Ltd. Oleanolic acid amidate derivatives, preparation methods and uses thereof
US20150119373A1 (en) 2012-04-24 2015-04-30 Hetero Research Foundation Novel betulinic acid derivatives as hiv inhibitors
US20130303607A1 (en) 2012-05-08 2013-11-14 Trustees Of Dartmouth College Triterpenoids and Compositions Containing the Same
US20150011627A1 (en) 2012-05-08 2015-01-08 Trustees Of Dartmouth College Triterpenoids and compositions containing the same
US9120839B2 (en) 2012-08-14 2015-09-01 Nexoligo Co., Ltd. Ursolic acid derivative and method for preparing same
US20150218206A1 (en) 2012-08-14 2015-08-06 Nexoligo Co., Ltd. Novel ursolic acid derivative and method for preparing same
US20140100227A1 (en) 2012-09-10 2014-04-10 Reata Pharmaceuticals, Inc. C17-alkanediyl and alkenediyl derivatives of oleanolic acid and methods of use thereof
US20140088188A1 (en) 2012-09-10 2014-03-27 Reata Pharmaceuticals, Inc. C13-hydroxy derivatives of oleanolic acid and methods of use thereof
US20140088163A1 (en) 2012-09-10 2014-03-27 Reata Pharmaceuticals, Inc. C17-heteroaryl derivatives of oleanolic acid and methods of use thereof
US20150337004A1 (en) 2012-12-31 2015-11-26 Hetero Research Foundation Novel betulinic acid proline derivatives as hiv inhibitors
US20140243298A1 (en) 2013-02-25 2014-08-28 Bristol-Myers Squibb Company C-3 alkyl and alkenyl modified betulinic acid derivatives
US20170130233A1 (en) * 2014-02-12 2017-05-11 Organobalance Gmbh Yeast strain and microbial method for production of pentacyclic triterpenes and/or triterpenoids
US20170204133A1 (en) 2014-06-19 2017-07-20 VIIV Healthcare UK (No.5) Limited Betulinic acid derivatives with hiv maturation inhibitory activity
US20160354396A1 (en) * 2015-06-08 2016-12-08 Cutanea Life Sciences, Inc. Therapeutic composition

Non-Patent Citations (38)

* Cited by examiner, † Cited by third party
Title
"Q3C Impurities: Residual Solvents", INTERNATIONAL CONFERENCE ON HARMONISATION OF TECHNICAL REQUIREMENTS FOR REGISTRATION OF PHARMACEUTICALS FOR HUMAN USE (ICH) GUIDANCE FOR INDUSTRY, 1997
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
ADOME ET AL., AFR. HEALTH SCI., vol. 3, no. 2, August 2003 (2003-08-01), pages 77 - 86
AYATOLLAHI ET AL., IRAN. J. PHARM. RES., vol. 10, no. 2, 2011, pages 287 - 294
BARROWS ET AL.: "A screen of FDA-approved drugs for inhibitors of Zikavirus infection", CELL HOST MICROBE, vol. 20, 2016, pages 259 - 270, XP029682741, DOI: 10.1016/j.chom.2016.07.004
BEGUM ET AL., PHYTOCHEMISTRY, vol. 50, no. 3, February 1999 (1999-02-01), pages 435 - 438
CAO ET AL.: "Ethylene glycol-linked amino acid diester prodrugs of oleanolic acid for PEPT1-mediated transport: synthesis, intestinal permeability and pharmacokinetics", MOL. PHARM., vol. 9, no. 8, August 2012 (2012-08-01), pages 2127 - 2135
CHEN ET AL.: "Oleanolic acid nanosuspensions: preparation, in-vitro characterization and enhanced hepatoprotective effect", J. PHARM. PHARMACOL., vol. 57, no. 2, February 2005 (2005-02-01), pages 259 - 264, XP055503197, DOI: 10.1211/0022357055407
CHEUNG: "Antiviral activity of lanatoside C against dengue virus infection", ANTIVIRAL RES., vol. 111, 2014, pages 93 - 99, XP029090000, DOI: 10.1016/j.antiviral.2014.09.007
EL-SHAZLY ET AL., J. EGYPT SOC. PARASITOL., vol. 26, no. 2, August 1996 (1996-08-01), pages 461 - 473
ERDEMOGLU ET AL., J. ETHNOPHARMACOL., vol. 89, no. 1, November 2003 (2003-11-01), pages 123 - 129
FUMIKO ET AL., BIOL. PHARM. BULL, vol. 25, no. 11, 2002, pages 1485 - 1487
GODUGU: "Approaches to improve the oral bioavailability and effects of novel anticancer drugs berberine and betulinic acid", PLOS ONE, vol. 9, no. 3, March 2014 (2014-03-01), pages e89919
JAGER ET AL., MOLECULES, vol. 14, 2009, pages 2016 - 2031
L. E SILVA, MOLECULES, vol. 17, 2012, pages 12197
LEE ET AL., BIOL. PHARM. BULL, vol. 33, no. 2, 2010, pages 330
LI ET AL.: "Development and evaluation of optimized sucrose ester stabilized oleanolic acid nanosuspensions prepared by wet ball milling with design of experiments", BIOL. PHARM. BULL., vol. 37, no. 6, 2014, pages 926 - 937, XP055503198, DOI: 10.1248/bpb.b13-00864
LI ET AL.: "Formulation, biological and pharmacokinetic studies of sucrose ester-stabilized nanosuspensions of oleanolic acid", PHARM. RES., vol. 28, no. 8, August 2011 (2011-08-01), pages 2020 - 2033, XP019921760, DOI: 10.1007/s11095-011-0428-3
MISHRA ET AL., PLOS ONE, vol. 11, no. 7, 25 July 2016 (2016-07-25), pages e0159430
MULLER ET AL., PHARMAZIE, vol. 46, no. 9, September 1991 (1991-09-01), pages 657 - 663
NEWMAN ET AL., J. HERBAL PHARMACOTHERAPY, vol. 1, 2001, pages 1 - 16
OGUZHAN AVCI ET AL.: "Determination of in vitro antiviral activity of Nerium Oleander distillate against to parainfluenza-3 virus", ANIMAL AND VETERINARY SCIENCES, vol. 2, no. 5, January 2014 (2014-01-01), pages 150 - 153, XP055762391, DOI: 10.11648/j.avs.20140205.14
OGUZHAN AVCI ET AL.: "Nerium oleander Distilati'nin Blue Tongue Virus Uzerine in vitro Etkisi - In vitro Effect of Nerium Oleander Distillate on Blue Tongue Virus", AVKAE DERG, vol. 4, 1 January 2014 (2014-01-01), pages 14 - 18, XP055792186
PI ET AL.: "Ursolic acid nanocrystals for dissolution rate and bioavailability enhancement: influence of different particle size", CURR. DRUG DELIV., vol. 13, no. 8, March 2016 (2016-03-01), pages 1358 - 1366
RAJBHANDARI M ET AL.: "Screening of Nepalese medicinal plants for antiviral activity", JOURNAL OF ETHNOPHARMACOLOGY, vol. 74, January 2001 (2001-01-01), pages 251 - 255, XP055792595
RUI ET AL., INT. J. MOL. SCI., vol. 13, 2012, pages 7648 - 7662
SINGH SHAILBALA ET AL.: "FITOTERAPIA", vol. 84, 2 November 2012, IDB HOLDING, article "Nerium oleander derived cardiac glycoside oleandrin is a novel inhibitor of HIV infectivity", pages: 32 - 39
TONG ET AL.: "Spray freeze drying with polyvinylpyrrolidone and sodium caprate for improved dissolution and oral bioavailablity of oleanolic acid, a BCS Class IV compound", INT. J. PHARM., vol. 404, no. 1-2, February 2011 (2011-02-01), pages 148 - 158, XP027575240
VAN KANEGAN ET AL., NATURE SCIENTIFIC REPORTS, vol. 6, May 2016 (2016-05-01), pages 25626
VLASENKO ET AL., FARMATSIIA, vol. 21, no. 5, September 1972 (1972-09-01), pages 46 - 47
WANG, MOLECULES, vol. 21, 2016, pages 139
WU ET AL., MOLECULES, vol. 16, 2011, pages 1 - 15
XI ET AL.: "Formulation development and bioavailability evaluation of a self-nanoemulsified drug delivery system of oleanolic acid", AAPS PHARMSCITECH, vol. 10, no. 1, 2009, pages 172 - 182, XP055127812, DOI: 10.1208/s12249-009-9190-9
YANG ET AL.: "Self-microemulsifying drug delivery system for improved oral bioavailability of oleanolic acid: design and evaluation", INT. J. NANOMED, vol. 8, no. 1, 2013, pages 2917 - 2926
YANG: "Physicochemical properties and oral bioavailability of ursolic acid nanoparticles using supercritical anti-solvent (SAS) process", FOOD CHEM., vol. 132, no. 1, May 2012 (2012-05-01), pages 319 - 325, XP028344727, DOI: 10.1016/j.foodchem.2011.10.083
ZHANG ET AL.: "Enhancement of oral bioavailability of triterpene through lipid nanospheres: preparation, characterization, and absorption evaluation", J. PHARM. SCI., vol. 103, no. 6, June 2014 (2014-06-01), pages 1711 - 1719
ZHAO: "Preparation and characterization of betulin nanoparticles for oral hypoglycemic drug by antisolvent precipitation", DRUG DELIV., vol. 21, no. 6, September 2014 (2014-09-01), pages 467 - 479
ZIA ET AL., J. ETHNOLPHARMACOL., vol. 49, no. 1, November 1995 (1995-11-01), pages 33 - 39

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112689510A (en) * 2020-03-31 2021-04-20 菲尼克斯生物技术公司 Methods and compositions for treating coronavirus infection
WO2021201903A1 (en) 2020-03-31 2021-10-07 Phoenix Biotechnology, Inc. Method and compositions for treating coronavirus infection
TWI753747B (en) * 2020-03-31 2022-01-21 美商菲尼克斯生物技術公司 Method and compositions for treating coronavirus infection
CN112689510B (en) * 2020-03-31 2022-02-01 菲尼克斯生物技术公司 Methods and compositions for treating coronavirus infection
TWI790048B (en) * 2020-03-31 2023-01-11 美商菲尼克斯生物技術公司 Method and compositions for preventing coronavirus infection
US11806359B2 (en) 2020-03-31 2023-11-07 Phoenix Biotechnology, Inc. Method and compositions for treating Coronavirus infection
EP4295854A2 (en) 2020-03-31 2023-12-27 Phoenix Biotechnology, Inc. Method and compositions for treating coronavirus infection
FR3109297A1 (en) * 2020-04-19 2021-10-22 Ephyla "New non-protein furin inhibitor"
WO2021213931A1 (en) * 2020-04-19 2021-10-28 Ephyla Novel non-proteinic furin inhibitor
WO2022094816A1 (en) * 2020-11-04 2022-05-12 Janssen Pharmaceuticals, Inc. Solid formulation

Also Published As

Publication number Publication date
EP3681508A1 (en) 2020-07-22
AU2018334386B2 (en) 2021-11-04
JP2020534293A (en) 2020-11-26
JP6820450B2 (en) 2021-01-27
IL285232A (en) 2021-09-30
KR102295179B1 (en) 2021-08-30
AU2018334386A1 (en) 2020-04-02
AU2020204236A1 (en) 2020-07-16
MX2020002883A (en) 2020-10-05
IL285232B (en) 2022-07-01
AU2020204236B2 (en) 2020-12-10
IL273162A (en) 2020-04-30
RU2020113341A (en) 2021-10-15
KR20200081356A (en) 2020-07-07
IL273162B (en) 2021-08-31
JP2022075755A (en) 2022-05-18
CA3075729A1 (en) 2019-03-21
RU2020113341A3 (en) 2021-10-15
JP7037673B2 (en) 2022-03-16
JP2021063105A (en) 2021-04-22
KR20210107904A (en) 2021-09-01
EP3681508A4 (en) 2021-05-19

Similar Documents

Publication Publication Date Title
US10596186B2 (en) Method and compositions for treating viral infections
EP3512524B1 (en) Method and compositions for treating viral infection
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 compositions for treating coronavirus infection
US20220047616A1 (en) Method and Compositions for Treating HTLV-1 Virus Infection
CA3091376C (en) Method and compositions for treating coronavirus infection
EP4295854A2 (en) Method and compositions for treating coronavirus infection

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 18857189

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3075729

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2020515911

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018334386

Country of ref document: AU

Date of ref document: 20180716

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2018857189

Country of ref document: EP

Effective date: 20200414