WO2021183463A1 - Compositions and methods for treating coronavirus - Google Patents

Compositions and methods for treating coronavirus Download PDF

Info

Publication number
WO2021183463A1
WO2021183463A1 PCT/US2021/021427 US2021021427W WO2021183463A1 WO 2021183463 A1 WO2021183463 A1 WO 2021183463A1 US 2021021427 W US2021021427 W US 2021021427W WO 2021183463 A1 WO2021183463 A1 WO 2021183463A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
cov
peptide
coronavirus
subject
Prior art date
Application number
PCT/US2021/021427
Other languages
French (fr)
Inventor
Longlong SI
Haiqing BAI
Rachelle PRANTIL-BAUN
Donald E. Ingber
Original Assignee
President And Fellows Of Harvard College
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by President And Fellows Of Harvard College filed Critical President And Fellows Of Harvard College
Publication of WO2021183463A1 publication Critical patent/WO2021183463A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/275Nitriles; Isonitriles
    • A61K31/277Nitriles; Isonitriles having a ring, e.g. verapamil
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/47064-Aminoquinolines; 8-Aminoquinolines, e.g. chloroquine, primaquine
    • 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
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20022New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/20011Coronaviridae
    • C12N2770/20033Use of viral protein as therapeutic agent other than vaccine, e.g. apoptosis inducing or anti-inflammatory

Definitions

  • SARS-CoV2 Severe Acute Respiratory Syndrome CoV
  • (Z)-toremifene e.g., FARESTON®
  • clomiphene e.g., CLOMID®
  • amodiaquine e.g., CAMOQUIN®
  • verapamil e.g., CALAN®
  • amiodarone e.g., NEXTERONE®
  • chloroquine e.g., ARALEN®
  • umifenovir e.g., ARBIDOL®
  • Baicalein is a natural product isolated from Chinese herbs, such as roots of Scutellaria baicalensis and Scutellaria lateriflora.
  • the synthetic HR2 peptide is derived from the HR2 domain of the spike protein of SARS-CoV2.
  • the data provided herein shows that the agents tested decrease viral infectivity and increase coronavirus inhibition rate (rate of inhibition of coronavirus infection), relative to a control.
  • Some aspects of the present disclosure provide methods for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of an agent (or a pharmaceutically acceptable salt of an agent) selected from the group consisting of (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide.
  • an agent or a pharmaceutically acceptable salt of an agent
  • compositions comprising a therapeutically effective amount of an agent (or a pharmaceutically acceptable salt of an agent) selected from the group consisting of (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide.
  • agent or a pharmaceutically acceptable salt of an agent
  • the coronavirus is a beta coronavirus.
  • the beta coronavirus may be SARS-CoV2.
  • the composition comprises a combination of agents (e.g., two, three, four, five, six, seven, eight, or nine agents) selected from the group consisting of (Z)- toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide.
  • the composition comprises (Z)-toremifene.
  • the composition comprises clomiphene.
  • the composition comprises amodiaquine.
  • the composition comprises verapamil.
  • the composition comprises umifenovir.
  • the composition comprises amiodarone. In some embodiments, the composition comprises chloroquine. In some embodiments, the composition comprises baicalein. In some embodiments, the composition comprises CoV HR2 peptide or a variant CoV HR2 peptide.
  • the composition comprises a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1. In some embodiments, the composition comprises a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1. In some embodiments, the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • the therapeutically effective amount is an amount effective for decreasing coronavirus infectivity, relative to a control.
  • the therapeutically effective amount may be an amount effective for decreasing viral infectivity by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control.
  • the therapeutically effective amount is an amount effective for increasing rate of inhibition of coronavirus infection, relative to a control.
  • the therapeutically effective amount may be an amount effective for increasing rate of inhibition of coronavirus infection by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control.
  • a control may be, for example, untreated subjects.
  • the composition further comprises a pharmaceutically-acceptable excipient.
  • the subject is a human subject. In some embodiments, the subject is immunocompromised. In some embodiments, the subject is a child or an elderly person.
  • the composition is administered nasally, orally, intravenously, intramuscularly, subcutaneously, or intraperitoneally. In some embodiments, the composition is administered via inhalation.
  • the composition is administered to the lung airway of the subject.
  • the composition is administered to the lung airway via an aerosol, a nebulizer, or a tracheal wash.
  • the composition is administered as a single dose.
  • the composition is administered as multiple doses.
  • the composition may be administered at multiple time points.
  • the composition is administered the composition weekly, monthly, every six months, yearly, every 5 years, or every 10 years.
  • compositions comprising a therapeutically effective amount of a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • compositions comprising a therapeutically effective amount of a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
  • a composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • composition further comprises a pharmaceutically-acceptable excipient.
  • compositions comprising a therapeutically effective amount of a SARS-CoV2 HR2 peptide that comprises an amino acid sequence having at least 95% identity (or 100% identity) to the amino acid sequence of SEQ ID NO: 1, wherein the composition is administered to a lung airway of the subject.
  • the composition is administered via an aerosol, a nebulizer, or a tracheal wash.
  • the coronavirus is a beta coronavirus, such as SARS-CoV2.
  • FIG. 1 The model of pseudotyped SARS-CoV2 (CoVpp) entry.
  • the genome of CoVpp is from the HIV genome but carries a luciferase reporter gene; while its envelope protein is the spike protein of coronavirus.
  • This pseudotyped virus can mimic the entry of coronavirus into host cells. Upon entry into host cells, we can detect report gene expression signal. If one drug can block viral entry, no signal can be detected. Using this model, we can test coronavirus entry inhibitors.
  • FIG. 2 The infection of CoVpp in Huh-7 cells.
  • Huh-7 (5,000 cells per well) was seeded into 96-well plate, cultured for 24 h, and infected with CoVpp in DMEM supplemented with 3% FBS. 72 h post-infection, luminescence was measured using the Bright-Glo reagent (Prornega) according to the manufacturer's protocol. The particles without the spike protein of SARS-CoV2 was used as control.
  • FIG. 3 Repurposing of FDA-approved drugs as entry inhibitors of SARS-CoV2.
  • Huh-7 cells 5,000 cell per well in 96-well plate
  • CoVpp infected with CoVpp in the presence or absence of test drugs at 1 uM or 5uM concentration.
  • Luciferase activity which correlates with the number of pseudoparticles in the host cells, was measured 3 days after infection using the Bright-Gio reagent (Prornega) according to the manufacturer's protocol.
  • Vesicular stomatitis vims envelope protein GP-carrying pseudotyped viral particle (VSVpp) was used to test the specificity and cytotoxicity of drugs.
  • Virus infectivity the presence and absence of drugs were calculated as above, *, P ⁇ 0.05; P ⁇ 0.01; ***, P ⁇ 0.001.
  • FIG. 4 Identification of SARS-CoV2 HR2 peptide and baicalein as entry inhibitors of SARS-CoV2.
  • Huh-7 cells 5,000 cell per well in 96-well plate) were infected with CoVpp in the presence or absence of test drugs.
  • the sequence of SARS-CoV2 HR2 peptide is GDIS GINAS V VNIQKEIDRFNE V AKNFNES FIDFQEFG (SEQ ID NO: 1).
  • Luciferase activity which correlates with the number of pseudoparticles in the host cells, was measured 3 days after infection using the Bright-Glo reagent (Promega) according to the manufacturer's protocol.
  • VSVpp Vesicular stomatitis virus envelope protein GP-carrying pseudotyped viral particle
  • FIGs. 5A-5B The expression of SARS-CoV2 receptor ACE2 and CoVpp infection in human Airway chip.
  • FIG. 5A Micrographs showing ZO-1, ACE2, Foxjl in the airway chip.
  • FIG. 5B Graph showing the infection of CoVpp in the airway chip. CoVpp was delivered into airway channel. 72 h later, cells were collected for detection of viral pol gene by qRT-PCR.
  • FIGs. 6A-6B The efficacy of nafamostat mesylate, ehloroquine diphosphate, arbidol hydrochloride, toremifene, ciomipbene citrate, amodiaqnine, verapamil hydrochloride, amiodarone.
  • the epithelium from the chips were collected for detection of viral pol gene by qRT-PCR; viral entry in untreated chips was set as 100%.
  • a t-test between the control and each compound (9 compounds + 1 protein — > 10 comparisons total) was performed, showing statistically significant viral inhibition in human Airway Chips following treatment with amodiaquine and toremifene (FIG. 6A).
  • Multiple hypothesis testing correction on these 10 comparisons were then performed to obtain the p- values for each compound.
  • the Bonferroni correction for multiple hypothesis testing showed statistically significant viral inhibition in human Airway Chips following treatment with amodiaquine, ciomipbene, and toremifene (FIG. 6B).
  • FIGs. 7A-7B Evaluation of the cytotoxicity of test drugs in Huh-7 cells and human Airway chips.
  • Huh-7 cells were treated with the test drugs at 1 or 5 mM for 48 h, and cell viability was measured by Celititer-Gio assay. The cell viability of untreated cells was set as 100%.
  • FIG. 7B Human Airway Chips were treated with the test drugs at their respective Cmax for 72 h, cell damage was measure by LDH assay. The LDH level of untreated human Airway Chips was set as 100%. Note that none of the drags produced any significant cytotoxicity at the doses used in these studies.
  • FIGs. 8A-8I Inhibition of infection by native SARS-CoV-2 virus in vitro and in vivo.
  • FIG. 8B Inhibition of wild type SARS-CoV-2 infection in ACE2-expressing A549 cells by 10 mM amodiaquine.
  • FIG. 8C Reduction of viral load in the lungs of hamsters treated once a day with amodiaquine (50 mg/kg) beginning 1 day prior to intranasal administration of SARS-CoV-2 virus (103 PFU) as measured by qPCR for subgenomic RNA encoding SARS-CoV-2 N protein. **, p ⁇ 0.01.
  • FIG. 8D Hematoxylin- and SARS-CoV-2 N-stained histological sections of lungs from animals that were mock treated, infected with SARS-CoV-2 and treated with vehicle alone, or infected with SARS-CoV-2 and treated with amodiaquine (50 mg/kg subcutaneously).
  • FIG. 8E Reduction of viral load in the lungs of hamsters treated once a day for 4 days with amodiaquine (50 mg/kg) beginning 1 day prior to co-caging with SARS-CoV-2 infected animals as measured by qPCR for RNA encoding SARS-CoV-2 N protein. *, p ⁇ 0.05.
  • FIG. 9 Effects of desethylamodiaquine on pseudotyped SARS-CoV-2 viral entry in human Airway Chips.
  • Desethylamodiaquine was delivered into apical and basal channels of the chip at its Cmax in human blood (1 mM), and one day later chips were infected with SARS-CoV- 2pp while in the continued presence of the drugs for 2 days.
  • the epithelium from the chips were collected for detection of viral gene by qRT-PCR; viral entry in untreated chips was set as 100%.
  • FIG. 10 PK profiles for amodiaquine and desethylamodiaquine administered subcutaneously in hamsters. Plasma concentration-time profiles showing mean concentration ( ⁇ s.d.) of amodiaquine (left) and desethylamodiaquine (right) at different time points after a single subcutaneous injection of amodiaquine (50 mg/kg).
  • FIG. 11 PK profiles for amodiaquine and desethylamodiaquine administered orally in hamsters. Plasma concentration-time profiles showing mean concentration ( ⁇ s.d.) of amodiaquine (left) and desethylamodiaquine (right) at different time points after a administration of a single dose of amodiaquine (75 mg/kg) via oral gavage.
  • FIGs. 12A-12B Effects of Amodiaquine treatment on gene expression in hamsters infected with SARS-CoV-2.
  • FIG. 12A Volcano plot showing distribution of expressed genes in hamster lungs infected with native SARS-CoV-2 and treated with amodiaquine compared to vehicle.
  • FIG. 12B Reduction of viral load measured in the lungs of hamsters in the study in which RNA seq data were obtained that are shown in (FIG. 12A).
  • compositions and methods for treating and/or preventing coronavirus infection such as SARS-CoV2 infection.
  • the compositions used herein include at least one agent selected from (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide (e.g., SEQ ID NO: 1).
  • Coronavimses are a large family of zoonotic viruses that are transmitted between animals and people, causing illness ranging from the common cold to more severe diseases such as Middle East Respiratory Syndrome (MERS-CoV) and Severe Acute Respiratory Syndrome (SARS-CoV).
  • MERS-CoV Middle East Respiratory Syndrome
  • SARS-CoV Severe Acute Respiratory Syndrome
  • coronaviruses include coronavirus 229E and NL63, which are common human alpha coronaviruses, and OC43 and HKU1, which are common human beta coronaviruses.
  • the methods and composition provided herein are used to treat an alpha coronavirus.
  • the methods and composition provided herein are used to treat a beta coronavirus.
  • Several known coronaviruses are circulating in animals that have not yet infected humans.
  • coronavirus infection Common signs of coronavirus infection include respiratory symptoms, fever, cough, shortness of breath, and breathing difficulties. In more severe cases, infection can cause pneumonia, severe acute respiratory syndrome, kidney failure, and even death.
  • COVID-19 coronavirus disease 2019
  • COVID-19 ‘corona,’ ‘VI’ for ‘vims,’ and ‘D’ for disease.
  • 2019 novel coronavirus or “2019-nCoV.”
  • the coronavirus infection being treated or prevented is COVID-19, also referred to as SARS-CoV2.
  • compositions provided herein include at least one of the following agents selected from the group consisting of: (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide.
  • Pharmaceutically acceptable salts of (Z)- (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, and baicalein are also encompassed by the present disclosure.
  • (Z)-Toremifene (IUPAC Name: 2-[4-[(Z)-4-chloro-1,2-diphenylbut-l-enyl]phenoxy]- N,N-dimethylethanamine) is a nonsteroidal triphenylethylene antiestrogen (see, e.g., Abeam, abl42467; CAS Number 89778-26-7).
  • toremifene is a selective estrogen receptor modulator (SERM). This agent binds competitively to estrogen receptors, thereby interfering with estrogen activity.
  • SERM selective estrogen receptor modulator
  • Toremifene has approved by the FDA for the treatment of metastatic breast cancer in postmenopausal women having estrogen receptor-positive or unknown tumors (FDA drug sheet, “FARESTON®”; reference ID: 4097849, 2017).
  • a therapeutically effective amount of (Z)-toremifene is administered.
  • (Z)-toremifene in some embodiments, may be administered orally as a 60 mg dose daily (Mari, The APRN and PA's Complete Guide to Prescribing Drug Therapy 2020; page 60).
  • the therapeutically effective amount of (Z)-toremifene can be a dose of 30 mg - 120 mg, 30 mg - 110 mg, 30 mg - 100 mg, 30 mg - 90 mg, 30 mg - 80 mg, 30 mg - 70 mg, 30 mg - 60 mg, 30 mg - 50 mg, 30 mg - 40 mg, 40 mg -120 mg, 40 mg - 110 mg, 40 mg - 100 mg, 40 mg - 90 mg, 40 mg - 80 mg, 40 mg - 70 mg, 40 mg - 60 mg, 40 mg - 50 mg, 50 mg
  • the therapeutically effective amount of (Z)-toremifene is 30 mg, 60 mg, 90 mg, 120 mg, 150 mg,
  • the therapeutically effective amount of (Z)- toremifene is 60 mg. In some embodiments, the (Z)-toremifene is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of (Z)-toremifene administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • Amodiaquine (IUPAC Name: 4-[(7-chloroquinolin-4-yl)amino]-2- (diethylaminomethyl)phenol), e.g., amodiaquin dihydrochloride dihydrate (see, e.g., MilliporeSigma, Cat. A2799; CAS Number 398-98-7), is an orally active 4-aminoquinoline derivative with antimalarial and anti-inflammatory properties. Similar in structure and activity to chloroquine, amodiaquine is effective against some chloroquine-resistant strains, particularly Plasmodium falciparum, the most deadly malaria parasite.
  • amodiaquine likely is able to inhibit heme polymerase activity in the body. This results in accumulation of free heme, which is toxic to the parasites. See U.S. National Library of Medicine, PubChem entry for “Amodiaquine.” Amodiaquine is an FDA-approved 4-aminoquinoline antimalarial drug, which is known to inhibit rRNA transcription, a rate-limiting step in the ribosome biosynthesis pathway (Espinoza et al, Cell Death & Diff. 2020, 27:773-789). In some embodiments, a therapeutically effective amount of amodiaquin dihydrochloride dihydrate is administered.
  • a therapeutically effective amount of amodiaquine e.g., AMDAQUINE®, AMOBIN®
  • a therapeutically effective amount of amodiaquin dihydrochloride dihydrate is administered.
  • Amodiaquine in some embodiments, may be administered orally as a 100 mg or 200 mg dose (Cairns et al, Antimicrobial Agents and Chemother. 2010, 54(3): 1265-74).
  • the therapeutically effective amount of amodiaquine can be a dose of 50 mg - 300 mg, 50 mg - 275 mg, 50 mg - 250 mg, 50 mg - 225 mg, 50 mg - 200 mg, 50 mg - 175 mg, 50 mg - 150 mg, 50 mg - 125 mg, 50 mg - 100 mg, 50 mg -75 mg, 75 mg - 300 mg, 75 mg - 275 mg, 75 mg - 250 mg, 75 mg - 225 mg, 75 mg - 200 mg, 75 mg - 175 mg, 75 mg - 150 mg, 75 mg - 125 mg, 75 mg - 100 mg, 100 mg -
  • the therapeutically effective amount of amodiaquine is 50 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, or 400 mg. In some embodiments, the therapeutically effective amount of amodiaquine is 200 mg. In some embodiments, the amodiaquine is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of amodiaquine administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • Clomiphene (IUPAC Name: 2-[4-(2-chloro-1,2-diphenylethenyl)phenoxy]-N,N- diethylethanamine) is a triphenyl ethylene stilbene derivative which is an estrogen agonist or antagonist depending on the target tissue.
  • Clomiphene has both estrogenic and anti-estrogenic properties, but its precise mechanism of action has not been determined.
  • Clomiphene appears to stimulate the release of gonadotropins, follicle- stimulating hormone (FSH), and luteinizing hormone (LH), which leads to the development and maturation of ovarian follicle, ovulation, and subsequent development and function of the corpus luteum, thus resulting in pregnancy.
  • FSH follicle- stimulating hormone
  • LH luteinizing hormone
  • Gonadotropin release may result from direct stimulation of the hypothalamic-pituitary axis or from a decreased inhibitory influence of estrogens on the hypothalamic-pituitary axis by competing with the endogenous estrogens of the uterus, pituitary, or hypothalamus.
  • Clomiphene has no apparent progestational, androgenic, or antiandrogenic effects and does not appear to interfere with pituitary- adrenal or pituitary-thyroid function. See U.S. National Library of Medicine, PubChem entry for “Clomiphene.”
  • a therapeutically effective amount of clomiphene also known as clomifene (e.g., BECLOM®, BEMOT®, BLESIFEN®, CHLORAMIPHENE®, CLOFERT®, CLOMENE®, CLOMHEXAL®, CLOM®, CLOMID®, CLOMIDAC®, CLOMIFEN®, CLOMIFENCITRAT®, CLOMIFENE®, CLOMIFENE®, CLOMIFENE CITRATE®, CLOMIFENI CITRAS®, CLOMIFENO®, CLOMIFERT®, CLOMIHEXAL®, CLOMIPHEN, CLOMIPHENE®, CLOMIPHENE CITRATE®, CLONINN®, CLOSTIL®, CLOSTILBEGYT®, CLOVERTIL®, CLOVUL®, DIPTHEN®, DUFINE®, DUINUM®, FENSIPROS®, FERTAB®, FERTEC®, FERTEX®, FERTICLO
  • a therapeutically effective amount of clomiphene citrate is administered.
  • Clomiphene has been approved by the FDA for treatment of ovulatory dysfunction in women who want to become pregnant (FDA drug sheet, “CLOMID®”; reference ID: 3206435, 2012).
  • Clomiphene in some embodiments, may be administered at a dose of 50 mg per day for five days (Mayo Clinic, “Clomiphene,” 2020).
  • the therapeutically effective dose of clomiphene can be a dose of 10 mg - 150 mg, 10 mg - 125 mg, 10 mg - 100 mg, 10 mg - 100 mg, 10 mg - 90 mg, 10 mg - 80 mg, 10 mg - 75 mg, 10 mg - 70 mg, 10 mg - 60 mg, 10 mg - 50 mg, 10 mg - 40 mg, 10 mg - 30 mg, 10 mg - 20 mg, 25 mg - 150 mg, 25 mg - 125 mg, 25 mg - 100 mg, 25 mg - 90 mg, 25 mg - 80 mg, 25 mg - 75 mg, 25 mg - 70 mg, 25 mg - 60 mg, 25 mg - 50 mg,
  • the therapeutically effective dose of clomiphene is 25 mg, 50 mg, 75 mg,
  • the therapeutically effective amount of clomiphene is 50 mg.
  • the clomiphene is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly.
  • a therapeutically effective amount of clomiphene administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • Verapamil (IUPAC Name: 2-(3,4-dimethoxyphenyl)-5-[2-(3,4-dimethoxyphenyl)ethyl- methylamino]-2-propan-2-ylpentanenitrile) is a phenylalkylamine calcium channel blocking agent. Verapamil inhibits the transmembrane influx of extracellular calcium ions into myocardial and vascular smooth muscle cells, causing dilatation of the main coronary and systemic arteries and decreasing myocardial contractility. This agent also inhibits the drug efflux pump P- glycoprotein which is overexpressed in some multi-drug resistant tumors and may improve the efficacy of some antineoplastic agents. See U.S. National Library of Medicine, PubChem entry for “Verapamil.”
  • a therapeutically effective amount of verapamil e.g., CALAN®, VERELAN®
  • a therapeutically effective amount of verapamil hydrochloride is administered.
  • Verapamil benzeneacetonitrile, a-[3-[[2-(3,4- dimethoxyphenyl)ethyl] methylamino]propyl]-3,4-dimethoxya-(l-methylethyl) hydrochloride
  • Verapamil is an FDA-approved calcium antagonist or slow-channel inhibitor for the treatment of supraventricular tachyarrhythmias (FDA drug sheet, “Verapamil”; reference ID: 4015901, 2016).
  • Verapamil in some embodiments, may be administered intravenously, for example as a 5 mg or
  • the therapeutically effective amount of verapamil can be a dose of 1 mg - 30 mg, 1 mg - 25 mg, 1 mg - 20 mg, 1 mg - 18 mg, 1 mg - 15 mg, 1 mg - 14 mg, 1 mg - 13 mg, 1 mg - 12 mg, 1 mg - 11 mg, 1 mg -10 mg, 1 mg - 9 mg, 1 mg - 8 mg, 1 mg - 7 mg, 1 mg - 6 mg,
  • the therapeutically effective amount of verapamil is 5 mg, 10 mg, 15 mg, 20 mg, 250 mg, 30 mg, or 40 mg. In some embodiments, the therapeutically effective amount of verapamil is 10 mg. In some embodiments, the verapamil is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of verapamil administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • Umifenovir (IUPAC Name: ethyl 6-bromo-4-[(dimethylamino)methyl]-5-hydroxy-l- methyl-2-(phenylsulfanylmethyl)indole-3-carboxylate) has been approved for use in Russia and China as a broad spectrum antiviral compound to treat influenza (Haviernik et al, Viruses, 2018, 10(4):E184).
  • a therapeutically effective amount of umifenovir e.g., a compound having the following properties of a therapeutically effective amount of umifenovir (e.g., a compound having the following properties of a therapeutically effective amount of umifenovir (e.g., a compound having the following properties of a therapeutically effective amount of umifenovir (e.g., a compound having the following properties of a therapeutically effective amount of umifenovir (e.g.,
  • ARB IDOL® is administered.
  • a therapeutically effective amount of arbidol hydrochloride is administered.
  • Umifenovir in some embodiments, may be administered orally in 400 mg doses (Pshenichnaya et al, Ter Arkh. 2019, 91(3): 56-63).
  • the therapeutically effective amount of umifenovir can be a dose of 200 mg - 1000 mg, 200 mg - 800 mg, 200 mg - 750 mg, 200 mg - 700 mg, 200 mg - 600 mg, 200 mg - 500 mg, 200 mg - 400 mg, 200 mg - 300 mg, 300 mg - 1000 mg, 300 mg - 800 mg, 300 mg - 750 mg, 300 mg - 700 mg, 300 mg - 600 mg, 300 mg - 500 mg, 300 mg - 400 mg, 400 mg - 1000 mg, 400 mg - 800 mg, 400 mg - 750 mg, 400 mg - 700 mg, 400 mg - 600 mg, 400 mg - 500 mg, 500 mg - 1000 mg, 500 mg - 800 mg, 500 mg - 750 mg, 500 mg - 700 mg, 500 mg - 600 mg, 600 mg - 1000 mg, 600 mg - 800 mg, 600 mg - 750 mg, 600 mg - 700 mg, 500 mg - 600 mg, 600 mg - 1000 mg, 600 mg - 800 mg, 600
  • the therapeutically effective amount of verapamil is 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, or 1600 mg.
  • the therapeutically effective amount of umifenovir is 400 mg.
  • the umifenovir is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly.
  • a therapeutically effective amount of umifenovir administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • Amiodarone (2- ⁇ 4- [(2-butyl- 1 -benzofuran-3 -yl)carbonyl] -2,6- diiodophenoxy ⁇ ethyl)diethylamine is an FDA-approved antiarrhythmic drug having predominantly Class III (Vaughn Williams’ classification) effects for use in life-threatening recurrent ventricular arrhythmias that have not responded to adequate doses of other antiarrhythmic agents (FDA drug sheet, “CORDARONE®”; reference ID: 2876651).
  • a therapeutically effective amount of amiodarone e.g., NEXTERONE®, CORDARONE®
  • a therapeutically effective amount of amiodarone hydrochloride is administered.
  • Amiodarone in some embodiments, may be administered orally in 800 mg doses (id.).
  • the therapeutically effective amount of amiodarone can be a dose of 100 mg - 2000 mg, 100 mg - 1800 mg, 100 mg - 1700 mg, 100 mg - 1600 mg, 100 mg - 1500 mg, 100 mg - 1400 mg, 100 mg - 1300 mg, 100 mg - 1200 mg, 100 mg - 1100 mg, 100 mg - 1000 mg, 100 mg - 900 mg, 100 mg - 800 mg, 100 mg
  • the therapeutically effective amount of amiodarone is 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1600 mg, or 2000 mg. In some embodiments, the therapeutically effective amount of amiodarone is 600 mg. In some embodiments, the amiodarone is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of amiodarone administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • Chloroquine (IUPAC Name: 4-N-(7-chloroquinolin-4-yl)-l-N,l-N-diethylpentane-1,4- diamine), e.g., chloroquine diphosphate, is a 4-aminoquinoline with antimalarial, anti- inflammatory, and potential chemosensitization and radiosensitization activities (see, e.g., Abeam, ab142116; CAS Number 50-63-5) Although the mechanism is not well understood, chloroquine is shown to inhibit the parasitic enzyme heme polymerase that converts the toxic heme into non-toxic hemazoin, thereby resulting in the accumulation of toxic heme within the parasite.
  • Chloroquine's potential chemosensitizing and radiosensitizing activities in cancer may be related to its inhibition of autophagy, a cellular mechanism involving lysosomal degradation that minimizes the production of reactive oxygen species (ROS) related to tumor reoxygenation and tumor exposure to chemotherapeutic agents and radiation.
  • ROS reactive oxygen species
  • chloroquine is an FDA-approved 4-aminoquinoline compound for the suppressive treatment and for acute attacks of malaria due to P. vivax, P. malariae, P. ovale, and susceptible strains of P. falciparum, in addition to extraintestinal amebiasis (FDA drug sheet, “ARALEN®”; reference ID: 3402523, 2013).
  • Chloroquine in some embodiments, may be administered orally in 300 mg doses (id.).
  • the therapeutically effective amount of amiodarone can be a dose of 100 mg - 900 mg, 100 mg - 800 mg, 100 mg - 700 mg, 100 mg - 600 mg, 100 mg - 500 mg, 100 mg - 400 mg, 100 mg - 300 mg, 100 mg - 200 mg, 200 mg - 900 mg, 200 mg - 800 mg, 200 mg - 700 mg, 200 mg - 600 mg, 200 mg - 500 mg, 200 mg - 400 mg, 200 mg - 300 mg, 300 mg - 900 mg, 300 mg - 800 mg, 300 mg - 700 mg, 300 mg - 600 mg, 300 mg - 500 mg, 300 mg - 400 mg, 400 mg - 900 mg, 400 mg - 800 mg, 400 mg - 700 mg, 400 mg - 600 mg, 400 mg - 500 mg, 500 mg - 600 mg, 400 mg, 400 mg - 900 mg, 400 mg - 800 mg, 400 mg - 700 mg, 400 mg - 600 mg, 400 mg - 500 mg, 500 mg - 600
  • the therapeutically effective amount of chloroquine is 150 mg, 300 mg, 450 mg, 600 mg, 750 mg, 900 mg, 1050 mg, or 1200 mg. In some embodiments, the therapeutically effective amount of chloroquine is 600 mg. In some embodiments, the chloroquine is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of chloroquine administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • Baicalein (IUPAC Name: 5,6,7-trihydroxy-2-phenylchromen-4-one) is a trihydroxyflavone with the hydroxy groups at positions C-5, -6 and -7. It has a role as an antioxidant, a hormone antagonist, a prostaglandin antagonist, an EC 1.13.11.31 (arachidonate 12-lipoxygenase) inhibitor, an EC 1.13.11.33 (arachidonate 15-lipoxygenase) inhibitor, a radical scavenger, an EC 3.4.21.26 (prolyl oligopeptidase) inhibitor, an anti-inflammatory agent and a plant metabolite. It is a conjugate acid of a baicalein(1-). See U.S.
  • Baicalein is a flavone, which was initially isolated from Scutellaria baicalensis and S. lateriflora.
  • the compound is a positive allosteric modulator of benzodiazepine site or the non-benzodiazepine site of the GABA A receptor and is selective for a and ⁇ 3 subunit-containing GABA A receptors, in addition to being an anti-inflammatory agent (Wang et al., Neuropharmacol, 2008, 55(7): 1231-7). It is also used in traditional Chinese medicine to treat inflammation, hepatitis, infections, and tumors (Liu et al., Front Pharmacol. 2019, 10:518).
  • the compound in some embodiments, may be administered orally, for example, as a 1000 mg dose (Li et al., J Ethnopharmacol. 2014, 154:210-5).
  • a therapeutically effective amount of baicalein is administered.
  • the therapeutically effective amount of baicalein can be a dose of 100 mg - 3000 mg, 100 mg - 2800 mg, 100 mg - 2600 mg, 100 mg - 2500 mg, 100 mg - 2200 mg, 100 mg - 2000 mg, 100 mg - 1800 mg, 100 mg - 1600 mg, 100 mg - 1500 mg, 100 mg - 1400 mg, 100 mg - 1200 mg, 100 mg - 1000 mg, 100 mg - 900 mg, 100 mg - 800 mg, 100 mg - 700 mg, 100 mg - 600 mg, 100 mg - 500 mg, 100 mg - 400 mg, 100 mg - 300 mg, 100 mg - 200 mg, 200 mg - 3000 mg, 200 mg - 2800 mg, 200 mg - 2600 mg, 200 mg - 2500 mg, 200 mg - 2200 mg, 200 mg - 2000 mg, 200 mg - 1800 mg, 200 mg - 1600 mg, 200 mg - 1500 mg, 200 mg - 1400
  • the therapeutically effective amount of baicalein is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1500 mg, 2000 mg, 2250 mg, 2500 mg, or 2800 mg. In some embodiments, the therapeutically effective amount of baicalein is 600 mg. In some embodiments, the baicalein is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of baicalein administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • the coronavirus spike protein is a type I membrane protein that associates into trimers on the surface of coronavirus membrane.
  • the distal subunit (S1) of the spike protein contains the receptor binding domain, and the membrane- anchored subunit (S2) contains a putative internal fusion peptide and two heptad repeat regions (HR1 and HR2).
  • the CoV HR2 peptide comprises an amino acid sequence having at least 95% identity to GDIS GINAS V VNIQKEIDRLNE V AKNLNES LIDLQELG (SEQ ID NO: 1).
  • the CoV HR2 peptide comprises the amino acid sequence of SEQ ID NO: 1.
  • identity refers to a relationship between the sequences of two or more peptides, as determined by comparing the sequences. Identity also refers to the degree of sequence relatedness between or among sequences as determined by the number of matches between strings of two or more amino acid residues. Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program. Identity of related peptides can be readily calculated by known methods. “Percent (%) identity” as it applies to peptide sequences is defined as the percentage of amino acid residues of a first sequence that is identical with the amino acid residues of a second sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity. Methods and computer programs for the alignment are well known in the art. It is understood that identity depends on a calculation of percent identity but may differ in value due to gaps and penalties introduced in the calculation.
  • a therapeutically effective amount of CoV HR2 peptide is administered.
  • the therapeutically effective amount of CoV HR2 peptide can be a dose of 1 mg - 50 mg, lmg - 45 mg, 1 mg - 40 mg, 1 mg - 35 mg, 1 mg - 30 mg, 1 mg - 25 mg, 1 mg - 20 mg, 1 mg - 15 mg, 1 mg - 10 mg, 5 mg - 50 mg, 5 mg - 45 mg, 5 mg - 40 mg, 5 mg - 35 mg, 5 mg
  • the therapeutically effective amount of CoV HR2 peptide is 1 mg, 2 mg, 5 mg, 10 mg, 20 mg, or 30 mg. In some embodiments, the therapeutically effective amount of CoV HR2 peptide is 10 mg.
  • the CoV HR2 peptide is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly.
  • a therapeutically effective amount of CoV HR2 peptide administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
  • an agent, or a combination of agents is administered in an amount effective for decreasing coronavirus infectivity.
  • coronavirus infectivity is decreased by at least 20%, relative to a control.
  • coronavirus infectivity may be decreased by at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%, relative to a control.
  • coronavirus infectivity is decreased by 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%- 60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60%, relative to a control.
  • an agent, or a combination of agents is administered in an amount effective for increasing the rate of inhibition of coronavirus infection.
  • the rate of inhibition of coronavirus infection is increased by at least 20%, relative to a control.
  • coronavirus infectivity may be increased by at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%, relative to a control.
  • the rate of inhibition of coronavirus infection is increased by 20%-100%, 20%- 90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%- 100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60%, relative to a control.
  • pharmaceutically acceptable salt refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of the present disclosure include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid or by using other methods known in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid or by using other methods known in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy- ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium, and N+(C1-4 alky1)4- salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate.
  • compositions provided herein include at least one pharmaceutically-acceptable excipient (e.g., carrier, buffer, and/or salt, etc.).
  • excipient e.g., carrier, buffer, and/or salt, etc.
  • a molecule or other substance/agent is considered “pharmaceutically acceptable” if it is approved or approvable by a regulatory agency of the Federal government or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans.
  • An excipient may be any inert (inactive), non-toxic agent, administered in combination with an agent provided herein.
  • Non-limiting examples of excipients include buffers (e.g ., sterile saline), salts, carriers, preservatives, fillers, surfactants, and coloring agents.
  • compositions provided herein may include, or may be administered in combination with, other agents, such as antiviral agents, antibacterial agents, and/or anti-inflammatory agents (e.g., nelfinavir, resdemivir, dexamethasone, and/or baricitinib).
  • agents such as antiviral agents, antibacterial agents, and/or anti-inflammatory agents (e.g., nelfinavir, resdemivir, dexamethasone, and/or baricitinib).
  • a composition provided herein includes, or is administered in combination with, an agent used to treat a secondary condition associated with coronavirus infection, such as pneumonia.
  • a method comprises administering to a subject a combination of two or more agents selected from (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide.
  • a method comprises administering to a subject a combination of (Z)- toremifene and clomiphene.
  • a method comprises administering to a subject a combination of (Z)-toremifene and amodiaquine.
  • a method comprises administering to a subject a combination of (Z)-toremifene and verapamil. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and umifenovir. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and amiodarone. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and chloroquine. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and baicalein.
  • a method comprises administering to a subject a combination of (Z)-toremifene and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of clomiphene and amodiaquine. In some embodiments, a method comprises administering to a subject a combination of clomiphene and verapamil. In some embodiments, a method comprises administering to a subject a combination of clomiphene and umifenovir. In some embodiments, a method comprises administering to a subject a combination of clomiphene and amiodarone. In some embodiments, a method comprises administering to a subject a combination of clomiphene and chloroquine.
  • a method comprises administering to a subject a combination of clomiphene and baicalein. In some embodiments, a method comprises administering to a subject a combination of clomiphene and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and verapamil. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and umifenovir. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and amiodarone. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and chloroquine.
  • a method comprises administering to a subject a combination of amodiaquine and baicalein. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of verapamil and umifenovir. In some embodiments, a method comprises administering to a subject a combination of verapamil and amiodarone.
  • a method comprises administering to a subject a combination of verapamil and chloroquine. In some embodiments, a method comprises administering to a subject a combination of verapamil and baicalein. In some embodiments, a method comprises administering to a subject a combination of verapamil and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of umifenovir and amiodarone. In some embodiments, a method comprises administering to a subject a combination of umifenovir and chloroquine. In some embodiments, a method comprises administering to a subject a combination of umifenovir and baicalein.
  • a method comprises administering to a subject a combination of umifenovir and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of amiodarone and chloroquine. In some embodiments, a method comprises administering to a subject a combination of amiodarone and baicalein. In some embodiments, a method comprises administering to a subject a combination of amiodarone and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of chloroquine and baicalein. In some embodiments, a method comprises administering to a subject a combination of chloroquine and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of baicalein and CoV HR2 peptide.
  • a composition provided herein includes two or more agents selected from (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide.
  • a composition comprises (Z)-toremifene and clomiphene.
  • a composition comprises (Z)-toremifene and amodiaquine.
  • a composition comprises (Z)-toremifene and verapamil.
  • a composition comprises (Z)-toremifene and umifenovir.
  • a composition comprises (Z)-toremifene and amiodarone. In some embodiments, a composition comprises (Z)-toremifene and chloroquine. In some embodiments, a composition comprises (Z)-toremifene and baicalein. In some embodiments, a composition comprises (Z)-toremifene and CoV HR2 peptide. In some embodiments, a composition comprises clomiphene and amodiaquine. In some embodiments, a composition comprises clomiphene and verapamil. In some embodiments, a composition comprises clomiphene and umifenovir. In some embodiments, a composition comprises clomiphene and amiodarone.
  • a composition comprises clomiphene and chloroquine. In some embodiments, a composition comprises clomiphene and baicalein. In some embodiments, a composition comprises clomiphene and CoV HR2 peptide. In some embodiments, a composition comprises amodiaquine and verapamil. In some embodiments, a composition comprises amodiaquine and umifenovir. In some embodiments, a composition comprises amodiaquine and amiodarone. In some embodiments, a composition comprises amodiaquine and chloroquine. In some embodiments, a composition comprises amodiaquine and baicalein. In some embodiments, a composition comprises amodiaquine and CoV HR2 peptide.
  • a composition comprises amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide.
  • a composition comprises verapamil and umifenovir.
  • a composition comprises verapamil and amiodarone.
  • a composition comprises verapamil and chloroquine.
  • a composition comprises verapamil and baicalein.
  • a composition comprises verapamil and CoV HR2 peptide.
  • a composition comprises umifenovir and amiodarone.
  • a composition comprises umifenovir and chloroquine.
  • a composition comprises umifenovir and baicalein. In some embodiments, a composition comprises umifenovir and CoV HR2 peptide. In some embodiments, a composition comprises amiodarone and chloroquine. In some embodiments, a composition comprises amiodarone and baicalein. In some embodiments, a composition comprises amiodarone and CoV HR2 peptide. In some embodiments, a composition comprises chloroquine and baicalein. In some embodiments, a composition comprises chloroquine and CoV HR2 peptide. In some embodiments, a composition comprises baicalein and CoV HR2 peptide.
  • composition and methods of the present disclosure may be used to prevent or treat coronavirus (e.g., SARS-CoV2) infection in any subject, such as a human subject.
  • a subject is a child, who is between the ages of 1 year and 5 years.
  • a subject is an elderly person, who is 65 years old, or older.
  • a subject is immunocompromised (having an impaired or weakened immune system). In some embodiments, a subject has another comorbidity, such as heart disease or liver disease.
  • compositions provided herein may vary depending on the specific agents.
  • a composition comprising an agent is administered orally (e.g., as a liquid suspension or tablet).
  • a composition comprising an agent is administered nasally, intravenously, intramuscularly, subcutaneously, or intraperitoneally.
  • the composition is administered via inhalation.
  • a composition is administered to the lung airway, for example, via aerosol, nebulizer, or tracheal wash.
  • compositions herein may be administered as a single dose or as multiple doses (e.g., a booster dose or multiple booster doses). For example, a composition may be administered weekly, monthly, every six months, yearly, every 5 years, or every 10 years.
  • a composition is administered as a therapeutic intervention after diagnosis of viral infection.
  • a composition is administered as a prophylactic treatment before diagnosis of viral infection.
  • a method for treating coronavirus infection comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of an agent selected from the group consisting of (Z)-toremifene, clomiphene (e.g., clomiphene citrate), amodiaquine (e.g., amodiaquin dihydrochloride dehydrate), verapamil (e.g., verapamil hydrochloride), umifenovir (e.g., arbidol hydrochloride), amiodarone (e.g., amiodarone hydrochloride), chloroquine (e.g., chloroquine diphosphate), baicalein, and CoV HR2 peptide.
  • an agent selected from the group consisting of (Z)-toremifene, clomiphene (e.g., clomiphene citrate), amodiaquine (e.g., amodiaquin dihydrochloride dehydrate), verapa
  • a method for preventing coronavirus infection comprising administering to a subject at risk of coronavirus a composition comprising a therapeutically effective amount of an agent selected from the group consisting of (Z)-toremifene, clomiphene (e.g., clomiphene citrate), amodiaquine (e.g., amodiaquin dihydrochloride dehydrate), verapamil (e.g., verapamil hydrochloride), umifenovir (e.g., arbidol hydrochloride), amiodarone (e.g., amiodarone hydrochloride), chloroquine (e.g., chloroquine diphosphate), baicalein, and CoV HR2 peptide.
  • an agent selected from the group consisting of (Z)-toremifene, clomiphene (e.g., clomiphene citrate), amodiaquine (e.g., amodiaquin dihydrochloride dehydrate), verapa
  • composition comprises two, three, four, five, six, seven, eight, or nine agents selected from the group consisting of (Z)- toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide.
  • agents selected from the group consisting of (Z)- toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide.
  • composition comprises clomiphene.
  • composition comprises amodiaquine.
  • composition comprises verapamil.
  • composition comprises umifenovir.
  • composition comprises amiodarone.
  • composition comprises chloroquine
  • composition comprises baicalein.
  • composition comprises a CoV HR2 peptide or a variant CoV HR2 peptide.
  • composition comprises a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • composition comprises a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
  • composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • the therapeutically effective amount is an amount effective for decreasing coronavirus infectivity.
  • the therapeutically effective amount is an amount effective for decreasing viral infectivity by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control.
  • the therapeutically effective amount is an amount effective for increasing the rate of inhibition of coronavirus infection, relative to a control.
  • the therapeutically effective amount is an amount effective for increasing the rate of inhibition of coronavirus infection by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control.
  • composition further comprises a pharmaceutically-acceptable excipient.
  • composition is administered nasally, orally, intravenously, intramuscularly, subcutaneously, or intraperitoneally.
  • composition is administered to the lung airway via an aerosol, a nebulizer, or a tracheal wash.
  • composition is administered as multiple doses.
  • composition is administered weekly, monthly, every six months, yearly, every 5 years, or every 10 years.
  • composition comprising at least two agents selected from the group consisting of (Z)- toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide.
  • composition comprising a therapeutically effective amount of a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • composition comprising a therapeutically effective amount of a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
  • composition of paragraph 35 wherein the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • composition of any one of paragraphs 33-35 further comprising a pharmaceutically- acceptable excipient.
  • a method for treating coronavirus infection comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of a SARS-CoV2 HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1, wherein the composition is administered to a lung airway of the subject.
  • composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
  • composition is administered via an aerosol, a nebulizer, or a tracheal wash.
  • the lifecycle of viruses includes three main steps: entry into host cells, replicate in host cells, and release from host cells. Inhibition of the first step (viral entry) should reduce virus propagation and dampen infection-related inflammatory responses. Therefore, entry inhibitors represent a new generation of antiviral drugs. 6
  • SARS-CoV2 entry inhibitors we generated SARS-CoV2 pseudoparticles (CoVpp) by packaging the SARS-CoV2 spike protein into a retroviral core (FIG. 1). This pseudovirus allows us to develop potential inhibitors for viral entry while alleviating biosafety concerns.
  • CoVpp The generation of CoVpp was conducted as described previously. 7 Briefly, HEK293T cells (5 x 10 5 ') were seeded into six- well plates 24 hours before transfection. Then, HEK293T cells were cotransfected with pNL4-3 plasmid, a pCMV3 plasmid that encodes the viral spike protein of SARS-CoV2, and pAdvantage plasmid using the transfection reagent MegaTran 1.0 (OriGene) according to the manufacturer's instructions. Supernatant was collected 72 hours later to obtain SARS-CoV2 spike protein pseudotyped virions (CoVpp).
  • the maximum infeetivity (100%) was derived from the control wells containing DMSO control; background (0%) from uninfected wells.
  • background (0%) from uninfected wells.
  • the luciferase intensity in each of the compound testing wells were subtracted by the background signals, divided by the average signals of the control values (DMSO control), and multiplied by 100%.
  • ARALEN® hydroxychloroquine
  • umifenovir e.g., ARB IDOL®
  • FIG. 3 data not shown for hydroxychloroquine, which was similar to the chloroquine data
  • FIGs. 7A-7B data not shown for hydroxychloroquine, which was similar to the chloroquine data
  • chloroquine has been reported to inhibit SARS-CoV2 in vitro.
  • umifenovir inhibited viral entry.
  • CoV HR2 peptide which is derived from the HR2 domain of the spike protein of S ARS-CoV2
  • baicalein which is a natural product isolated from Chinese herbs, such as roots of Scutellaria baicalensis and Scutellaria lateriflora, exhibited inhibitory activity on the entry of SARS-CoV2 in a dose- dependent manner (FIG. 4).
  • Example 3 Evaluation of repurposed entry inhibitors of SARS-CoV2 in human Lung Airway Chips.
  • amodiaquine was an effective inhibitor of SARS-CoV-2 viral entry while the related anti-malarial drugs hydroxychloroquine and chloroquine were not, quantitative mass spectrometry was performed to compare the effects of these three drugs on the proteome of airway epithelial cells.
  • Proteomics analysis revealed that amodiaquine triggered distinct and broader pertubations in host proteome compared to the other related anti-malarial drugs (data not shown) with the most differentially affected proteins being related to regulation of cilia and expression of lysosomal proteins, which may be responsible for it having greater effects against viral entry.
  • amodiaquine reduced viral load by ⁇ 3 logs in ACE2-expressing human lung A549 cells infected with native SARS-CoV-2 when administered at 10 mM (FIG. 8B).
  • amodiaquine in a hamster COVID-19 prevention model in which the drug was first administered subcutaneously (50 mg/kg) one day before the animals were infected intranasally with SARS- CoV-2 vims (10 3 PFU), and then treated daily with the same dose for 3 additional days.
  • the animals were treated once a day for 4 days with amodiaquine (50 mg/kg via subcutaneous injection) beginning one day prior to SARS-CoV-2 infection.
  • the dosing regimen was selected based on a PK study for amodiaquine that was carried out in healthy hamsters in parallel.
  • Prophylaxis of infected hamsters with amodiaquine beginning 1 day before infection and treating daily over the following 3 days using this dosing regimen resulted in -70% reduction in SARS-CoV-2 viral load measured by RT-qPCR of the viral N transcript when measured on the third day after the viral challenge (FIG. 8C), which is the day at which peak viral loads are observed in this model.
  • Immunohistochemical analysis of lungs from these animals confirmed that amodiaquine treatment resulted in a significant reduction in expression of SARS-CoV-2 N protein in these tissues (FIG. 8D).
  • amodiaquine is commonly administered as an oral dose clinically, we repeated these studies with drug administered through oral gavage at a dose (75 mg/kg) that produced similar PK parameters to those observed with the subcutaneous administration (Cmax for amodiaquine and desethylamodiaquine of ⁇ 1.8 and 4.5 mM, respectively; Tl/2 for amodiaquine was 12.8 hours and again greater than 24 hours for its active metabolite (FIG. 11, Tables 5).
  • oral administration resulted in even higher levels in tissues compared to plasma (enhanced 29- to 331-fold for amodiaquine and 8- to 119-fold for desethylamodiaquine) (Table 6).
  • RNA seq analysis of infected hamsters treated with oral amodiaquine revealed that drug treatment resulted in a significant down regulation of genes associated with the inflammatory response, including those involved in signaling through TNF-a and NF-kB, IL-6 JAK STAT3, and interferon-g (FIG. 12, Table 7).
  • RNA Lung RNA was extracted by phenol chloroform extraction and DNase treated using DNA-free DNA removal kit (Invitrogen). After cDNA synthesis of RNA samples by reverse transcription using Superscript II Reverse Transcriptase (invitrogen) with oligo d(T) primers, quantitative RT-PCR was performed using KAPA SYBR FAST qPCR Master Mix Kit (Kapa Biosystems) on a LightCycler 480 Instrument II (Roche) for subgenomic nucleocapsid (N) RNA (sgRNA) and actin using the following primers: Actin forward primer: 5’- CC AAGGCC AACCGTGAAAAG-3 ’ , Actin reverse primer 5’- ATGGCTACGTAC ATGGCTGG-3 ’ , N sgRNA forward primer: 5’- CTCTTGTAGATCTGTTCTCTAAACGAAC-3’, N sgRNA reverse primer: 5’- GGTCCACCAAACGTAATGCG-3’ Relative sgRNA
  • SARS-related coronavirus 2 SARS-CoV-2
  • Isolate USA-WA1/ 2020 NR-52281
  • SARS-CoV-2 was propagated in Vero E6 cells in DMEM supplemented with 2% FBS, 4.5 g/L D-glucose, 4 mM L-glutamine, 10 mM Non-Essential Amino Acids, 1 mM Sodium Pyruvate and 10 mM HEPES and filtered through an Amicon Ultracel 15 (100kDa) centrifugal filter. Flow through was discarded and virus resuspended in DMEM supplemented as above.
  • Infectious titers of SARS-CoV-2 stock and hamster lung homogenates were determined by plaque assay in Vero E6 cells in Minimum Essential Media supplemented with 2% FBS, 4 mM L-glutamine, 0.2% BSA, 10 mM HEPES and 0.12% NaHCO 3 and 0.7% agar.
  • the efficacy of the agents described herein may be farther increased by increasing drug levels, either via intravenous administration, through delivery of drug directly to the lung airway (e.g., via aerosol, nebulizer, or tracheal wash), or both.
  • the efficacy may increase further when used against native SARS-CoV2, as inhibition of viral entry will effectively block the propagation of many progeny virus, and thus the untreated virus will increase many more fold in number in controls whereas the drug treated should show a similar total low number of progeny.
  • inhibitors of viral entry should synergize with inhibitors of viral infection and response to infection that work via distinct mechanisms, such as inhibitors of viral budding, inhibitors of proteases required for activation of entry ' (e.g,, nafarnostat, casmoslat, and aprotinin), inhibitors of viral replication, and inhibitors of membrane fusion, as well as modulators of the host inflammatory response.
  • inhibitors of viral budding inhibitors of proteases required for activation of entry ' (e.g, nafarnostat, casmoslat, and aprotinin)
  • inhibitors of viral replication e.g, nafarnostat, casmoslat, and aprotinin
  • inhibitors of membrane fusion e.g, as well as modulators of the host inflammatory response.

Abstract

The present disclosure provides compositions and methods for preventing and/or treating infection by coronaviruses, including SARS-CoV2 infection.

Description

COMPOSITIONS AND METHODS FOR TREATING CORONAVIRUS RELATED APPLICATIONS This application claims the benefit under 35 U.S.C. § 119(e) of U.S. provisional application number 62/987,024, filed March 9, 2020, U.S. provisional application number 62/993,816, filed March 24, 2020, U.S. provisional application number 63/000,772, filed March 27, 2020, U.S. provisional application number 63/028,312, filed May 21, 2020, U.S. provisional application number 63/051,601, filed July 14, 2020 and U.S. provisional application number 63/069,583, filed August 24, 2020, each of which is incorporated by reference herein in its entirety. BACKGROUND In December 2019, a novel pneumonia caused by a previously unknown pathogen emerged in Wuhan, a city of 11 million people in central China, and is rapidly spreading to many counties. The pathogen was soon identified as a novel coronavirus that is closely related to Severe Acute Respiratory Syndrome CoV (SARS-CoV), and thus is referred to as SARS-CoV2 (or COVID-19). Currently, there is no specific treatment against this new virus. Therefore, identifying effective antiviral agents to combat the disease is urgently needed, and especially existing FDA-approved drugs that could be rapidly purposed for clinical testing in the field. SUMMARY In the studies provided herein, several agents were evaluated for efficacy against SARS- CoV2, specifically, their ability to inhibit entry of a SARS-CoV2 Spike protein pseudotyped virus into human Huh-7 liver cells, which has been shown to mimic the response of native SARS-CoV2 virus in other cultured cells, and in more clinically-relevant human lung airway epithelial cells grown within human organ-on-a-chip microfluidic models of the human lung that have been shown to recapitulate human lung physiology and pathophysiology1-5. Among the agents tested, (Z)-toremifene (e.g., FARESTON®), clomiphene (e.g., CLOMID®), amodiaquine (e.g., CAMOQUIN®), verapamil (e.g., CALAN®), amiodarone (e.g., NEXTERONE®), chloroquine (e.g., ARALEN®), and umifenovir (e.g., ARBIDOL®), are FDA-approved for other indications. Baicalein is a natural product isolated from Chinese herbs, such as roots of Scutellaria baicalensis and Scutellaria lateriflora. The synthetic HR2 peptide is derived from the HR2 domain of the spike protein of SARS-CoV2. The data provided herein shows that the agents tested decrease viral infectivity and increase coronavirus inhibition rate (rate of inhibition of coronavirus infection), relative to a control. Some aspects of the present disclosure provide methods for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of an agent (or a pharmaceutically acceptable salt of an agent) selected from the group consisting of (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide.
Other aspects of the present disclosure provide methods for preventing coronavirus infection, comprising administering to a subject at risk of coronavirus a composition comprising a therapeutically effective amount of an agent (or a pharmaceutically acceptable salt of an agent) selected from the group consisting of (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide.
In some embodiments, the coronavirus is a beta coronavirus. For example, the beta coronavirus may be SARS-CoV2.
In some embodiments, the composition comprises a combination of agents (e.g., two, three, four, five, six, seven, eight, or nine agents) selected from the group consisting of (Z)- toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide. In some embodiments, the composition comprises (Z)-toremifene. In some embodiments, the composition comprises clomiphene. In some embodiments, the composition comprises amodiaquine. In some embodiments, the composition comprises verapamil. In some embodiments, the composition comprises umifenovir. In some embodiments, the composition comprises amiodarone. In some embodiments, the composition comprises chloroquine. In some embodiments, the composition comprises baicalein. In some embodiments, the composition comprises CoV HR2 peptide or a variant CoV HR2 peptide.
In some embodiments, the composition comprises a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1. In some embodiments, the composition comprises a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1. In some embodiments, the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
In some embodiments, the therapeutically effective amount is an amount effective for decreasing coronavirus infectivity, relative to a control. For example, the therapeutically effective amount may be an amount effective for decreasing viral infectivity by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control. In some embodiments, the therapeutically effective amount is an amount effective for increasing rate of inhibition of coronavirus infection, relative to a control. For example, the therapeutically effective amount may be an amount effective for increasing rate of inhibition of coronavirus infection by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control. A control may be, for example, untreated subjects.
In some embodiments, the composition further comprises a pharmaceutically-acceptable excipient.
In some embodiments, the subject is a human subject. In some embodiments, the subject is immunocompromised. In some embodiments, the subject is a child or an elderly person.
In some embodiments, the composition is administered nasally, orally, intravenously, intramuscularly, subcutaneously, or intraperitoneally. In some embodiments, the composition is administered via inhalation.
In some embodiments, the composition is administered to the lung airway of the subject.
In some embodiments, the composition is administered to the lung airway via an aerosol, a nebulizer, or a tracheal wash.
In some embodiments, the composition is administered as a single dose.
In other embodiments, the composition is administered as multiple doses. For example, the composition may be administered at multiple time points. In some embodiments, the composition is administered the composition weekly, monthly, every six months, yearly, every 5 years, or every 10 years.
Still other aspects of the present disclosure provide a composition comprising a therapeutically effective amount of a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
Further aspects of the present disclosure provide a composition comprising a therapeutically effective amount of a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
In some embodiments, a composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
In some embodiments, a composition further comprises a pharmaceutically-acceptable excipient.
Yet other aspects of the present disclosure provide a method for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of a SARS-CoV2 HR2 peptide that comprises an amino acid sequence having at least 95% identity (or 100% identity) to the amino acid sequence of SEQ ID NO: 1, wherein the composition is administered to a lung airway of the subject. In some embodiments, the composition is administered via an aerosol, a nebulizer, or a tracheal wash.
Further aspects of the present disclosure provide a method for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of amodiaquine, wherein viral load in the subject is reduced by at least 40%, at least 50%, at least 60%, or at least 70% relative to a control ( e.g ., as measured by RT-qPCR of the viral N transcript when measured, for example, at least 3 days after viral challenge). In some embodiments, the coronavirus is a beta coronavirus, such as SARS-CoV2.
BRIEF DESCRIPTION OF THE DRAWINGS
FIG. 1. The model of pseudotyped SARS-CoV2 (CoVpp) entry. The genome of CoVpp is from the HIV genome but carries a luciferase reporter gene; while its envelope protein is the spike protein of coronavirus. This pseudotyped virus can mimic the entry of coronavirus into host cells. Upon entry into host cells, we can detect report gene expression signal. If one drug can block viral entry, no signal can be detected. Using this model, we can test coronavirus entry inhibitors.
FIG. 2. The infection of CoVpp in Huh-7 cells. Huh-7 (5,000 cells per well) was seeded into 96-well plate, cultured for 24 h, and infected with CoVpp in DMEM supplemented with 3% FBS. 72 h post-infection, luminescence was measured using the Bright-Glo reagent (Prornega) according to the manufacturer's protocol. The particles without the spike protein of SARS-CoV2 was used as control.
FIG. 3. Repurposing of FDA-approved drugs as entry inhibitors of SARS-CoV2. Huh-7 cells (5,000 cell per well in 96-well plate) were infected with CoVpp in the presence or absence of test drugs at 1 uM or 5uM concentration. 72 hours post-infection, Luciferase activity, which correlates with the number of pseudoparticles in the host cells, was measured 3 days after infection using the Bright-Gio reagent (Prornega) according to the manufacturer's protocol. Vesicular stomatitis vims envelope protein GP-carrying pseudotyped viral particle (VSVpp) was used to test the specificity and cytotoxicity of drugs. Virus infectivity the presence and absence of drugs were calculated as above, *, P < 0.05; P < 0.01; ***, P < 0.001.
FIG. 4. Identification of SARS-CoV2 HR2 peptide and baicalein as entry inhibitors of SARS-CoV2. Huh-7 cells (5,000 cell per well in 96-well plate) were infected with CoVpp in the presence or absence of test drugs. The sequence of SARS-CoV2 HR2 peptide is GDIS GINAS V VNIQKEIDRFNE V AKNFNES FIDFQEFG (SEQ ID NO: 1). 72 hours post- infection, Luciferase activity, which correlates with the number of pseudoparticles in the host cells, was measured 3 days after infection using the Bright-Glo reagent (Promega) according to the manufacturer's protocol. Vesicular stomatitis virus envelope protein GP-carrying pseudotyped viral particle (VSVpp) was used to test the specificity and cytotoxicity of drugs. Virus infectivity the presence and absence of drugs were calculated as above. *, P < 0.05; **, P < 0.01: ***, P < 0,001.
FIGs. 5A-5B. The expression of SARS-CoV2 receptor ACE2 and CoVpp infection in human Airway chip. (FIG. 5A) Micrographs showing ZO-1, ACE2, Foxjl in the airway chip. (FIG. 5B) Graph showing the infection of CoVpp in the airway chip. CoVpp was delivered into airway channel. 72 h later, cells were collected for detection of viral pol gene by qRT-PCR.
FIGs. 6A-6B. The efficacy of nafamostat mesylate, ehloroquine diphosphate, arbidol hydrochloride, toremifene, ciomipbene citrate, amodiaqnine, verapamil hydrochloride, amiodarone. HR2 protein, and imatinib in human Airway Chips infected with CoV-2pp. These compounds were delivered into apical and basal channels of the chip at their respective Cmax in human blood (except for nafamostat), and one day later chips were infected with CoV-2pp while in the continued presence of the drugs for 2 more days. The epithelium from the chips were collected for detection of viral pol gene by qRT-PCR; viral entry in untreated chips was set as 100%. A t-test between the control and each compound (9 compounds + 1 protein — > 10 comparisons total) was performed, showing statistically significant viral inhibition in human Airway Chips following treatment with amodiaquine and toremifene (FIG. 6A). Multiple hypothesis testing correction on these 10 comparisons were then performed to obtain the p- values for each compound. P < 0.05; **, P < 0.01; ***, P < 0.001; 2-6 replicate Airway Chips for each compound tested. Further assessment with another statistical analysis tool, the Bonferroni correction for multiple hypothesis testing, showed statistically significant viral inhibition in human Airway Chips following treatment with amodiaquine, ciomipbene, and toremifene (FIG. 6B).
FIGs. 7A-7B. Evaluation of the cytotoxicity of test drugs in Huh-7 cells and human Airway chips. (FIG. 7.4) Huh-7 cells were treated with the test drugs at 1 or 5 mM for 48 h, and cell viability was measured by Celititer-Gio assay. The cell viability of untreated cells was set as 100%. (FIG. 7B) Human Airway Chips were treated with the test drugs at their respective Cmax for 72 h, cell damage was measure by LDH assay. The LDH level of untreated human Airway Chips was set as 100%. Note that none of the drags produced any significant cytotoxicity at the doses used in these studies.
FIGs. 8A-8I. Inhibition of infection by native SARS-CoV-2 virus in vitro and in vivo. (FIG. 8A) Dose-response curves for amodiaquine and its metabolite desethylamodiaquine demonstrating their ability to inhibit GFP-SARS-CoV-2 infection (MOI = 0.1) in a dose- dependent manner in Vero E6 cells. (FIG. 8B) Inhibition of wild type SARS-CoV-2 infection in ACE2-expressing A549 cells by 10 mM amodiaquine. (FIG. 8C) Reduction of viral load in the lungs of hamsters treated once a day with amodiaquine (50 mg/kg) beginning 1 day prior to intranasal administration of SARS-CoV-2 virus (103 PFU) as measured by qPCR for subgenomic RNA encoding SARS-CoV-2 N protein. **, p< 0.01. (FIG. 8D) Hematoxylin- and SARS-CoV-2 N-stained histological sections of lungs from animals that were mock treated, infected with SARS-CoV-2 and treated with vehicle alone, or infected with SARS-CoV-2 and treated with amodiaquine (50 mg/kg subcutaneously). (FIG. 8E) Reduction of viral load in the lungs of hamsters treated once a day for 4 days with amodiaquine (50 mg/kg) beginning 1 day prior to co-caging with SARS-CoV-2 infected animals as measured by qPCR for RNA encoding SARS-CoV-2 N protein. *, p< 0.05. (FIG. 8F) Graph depicting plaque forming units (PFU) per mL of lung homogenate from hamsters pretreated with vehicle or amodiaquine one day prior to being exposed to infected animals. Each cohort was comprised of 4 animals; p-value = 0.037. (FIG. 8G) Reduction of viral load in the lungs of hamsters treated once a day with oral amodiaquine (75 mg/kg) versus oral hydroxychloroquine (50 mg/kg) beginning 1 day prior to intranasal administration of SARS-CoV-2 virus (103 PFU) as measured by qPCR for subgenomic RNA encoding SARS-CoV-2 N protein. **, p< 0.01. (FIG. 8H) Reduction of viral load measured on day 3 (left) and 7 (right) in the lungs of hamsters treated daily with oral amodiaquine (75 mg/kg) beginning 1 day after intranasal administration of SARS-CoV-2 virus (103 PFU) as measured by qPCR for subgenomic RNA encoding SARS-CoV-2 N protein. **, p< 0.01. (FIG. 81) Amodiaquine and its active metabolite inhibit SARS-CoV-2 infection in human ACE2-expressing A549 lung epithelial cells.
FIG. 9. Effects of desethylamodiaquine on pseudotyped SARS-CoV-2 viral entry in human Airway Chips. Desethylamodiaquine was delivered into apical and basal channels of the chip at its Cmax in human blood (1 mM), and one day later chips were infected with SARS-CoV- 2pp while in the continued presence of the drugs for 2 days. The epithelium from the chips were collected for detection of viral gene by qRT-PCR; viral entry in untreated chips was set as 100%.
FIG. 10. PK profiles for amodiaquine and desethylamodiaquine administered subcutaneously in hamsters. Plasma concentration-time profiles showing mean concentration (± s.d.) of amodiaquine (left) and desethylamodiaquine (right) at different time points after a single subcutaneous injection of amodiaquine (50 mg/kg).
FIG. 11. PK profiles for amodiaquine and desethylamodiaquine administered orally in hamsters. Plasma concentration-time profiles showing mean concentration (± s.d.) of amodiaquine (left) and desethylamodiaquine (right) at different time points after a administration of a single dose of amodiaquine (75 mg/kg) via oral gavage.
FIGs. 12A-12B. Effects of Amodiaquine treatment on gene expression in hamsters infected with SARS-CoV-2. (FIG. 12A) Volcano plot showing distribution of expressed genes in hamster lungs infected with native SARS-CoV-2 and treated with amodiaquine compared to vehicle. (FIG. 12B) Reduction of viral load measured in the lungs of hamsters in the study in which RNA seq data were obtained that are shown in (FIG. 12A). Animals were treated once a day with oral amodiaquine (75 mg/kg) beginning 1 day prior to intranasal administration of SARS-CoV-2 vims (102 PFU) as measured by qPCR for subgenomic RNA encoding SARS- CoV-2 N protein. **, p< 0.01.
DETAILED DESCRIPTION
Provided herein, in some aspects, are compositions and methods for treating and/or preventing coronavirus infection, such as SARS-CoV2 infection. The compositions used herein include at least one agent selected from (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide (e.g., SEQ ID NO: 1).
Coronavirus
Coronavimses (CoV) are a large family of zoonotic viruses that are transmitted between animals and people, causing illness ranging from the common cold to more severe diseases such as Middle East Respiratory Syndrome (MERS-CoV) and Severe Acute Respiratory Syndrome (SARS-CoV). Other non-limiting examples of coronaviruses include coronavirus 229E and NL63, which are common human alpha coronaviruses, and OC43 and HKU1, which are common human beta coronaviruses. In some embodiments, the methods and composition provided herein are used to treat an alpha coronavirus. In some embodiments, the methods and composition provided herein are used to treat a beta coronavirus. Several known coronaviruses are circulating in animals that have not yet infected humans.
Common signs of coronavirus infection include respiratory symptoms, fever, cough, shortness of breath, and breathing difficulties. In more severe cases, infection can cause pneumonia, severe acute respiratory syndrome, kidney failure, and even death.
On February 11, 2020 the World Health Organization (WHO) announced an official name for the disease that is causing the 2019 novel coronavirus outbreak, first identified in Wuhan City, Hubei Province, China - “coronavirus disease 2019”, abbreviated as “COVID-19.” In COVID-19, ‘CO’ stands for ‘corona,’ ‘VI’ for ‘vims,’ and ‘D’ for disease. Formerly, this disease was referred to as “2019 novel coronavirus” or “2019-nCoV.” In some embodiments, the coronavirus infection being treated or prevented is COVID-19, also referred to as SARS-CoV2.
Compositions
The compositions provided herein include at least one of the following agents selected from the group consisting of: (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide. Pharmaceutically acceptable salts of (Z)- (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, and baicalein are also encompassed by the present disclosure.
(Z)-Toremifene (IUPAC Name: 2-[4-[(Z)-4-chloro-1,2-diphenylbut-l-enyl]phenoxy]- N,N-dimethylethanamine) is a nonsteroidal triphenylethylene antiestrogen (see, e.g., Abeam, abl42467; CAS Number 89778-26-7). Chemically related to tamoxifen, toremifene is a selective estrogen receptor modulator (SERM). This agent binds competitively to estrogen receptors, thereby interfering with estrogen activity. Toremifene also has intrinsic estrogenic properties, which are manifested according to tissue type or species. See U.S. National Library of Medicine, PubChem entry for “Toremifene.” Toremifene has approved by the FDA for the treatment of metastatic breast cancer in postmenopausal women having estrogen receptor-positive or unknown tumors (FDA drug sheet, “FARESTON®”; reference ID: 4097849, 2017).
(Z)-Toremifene chemical structure:
Figure imgf000010_0001
In some embodiments, a therapeutically effective amount of (Z)-toremifene (e.g., FARESTON®) is administered. (Z)-toremifene, in some embodiments, may be administered orally as a 60 mg dose daily (Mari, The APRN and PA's Complete Guide to Prescribing Drug Therapy 2020; page 60). The therapeutically effective amount of (Z)-toremifene can be a dose of 30 mg - 120 mg, 30 mg - 110 mg, 30 mg - 100 mg, 30 mg - 90 mg, 30 mg - 80 mg, 30 mg - 70 mg, 30 mg - 60 mg, 30 mg - 50 mg, 30 mg - 40 mg, 40 mg -120 mg, 40 mg - 110 mg, 40 mg - 100 mg, 40 mg - 90 mg, 40 mg - 80 mg, 40 mg - 70 mg, 40 mg - 60 mg, 40 mg - 50 mg, 50 mg
- 120 mg, 50 mg - 110 mg, 50 mg - 100 mg, 50 mg - 90 mg, 50 mg - 80 mg, 50 mg - 70 mg,
50 mg - 60 mg, 60 mg - 120 mg, 60 mg - 110 mg, 60 mg - 100 mg, 60 mg - 90 mg, 60 mg - 80 mg, 60 mg - 70 mg, 70 mg - 120 mg, 70 mg - 110 mg, 70 mg - 100 mg, 70 mg - 90 mg, 70 mg
- 80 mg, 80 mg - 120 mg, 80 mg - 110 mg, 80 mg - 100 mg, 80 mg - 90 mg, 90 mg - 120 mg, 90 mg - 110 mg, 90 mg - 100 mg, or 100 mg - 120 mg. In some embodiments, the therapeutically effective amount of (Z)-toremifene is 30 mg, 60 mg, 90 mg, 120 mg, 150 mg,
180 mg, or 210 mg. In some embodiments, the therapeutically effective amount of (Z)- toremifene is 60 mg. In some embodiments, the (Z)-toremifene is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of (Z)-toremifene administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
Amodiaquine (IUPAC Name: 4-[(7-chloroquinolin-4-yl)amino]-2- (diethylaminomethyl)phenol), e.g., amodiaquin dihydrochloride dihydrate (see, e.g., MilliporeSigma, Cat. A2799; CAS Number 398-98-7), is an orally active 4-aminoquinoline derivative with antimalarial and anti-inflammatory properties. Similar in structure and activity to chloroquine, amodiaquine is effective against some chloroquine-resistant strains, particularly Plasmodium falciparum, the most deadly malaria parasite. Although the mechanism of plasmodicidal action has not been fully elucidated, like other quinoline derivatives, amodiaquine likely is able to inhibit heme polymerase activity in the body. This results in accumulation of free heme, which is toxic to the parasites. See U.S. National Library of Medicine, PubChem entry for “Amodiaquine.” Amodiaquine is an FDA-approved 4-aminoquinoline antimalarial drug, which is known to inhibit rRNA transcription, a rate-limiting step in the ribosome biosynthesis pathway (Espinoza et al, Cell Death & Diff. 2020, 27:773-789). In some embodiments, a therapeutically effective amount of amodiaquin dihydrochloride dihydrate is administered.
Amodiaquin Dihydrochloride Dihydrate Chemical Structure: C20H22CIN3O · 2HC1 ·
2H2O.
In some embodiments, a therapeutically effective amount of amodiaquine (e.g., AMDAQUINE®, AMOBIN®) is administered. In some embodiments, a therapeutically effective amount of amodiaquin dihydrochloride dihydrate is administered. Amodiaquine, in some embodiments, may be administered orally as a 100 mg or 200 mg dose (Cairns et al, Antimicrobial Agents and Chemother. 2010, 54(3): 1265-74). The therapeutically effective amount of amodiaquine can be a dose of 50 mg - 300 mg, 50 mg - 275 mg, 50 mg - 250 mg, 50 mg - 225 mg, 50 mg - 200 mg, 50 mg - 175 mg, 50 mg - 150 mg, 50 mg - 125 mg, 50 mg - 100 mg, 50 mg -75 mg, 75 mg - 300 mg, 75 mg - 275 mg, 75 mg - 250 mg, 75 mg - 225 mg, 75 mg - 200 mg, 75 mg - 175 mg, 75 mg - 150 mg, 75 mg - 125 mg, 75 mg - 100 mg, 100 mg -
300 mg, 100 mg - 275 mg, 100 mg - 250 mg, 100 mg - 225 mg, 100 mg - 200 mg, 100 mg -
175 mg, 100 mg - 150 mg, 100 mg - 125 mg, 125 mg - 300 mg, 125 mg - 275 mg, 125 mg -
250 mg, 125 mg - 225 mg, 125 mg - 200 mg, 125 mg - 175 mg, 125 mg - 150 mg, 150 mg - 300 mg, 150 mg - 275 mg, 150 mg - 250 mg, 150 mg - 225 mg, 150 mg - 200 mg, 150 mg - 175 mg, 175 mg - 300 mg, 175 mg - 275 mg, 175 mg - 250 mg, 175 mg - 225 mg, 175 mg - 200 mg, 200 mg - 300 mg, 200 mg - 275 mg, 200 mg - 250 mg, 200 mg - 225 mg, 200 mg - 350 mg, 200 mg - 400 mg, 200 mg - 450 mg, 200 mg - 500 mg, 225 mg - 300 mg, 225 mg - 275 mg, 225 mg - 250 mg, 250 mg - 300 mg, 250 mg - 275 mg, or 275 mg - 300 mg. In some embodiments, the therapeutically effective amount of amodiaquine is 50 mg, 100 mg, 150 mg, 200 mg, 250 mg, 300 mg, or 400 mg. In some embodiments, the therapeutically effective amount of amodiaquine is 200 mg. In some embodiments, the amodiaquine is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of amodiaquine administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
Clomiphene (IUPAC Name: 2-[4-(2-chloro-1,2-diphenylethenyl)phenoxy]-N,N- diethylethanamine) is a triphenyl ethylene stilbene derivative which is an estrogen agonist or antagonist depending on the target tissue. Clomiphene has both estrogenic and anti-estrogenic properties, but its precise mechanism of action has not been determined. Clomiphene appears to stimulate the release of gonadotropins, follicle- stimulating hormone (FSH), and luteinizing hormone (LH), which leads to the development and maturation of ovarian follicle, ovulation, and subsequent development and function of the corpus luteum, thus resulting in pregnancy. Gonadotropin release may result from direct stimulation of the hypothalamic-pituitary axis or from a decreased inhibitory influence of estrogens on the hypothalamic-pituitary axis by competing with the endogenous estrogens of the uterus, pituitary, or hypothalamus. Clomiphene has no apparent progestational, androgenic, or antiandrogenic effects and does not appear to interfere with pituitary- adrenal or pituitary-thyroid function. See U.S. National Library of Medicine, PubChem entry for “Clomiphene.”
In some embodiments, a therapeutically effective amount of clomiphene, also known as clomifene (e.g., BECLOM®, BEMOT®, BLESIFEN®, CHLORAMIPHENE®, CLOFERT®, CLOMENE®, CLOMHEXAL®, CLOM®, CLOMID®, CLOMIDAC®, CLOMIFEN®, CLOMIFENCITRAT®, CLOMIFENE®, CLOMIFENE®, CLOMIFENE CITRATE®, CLOMIFENI CITRAS®, CLOMIFENO®, CLOMIFERT®, CLOMIHEXAL®, CLOMIPHEN, CLOMIPHENE®, CLOMIPHENE CITRATE®, CLONINN®, CLOSTIL®, CLOSTILBEGYT®, CLOVERTIL®, CLOVUL®, DIPTHEN®, DUFINE®, DUINUM®, FENSIPROS®, FERTAB®, FERTEC®, FERTEX®, FERTICLO®, FERTIL®, FERTILAN®, FERTILPHEN®, FERTIN®, FERTOMID®, FERTON®, FERTOTAB®, FERTYL®, FETROP®, FOLISTIM®, GENOCLOM®, GENOZYM®, HETE®, I-CLOM®, IKACLOMIN®, KLOFIT®, KLOMEN®, KLOMIFEN®, LOMIFEN®, MER 41®, MILOPHENE®, OFERTIL®, OMIFIN®, OVA-MGG®, OVAMIT®, OVINUM®, OVIPREG®, OVOFAR®, OVUCLON®, OVULET®, PERGOTIME®, PINFETIL®, PROFERTIL®, PROLIFEN®, PROVULA®, REOMEN®, SEROFENE®, SEROPHENE®, SERPAFAR®, SERPAFAR®, SUROLE®, TOCOFENO®, ZIMAQUIN®), is administered. In some embodiments, a therapeutically effective amount of clomiphene citrate is administered. Clomiphene has been approved by the FDA for treatment of ovulatory dysfunction in women who want to become pregnant (FDA drug sheet, “CLOMID®”; reference ID: 3206435, 2012). Clomiphene, in some embodiments, may be administered at a dose of 50 mg per day for five days (Mayo Clinic, “Clomiphene,” 2020). The therapeutically effective dose of clomiphene can be a dose of 10 mg - 150 mg, 10 mg - 125 mg, 10 mg - 100 mg, 10 mg - 100 mg, 10 mg - 90 mg, 10 mg - 80 mg, 10 mg - 75 mg, 10 mg - 70 mg, 10 mg - 60 mg, 10 mg - 50 mg, 10 mg - 40 mg, 10 mg - 30 mg, 10 mg - 20 mg, 25 mg - 150 mg, 25 mg - 125 mg, 25 mg - 100 mg, 25 mg - 90 mg, 25 mg - 80 mg, 25 mg - 75 mg, 25 mg - 70 mg, 25 mg - 60 mg, 25 mg - 50 mg,
25 mg - 40 mg, 25 mg - 30 mg, 30 mg - 150 mg, 30 mg - 125 mg, 30 mg - 100 mg, 30 mg - 90 mg, 30 mg - 80 mg, 30 mg -75 mg, 30 mg - 70 mg, 30 mg - 60 mg, 30 mg - 50 mg, 30 mg - 40 mg, 40 mg - 150 mg, 40 mg - 125 mg, 0 mg - 100 mg, 40 mg - 90 mg, 40 mg - 80 mg, 40 mg - 75 mg, 40 mg - 70 mg, 40 mg - 60 mg, 40 mg - 50 mg, 50 mg - 150 mg, 50 mg - 125 mg, 50 mg - 100 mg, 50 mg - 90 mg, 50 mg - 80 mg, 50 mg -75 mg, 50 mg - 70 mg, 50 mg - 60 mg,
60 mg - 150 mg, 60 mg - 125 mg, 60 mg - 100 mg, 60 mg - 90 mg, 60 mg - 80 mg, 60 mg -75 mg, 60 mg - 70 mg, 75 mg - 150 mg, 75 mg - 125 mg, 75 mg - 100 mg, 75 mg - 90 mg, 75 mg - 80 mg, 80 mg - 150 mg, 80 mg - 125 mg, 80 mg - 100 mg, 80 mg - 90 mg, 90 mg - 150 mg, 90 mg - 125 mg, 90 mg - 100 mg, 100 mg - 150 mg, 100 mg - 125 mg, or 125 mg - 150 mg. In some embodiments, the therapeutically effective dose of clomiphene is 25 mg, 50 mg, 75 mg,
100 mg, 125 mg, or 150 mg. In some embodiments, the therapeutically effective amount of clomiphene is 50 mg. In some embodiments, the clomiphene is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of clomiphene administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
Verapamil (IUPAC Name: 2-(3,4-dimethoxyphenyl)-5-[2-(3,4-dimethoxyphenyl)ethyl- methylamino]-2-propan-2-ylpentanenitrile) is a phenylalkylamine calcium channel blocking agent. Verapamil inhibits the transmembrane influx of extracellular calcium ions into myocardial and vascular smooth muscle cells, causing dilatation of the main coronary and systemic arteries and decreasing myocardial contractility. This agent also inhibits the drug efflux pump P- glycoprotein which is overexpressed in some multi-drug resistant tumors and may improve the efficacy of some antineoplastic agents. See U.S. National Library of Medicine, PubChem entry for “Verapamil.”
In some embodiments, a therapeutically effective amount of verapamil (e.g., CALAN®, VERELAN®) is administered. In some embodiments, a therapeutically effective amount of verapamil hydrochloride is administered. Verapamil (benzeneacetonitrile, a-[3-[[2-(3,4- dimethoxyphenyl)ethyl] methylamino]propyl]-3,4-dimethoxya-(l-methylethyl) hydrochloride) is an FDA-approved calcium antagonist or slow-channel inhibitor for the treatment of supraventricular tachyarrhythmias (FDA drug sheet, “Verapamil”; reference ID: 4015901, 2016). Verapamil, in some embodiments, may be administered intravenously, for example as a 5 mg or
10 mg bolus (id.). The therapeutically effective amount of verapamil can be a dose of 1 mg - 30 mg, 1 mg - 25 mg, 1 mg - 20 mg, 1 mg - 18 mg, 1 mg - 15 mg, 1 mg - 14 mg, 1 mg - 13 mg, 1 mg - 12 mg, 1 mg - 11 mg, 1 mg -10 mg, 1 mg - 9 mg, 1 mg - 8 mg, 1 mg - 7 mg, 1 mg - 6 mg,
I mg - 5 mg, 1 mg - 4 mg, 1 mg - 3 mg, 1 mg - 2 mg, 1 mg - 30 mg, 1 mg - 25 mg, 1 mg - 20 mg, 1 mg - 18 mg, 1 mg - 15 mg, 1 mg - 14 mg, 1 mg - 13 mg, 1 mg - 12 mg, 1 mg - 11 mg, 1 mg -10 mg, 1 mg - 9 mg, 1 mg - 8 mg, 1 mg - 7 mg, 1 mg - 6 mg, 1 mg - 5 mg, 1 mg - 4 mg, 1 mg - 3 mg, 1 mg - 2 mg, 3 mg - 30 mg, 3 mg - 25 mg, 3 mg - 20 mg, 3 mg - 18 mg, 3 mg - 15 mg, 3 mg - 14 mg, 3 mg - 13 mg, 3 mg - 12 mg, 3 mg - 11 mg, 3 mg -10 mg, 3 mg - 9 mg, 3 mg - 8 mg, 3 mg - 7 mg, 3 mg - 6 mg, 3 mg - 5 mg, 3 mg - 4 mg, 5 mg - 30 mg, 5 mg - 25 mg, 5 mg - 20 mg, 5 mg - 18 mg, 5 mg - 15 mg, 5 mg - 14 mg, 5 mg - 13 mg, 5 mg - 12 mg, 5 mg - 11 mg, 5 mg - 10 mg, 5 mg - 9 mg, 5 mg - 8 mg, 5 mg - 7 mg, 5 mg - 6 mg, 6 mg - 30 mg, 6 mg - 25 mg, 6 mg - 20 mg, 6 mg - 18 mg, 6 mg - 15 mg, 6 mg - 14 mg, 6 mg - 13 mg, 6 mg - 12 mg, 6 mg - 11 mg, 6 mg - 10 mg, 6 mg - 9 mg, 6 mg - 8 mg, 7 mg - 30 mg, 7 mg - 25 mg, 7 mg - 20 mg, 7 mg - 18 mg, 7 mg - 15 mg, 7 mg - 14 mg, 7 mg - 13 mg, 7 mg - 12 mg, 7 mg -
11 mg, 7 mg -10 mg, 7 mg - 9 mg, 7 mg - 8 mg, 8 mg - 30 mg, 8 mg - 25 mg, 8 mg - 20 mg, 8 mg - 18 mg, 8 mg - 15 mg, 8 mg - 14 mg, 8 mg - 13 mg, 8 mg - 12 mg, 8 mg - 11 mg, 8 mg - 10 mg, 8 mg - 9 mg, 9 mg - 30 mg, 9 mg - 25 mg, 9 mg - 20 mg, 9 mg - 18 mg, 9 mg - 15 mg, 9 mg - 14 mg, 9 mg - 13 mg, 9 mg - 12 mg, 9 mg - 11 mg, 9 mg -10 mg, 10 mg - 30 mg, 10 mg
- 25 mg, 10 mg - 20 mg, 10 mg - 18 mg, 10 mg - 15 mg, 10 mg - 14 mg, 10 mg - 13 mg, 10 mg - 12 mg, 10 mg - 11 mg, 11 mg - 30 mg, 11 mg - 25 mg, 11 mg - 20 mg, 11 mg - 18 mg,
I I mg - 15 mg, 11 mg - 14 mg, 11 mg - 13 mg, 11 mg - 12 mg, 12 mg - 30 mg, 12 mg - 25 mg, 12 mg - 20 mg, 12 mg - 18 mg, 12 mg - 15 mg, 12 mg - 14 mg, 12 mg - 13 mg, 13 mg - 30 mg, 13 mg - 25 mg, 13 mg - 20 mg, 13 mg - 18 mg, 13 mg - 15 mg, 13 mg - 14 mg, 15 mg
- 30 mg, 15 mg - 25 mg, 15 mg - 20 mg, 15 mg - 18 mg, 20 mg - 30 mg, 20 mg - 27 mg, 20 mg - 25 mg, 20 mg - 22 mg or 25 mg - 30 mg. In some embodiments, the therapeutically effective amount of verapamil is 5 mg, 10 mg, 15 mg, 20 mg, 250 mg, 30 mg, or 40 mg. In some embodiments, the therapeutically effective amount of verapamil is 10 mg. In some embodiments, the verapamil is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of verapamil administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
Umifenovir (IUPAC Name: ethyl 6-bromo-4-[(dimethylamino)methyl]-5-hydroxy-l- methyl-2-(phenylsulfanylmethyl)indole-3-carboxylate) has been approved for use in Russia and China as a broad spectrum antiviral compound to treat influenza (Haviernik et al, Viruses, 2018, 10(4):E184).
In some embodiments, a therapeutically effective amount of umifenovir (e.g.,
ARB IDOL®) is administered. In some embodiments, a therapeutically effective amount of arbidol hydrochloride is administered. Umifenovir, in some embodiments, may be administered orally in 400 mg doses (Pshenichnaya et al, Ter Arkh. 2019, 91(3): 56-63). The therapeutically effective amount of umifenovir can be a dose of 200 mg - 1000 mg, 200 mg - 800 mg, 200 mg - 750 mg, 200 mg - 700 mg, 200 mg - 600 mg, 200 mg - 500 mg, 200 mg - 400 mg, 200 mg - 300 mg, 300 mg - 1000 mg, 300 mg - 800 mg, 300 mg - 750 mg, 300 mg - 700 mg, 300 mg - 600 mg, 300 mg - 500 mg, 300 mg - 400 mg, 400 mg - 1000 mg, 400 mg - 800 mg, 400 mg - 750 mg, 400 mg - 700 mg, 400 mg - 600 mg, 400 mg - 500 mg, 500 mg - 1000 mg, 500 mg - 800 mg, 500 mg - 750 mg, 500 mg - 700 mg, 500 mg - 600 mg, 600 mg - 1000 mg, 600 mg - 800 mg, 600 mg - 750 mg, 600 mg - 700 mg, 700 mg - 1000 mg, 700 mg - 800 mg, 800 mg - 1000 mg, 800 mg - 900 mg, 800 mg - 1200 mg, 800 mg - 1600 mg, 900 mg - 1000 mg, 900 mg - 1200 mg, 900 mg - 1600 mg, 1000 mg - 1600 mg, or 1000 mg - 1200. In some embodiments, the therapeutically effective amount of verapamil is 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, or 1600 mg. In some embodiments, the therapeutically effective amount of umifenovir is 400 mg. In some embodiments, the umifenovir is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of umifenovir administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
Amiodarone (2- { 4- [(2-butyl- 1 -benzofuran-3 -yl)carbonyl] -2,6- diiodophenoxy}ethyl)diethylamine is an FDA-approved antiarrhythmic drug having predominantly Class III (Vaughn Williams’ classification) effects for use in life-threatening recurrent ventricular arrhythmias that have not responded to adequate doses of other antiarrhythmic agents (FDA drug sheet, “CORDARONE®”; reference ID: 2876651).
In some embodiments, a therapeutically effective amount of amiodarone (e.g., NEXTERONE®, CORDARONE®) is administered. In some embodiments, a therapeutically effective amount of amiodarone hydrochloride is administered. Amiodarone, in some embodiments, may be administered orally in 800 mg doses (id.). The therapeutically effective amount of amiodarone can be a dose of 100 mg - 2000 mg, 100 mg - 1800 mg, 100 mg - 1700 mg, 100 mg - 1600 mg, 100 mg - 1500 mg, 100 mg - 1400 mg, 100 mg - 1300 mg, 100 mg - 1200 mg, 100 mg - 1100 mg, 100 mg - 1000 mg, 100 mg - 900 mg, 100 mg - 800 mg, 100 mg
- 700 mg, 100 mg - 600 mg, 100 mg - 500 mg, 100 mg - 400 mg, 100 mg - 300 mg, 100 mg - 200 mg, 200 mg - 2000 mg, 200 mg - 1800 mg, 200 mg - 1700 mg, 200 mg - 1600 mg, 200 mg
- 1500 mg, 200 mg - 1400 mg, 200 mg - 1300 mg, 200 mg - 1200 mg, 200 mg - 1100 mg, 200 mg - 1000 mg, 200 mg - 900 mg, 200 mg - 800 mg, 200 mg - 700 mg, 200 mg - 600 mg, 200 mg - 500 mg, 200 mg - 400 mg, 200 mg - 300 mg, 400 mg - 2000 mg, 400 mg - 1800 mg, 400 mg - 1700 mg, 400 mg - 1600 mg, 400 mg - 1500 mg, 400 mg - 1400 mg, 400 mg - 1300 mg, 400 mg - 1200 mg, 400 mg - 1100 mg, 400 mg - 1000 mg, 400 mg - 900 mg, 400 mg - 800 mg, 400 mg - 700 mg, 400 mg - 600 mg, 400 mg - 500 mg, 600 mg - 2000 mg, 600 mg - 1800 mg, 600 mg - 1700 mg, 600 mg - 1600 mg, 600 mg - 1500 mg, 600 mg - 1400 mg, 600 mg - 1300 mg, 600 mg - 1200 mg, 600 mg - 1100 mg, 600 mg - 1000 mg, 600 mg - 900 mg, 600 mg
- 800 mg, 600 mg - 700 mg, 800 mg - 2000 mg, 800 mg - 1800 mg, 800 mg - 1700 mg, 800 mg - 1600 mg, 800 mg - 1500 mg, 800 mg - 1400 mg, 800 mg - 1300 mg, 800 mg - 1200 mg, 800 mg - 1100 mg, 800 mg - 1000 mg, 800 mg - 900 mg, 1000 mg - 2000 mg, 1000 mg - 1800 mg, 1000 mg - 1700 mg, 1000 mg - 1600 mg, 1000 mg - 1500 mg, 1000 mg - 1400 mg, 1000 mg - 1300 mg, 1000 mg - 1200 mg, 1000 mg - 1100 mg, 1200 mg - 2000 mg, 1200 mg - 1800 mg, 1200 mg - 1700 mg, 1200 mg - 1600 mg, 1200 mg - 1500 mg, 1200 mg - 1400 mg, 1200 mg - 1300 mg, 1400 mg - 2000 mg, 1400 mg - 1800 mg, 1400 mg - 1700 mg, 1400 mg - 1600 mg, 1400 mg - 1500 mg, 1600 mg - 2000 mg, 1600 mg - 1800 mg, 1600 mg - 1700 mg, 1800 mg - 2000 mg, or 1900 mg - 2000 mg. In some embodiments, the therapeutically effective amount of amiodarone is 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1200 mg, 1600 mg, or 2000 mg. In some embodiments, the therapeutically effective amount of amiodarone is 600 mg. In some embodiments, the amiodarone is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of amiodarone administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein. Chloroquine (IUPAC Name: 4-N-(7-chloroquinolin-4-yl)-l-N,l-N-diethylpentane-1,4- diamine), e.g., chloroquine diphosphate, is a 4-aminoquinoline with antimalarial, anti- inflammatory, and potential chemosensitization and radiosensitization activities (see, e.g., Abeam, ab142116; CAS Number 50-63-5) Although the mechanism is not well understood, chloroquine is shown to inhibit the parasitic enzyme heme polymerase that converts the toxic heme into non-toxic hemazoin, thereby resulting in the accumulation of toxic heme within the parasite. This agent may also interfere with the biosynthesis of nucleic acids. Chloroquine's potential chemosensitizing and radiosensitizing activities in cancer may be related to its inhibition of autophagy, a cellular mechanism involving lysosomal degradation that minimizes the production of reactive oxygen species (ROS) related to tumor reoxygenation and tumor exposure to chemotherapeutic agents and radiation. See U.S. National Library of Medicine, PubChem entry for “Chloroquine.”
Chloroquine Diphosphate Chemical Structure:
Figure imgf000017_0001
In some embodiments, a therapeutically effective amount of chloroquine (e.g., ARALEN®) is administered. Chloroquine is an FDA-approved 4-aminoquinoline compound for the suppressive treatment and for acute attacks of malaria due to P. vivax, P. malariae, P. ovale, and susceptible strains of P. falciparum, in addition to extraintestinal amebiasis (FDA drug sheet, “ARALEN®”; reference ID: 3402523, 2013). Chloroquine, in some embodiments, may be administered orally in 300 mg doses (id.). The therapeutically effective amount of amiodarone can be a dose of 100 mg - 900 mg, 100 mg - 800 mg, 100 mg - 700 mg, 100 mg - 600 mg, 100 mg - 500 mg, 100 mg - 400 mg, 100 mg - 300 mg, 100 mg - 200 mg, 200 mg - 900 mg, 200 mg - 800 mg, 200 mg - 700 mg, 200 mg - 600 mg, 200 mg - 500 mg, 200 mg - 400 mg, 200 mg - 300 mg, 300 mg - 900 mg, 300 mg - 800 mg, 300 mg - 700 mg, 300 mg - 600 mg, 300 mg - 500 mg, 300 mg - 400 mg, 400 mg - 900 mg, 400 mg - 800 mg, 400 mg - 700 mg, 400 mg - 600 mg, 400 mg - 500 mg, 500 mg - 900 mg, 500 mg - 800 mg, 500 mg - 700 mg, 500 mg - 600 mg, 700 mg - 900 mg, 700 mg - 800 mg, or 800-900. In some embodiments, the therapeutically effective amount of chloroquine is 150 mg, 300 mg, 450 mg, 600 mg, 750 mg, 900 mg, 1050 mg, or 1200 mg. In some embodiments, the therapeutically effective amount of chloroquine is 600 mg. In some embodiments, the chloroquine is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of chloroquine administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
Baicalein (IUPAC Name: 5,6,7-trihydroxy-2-phenylchromen-4-one) is a trihydroxyflavone with the hydroxy groups at positions C-5, -6 and -7. It has a role as an antioxidant, a hormone antagonist, a prostaglandin antagonist, an EC 1.13.11.31 (arachidonate 12-lipoxygenase) inhibitor, an EC 1.13.11.33 (arachidonate 15-lipoxygenase) inhibitor, a radical scavenger, an EC 3.4.21.26 (prolyl oligopeptidase) inhibitor, an anti-inflammatory agent and a plant metabolite. It is a conjugate acid of a baicalein(1-). See U.S. National Library of Medicine, PubChem entry for “Baicalein.” Baicalein is a flavone, which was initially isolated from Scutellaria baicalensis and S. lateriflora. The compound is a positive allosteric modulator of benzodiazepine site or the non-benzodiazepine site of the GABAA receptor and is selective for a and α3 subunit-containing GABAA receptors, in addition to being an anti-inflammatory agent (Wang et al., Neuropharmacol, 2008, 55(7): 1231-7). It is also used in traditional Chinese medicine to treat inflammation, hepatitis, infections, and tumors (Liu et al., Front Pharmacol. 2019, 10:518). The compound, in some embodiments, may be administered orally, for example, as a 1000 mg dose (Li et al., J Ethnopharmacol. 2014, 154:210-5).
In some embodiments, a therapeutically effective amount of baicalein is administered. The therapeutically effective amount of baicalein can be a dose of 100 mg - 3000 mg, 100 mg - 2800 mg, 100 mg - 2600 mg, 100 mg - 2500 mg, 100 mg - 2200 mg, 100 mg - 2000 mg, 100 mg - 1800 mg, 100 mg - 1600 mg, 100 mg - 1500 mg, 100 mg - 1400 mg, 100 mg - 1200 mg, 100 mg - 1000 mg, 100 mg - 900 mg, 100 mg - 800 mg, 100 mg - 700 mg, 100 mg - 600 mg, 100 mg - 500 mg, 100 mg - 400 mg, 100 mg - 300 mg, 100 mg - 200 mg, 200 mg - 3000 mg, 200 mg - 2800 mg, 200 mg - 2600 mg, 200 mg - 2500 mg, 200 mg - 2200 mg, 200 mg - 2000 mg, 200 mg - 1800 mg, 200 mg - 1600 mg, 200 mg - 1500 mg, 200 mg - 1400 mg, 200 mg - 1200 mg, 200 mg - 1000 mg, 200 mg - 900 mg, 200 mg - 800 mg, 200 mg - 700 mg, 200 mg - 600 mg, 200 mg - 500 mg, 200 mg - 400 mg, 200 mg - 300 mg, 500 mg - 3000 mg, 500 mg - 2800 mg, 500 mg - 2600 mg, 500 mg - 2500 mg, 500 mg - 2200 mg, 500 mg - 2000 mg, 500 mg - 1800 mg, 500 mg - 1600 mg, 500 mg - 1500 mg, 500 mg - 1400 mg, 500 mg - 1200 mg, 500 mg - 1000 mg, 500 mg - 900 mg, 500 mg - 800 mg, 500 mg - 700 mg, 500 mg - 600 mg, 750 mg - 3000 mg, 750 mg - 2800 mg, 750 mg - 2600 mg, 750 mg - 2500 mg, 750 mg - 2200 mg, 750 mg - 2000 mg, 750 mg - 1800 mg, 750 mg - 1600 mg, 750 mg - 1500 mg, 750 mg - 1400 mg, 750 mg - 1200 mg, 750 mg - 1000 mg, 750 mg - 900 mg, 750 mg - 800 mg, 750 mg - 700 mg, 1000 mg - 3000 mg, 1000 mg - 2800 mg, 1000 mg - 2600 mg, 1000 mg - 2500 mg, 1000 mg - 2200 mg, 1000 mg - 2000 mg, 1000 mg - 1800 mg, 1000 mg - 1600 mg, 1000 mg -
1500 mg, 1000 mg - 1400 mg, 1000 mg - 1200 mg, 1250 mg - 3000 mg, 1250 mg - 2800 mg,
1250 mg - 2600 mg, 1250 mg - 2500 mg, 1250 mg - 2200 mg, 1250 mg - 2000 mg, 1250 mg -
1800 mg, 1250 mg - 1600 mg, 1250 mg - 1500 mg, 1250 mg - 1400 mg, 1500 mg - 3000 mg,
1500 mg - 2800 mg, 1500 mg - 2600 mg, 1500 mg - 2500 mg, 1500 mg - 2200 mg, 1500 mg -
2000 mg, 1500 mg - 1800 mg, 1500 mg - 1600 mg, 1750 mg - 3000 mg, 1750 mg - 2800 mg,
1750 mg - 2600 mg, 1750 mg - 2500 mg, 1750 mg - 2200 mg, 1750 mg - 2000 mg, 1750 mg -
1800 mg, 2000 mg - 3000 mg, 2000 mg - 2800 mg, 2000 mg - 2600 mg, 2000 mg - 2500 mg,
2000 mg - 2200 mg, 2250 mg - 3000 mg, 2250 mg - 2250 mg, 2250 mg - 2600 mg, 2250 mg -
2500 mg, 2500 mg - 3000 mg, 2500 mg - 2800 mg, or 2750 mg - 3000 mg. In some embodiments, the therapeutically effective amount of baicalein is 100 mg, 200 mg, 400 mg, 600 mg, 800 mg, 1000 mg, 1500 mg, 2000 mg, 2250 mg, 2500 mg, or 2800 mg. In some embodiments, the therapeutically effective amount of baicalein is 600 mg. In some embodiments, the baicalein is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of baicalein administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
The coronavirus spike protein is a type I membrane protein that associates into trimers on the surface of coronavirus membrane. The distal subunit (S1) of the spike protein contains the receptor binding domain, and the membrane- anchored subunit (S2) contains a putative internal fusion peptide and two heptad repeat regions (HR1 and HR2). In some embodiments, the CoV HR2 peptide comprises an amino acid sequence having at least 95% identity to GDIS GINAS V VNIQKEIDRLNE V AKNLNES LIDLQELG (SEQ ID NO: 1). In some embodiments, the CoV HR2 peptide comprises the amino acid sequence of SEQ ID NO: 1.
The term “identity” refers to a relationship between the sequences of two or more peptides, as determined by comparing the sequences. Identity also refers to the degree of sequence relatedness between or among sequences as determined by the number of matches between strings of two or more amino acid residues. Identity measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program. Identity of related peptides can be readily calculated by known methods. “Percent (%) identity” as it applies to peptide sequences is defined as the percentage of amino acid residues of a first sequence that is identical with the amino acid residues of a second sequence after aligning the sequences and introducing gaps, if necessary, to achieve the maximum percent identity. Methods and computer programs for the alignment are well known in the art. It is understood that identity depends on a calculation of percent identity but may differ in value due to gaps and penalties introduced in the calculation.
In some embodiments, a therapeutically effective amount of CoV HR2 peptide is administered. The therapeutically effective amount of CoV HR2 peptide can be a dose of 1 mg - 50 mg, lmg - 45 mg, 1 mg - 40 mg, 1 mg - 35 mg, 1 mg - 30 mg, 1 mg - 25 mg, 1 mg - 20 mg, 1 mg - 15 mg, 1 mg - 10 mg, 5 mg - 50 mg, 5 mg - 45 mg, 5 mg - 40 mg, 5 mg - 35 mg, 5 mg
- 30 mg, 5 mg - 25 mg, 5 mg - 20 mg, 5 mg - 15 mg, 5 mg - 10 mg, 10 mg - 50 mg, 10 mg - 45 mg, 10 mg - 40 mg, 10 mg - 35 mg, 10 mg - 30 mg, 10 mg - 25 mg, 10 mg - 20 mg, 10 mg
- 15 mg, 15 mg - 50 mg, 15 mg - 45 mg, 15 mg - 40 mg, 15 mg - 35 mg, 15 mg - 30 mg, 15 mg - 25 mg, 15 mg - 20 mg, 20 mg - 50 mg, 20 mg - 45 mg, 20 mg - 40 mg, 20 mg - 35 mg,
20 mg - 30 mg, 20 mg - 25 mg, 25 mg - 50 mg, 25 mg - 45 mg, 25 mg - 40 mg, 25 mg - 35 mg, 25 mg - 30 mg, 40 mg - 50 mg, 40 mg - 45 mg, or 45 mg - 50 mg. In some embodiments, the therapeutically effective amount of CoV HR2 peptide is 1 mg, 2 mg, 5 mg, 10 mg, 20 mg, or 30 mg. In some embodiments, the therapeutically effective amount of CoV HR2 peptide is 10 mg. In some embodiments, the CoV HR2 peptide is administered five times a day, four times a day, three times a day, twice a day, daily, every other day, every second day, every third day, every fourth day, every fifth day, every sixth day, or weekly. In some embodiments, a therapeutically effective amount of CoV HR2 peptide administered can be any amount that is suitable for treating or preventing coronavirus infection as described herein.
In some embodiments, an agent, or a combination of agents, is administered in an amount effective for decreasing coronavirus infectivity. In some embodiments, coronavirus infectivity is decreased by at least 20%, relative to a control. For example, coronavirus infectivity may be decreased by at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%, relative to a control. In some embodiments, coronavirus infectivity is decreased by 20%-100%, 20%-90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%- 60%, 40%-50%, 50%-100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60%, relative to a control.
In some embodiments, an agent, or a combination of agents, is administered in an amount effective for increasing the rate of inhibition of coronavirus infection. In some embodiments, the rate of inhibition of coronavirus infection is increased by at least 20%, relative to a control. For example, coronavirus infectivity may be increased by at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90%, relative to a control. In some embodiments, the rate of inhibition of coronavirus infection is increased by 20%-100%, 20%- 90%, 20%-80%, 20%-70%, 20%-60%, 20%-50%, 30%-100%, 30%-90%, 30%-80%, 30%-70%, 30%-60%, 30%-50%, 40%-100%, 40%-90%, 40%-80%, 40%-70%, 40%-60%, 40%-50%, 50%- 100%, 50%-90%, 50%-80%, 50%-70%, or 50%-60%, relative to a control.
The term “pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response, and the like, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically acceptable salts are well known in the art. For example, Berge et al. describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of the present disclosure include those derived from suitable inorganic and organic acids and bases. Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids, such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and perchloric acid or with organic acids, such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid, or malonic acid or by using other methods known in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2-hydroxy- ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like. Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium, and N+(C1-4 alky1)4- salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like. Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, lower alkyl sulfonate, and aryl sulfonate.
The compositions provided herein, in some embodiments, include at least one pharmaceutically-acceptable excipient (e.g., carrier, buffer, and/or salt, etc.). A molecule or other substance/agent is considered “pharmaceutically acceptable” if it is approved or approvable by a regulatory agency of the Federal government or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, including humans. An excipient may be any inert (inactive), non-toxic agent, administered in combination with an agent provided herein. Non-limiting examples of excipients include buffers ( e.g ., sterile saline), salts, carriers, preservatives, fillers, surfactants, and coloring agents.
Combination Therapy
The compositions provided herein may include, or may be administered in combination with, other agents, such as antiviral agents, antibacterial agents, and/or anti-inflammatory agents (e.g., nelfinavir, resdemivir, dexamethasone, and/or baricitinib). In some embodiments, a composition provided herein includes, or is administered in combination with, an agent used to treat a secondary condition associated with coronavirus infection, such as pneumonia.
In some embodiments, a method comprises administering to a subject a combination of two or more agents selected from (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, nafamostat, imatinib, and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of (Z)- toremifene and clomiphene. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and amodiaquine. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and verapamil. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and umifenovir. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and amiodarone. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and chloroquine. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and baicalein. In some embodiments, a method comprises administering to a subject a combination of (Z)-toremifene and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of clomiphene and amodiaquine. In some embodiments, a method comprises administering to a subject a combination of clomiphene and verapamil. In some embodiments, a method comprises administering to a subject a combination of clomiphene and umifenovir. In some embodiments, a method comprises administering to a subject a combination of clomiphene and amiodarone. In some embodiments, a method comprises administering to a subject a combination of clomiphene and chloroquine. In some embodiments, a method comprises administering to a subject a combination of clomiphene and baicalein. In some embodiments, a method comprises administering to a subject a combination of clomiphene and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and verapamil. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and umifenovir. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and amiodarone. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and chloroquine. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and baicalein. In some embodiments, a method comprises administering to a subject a combination of amodiaquine and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of verapamil and umifenovir. In some embodiments, a method comprises administering to a subject a combination of verapamil and amiodarone. In some embodiments, a method comprises administering to a subject a combination of verapamil and chloroquine. In some embodiments, a method comprises administering to a subject a combination of verapamil and baicalein. In some embodiments, a method comprises administering to a subject a combination of verapamil and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of umifenovir and amiodarone. In some embodiments, a method comprises administering to a subject a combination of umifenovir and chloroquine. In some embodiments, a method comprises administering to a subject a combination of umifenovir and baicalein. In some embodiments, a method comprises administering to a subject a combination of umifenovir and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of amiodarone and chloroquine. In some embodiments, a method comprises administering to a subject a combination of amiodarone and baicalein. In some embodiments, a method comprises administering to a subject a combination of amiodarone and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of chloroquine and baicalein. In some embodiments, a method comprises administering to a subject a combination of chloroquine and CoV HR2 peptide. In some embodiments, a method comprises administering to a subject a combination of baicalein and CoV HR2 peptide.
In some embodiments, a composition provided herein includes two or more agents selected from (Z)-toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide. In some embodiments, a composition comprises (Z)-toremifene and clomiphene. In some embodiments, a composition comprises (Z)-toremifene and amodiaquine. In some embodiments, a composition comprises (Z)-toremifene and verapamil. In some embodiments, a composition comprises (Z)-toremifene and umifenovir. In some embodiments, a composition comprises (Z)-toremifene and amiodarone. In some embodiments, a composition comprises (Z)-toremifene and chloroquine. In some embodiments, a composition comprises (Z)-toremifene and baicalein. In some embodiments, a composition comprises (Z)-toremifene and CoV HR2 peptide. In some embodiments, a composition comprises clomiphene and amodiaquine. In some embodiments, a composition comprises clomiphene and verapamil. In some embodiments, a composition comprises clomiphene and umifenovir. In some embodiments, a composition comprises clomiphene and amiodarone. In some embodiments, a composition comprises clomiphene and chloroquine. In some embodiments, a composition comprises clomiphene and baicalein. In some embodiments, a composition comprises clomiphene and CoV HR2 peptide. In some embodiments, a composition comprises amodiaquine and verapamil. In some embodiments, a composition comprises amodiaquine and umifenovir. In some embodiments, a composition comprises amodiaquine and amiodarone. In some embodiments, a composition comprises amodiaquine and chloroquine. In some embodiments, a composition comprises amodiaquine and baicalein. In some embodiments, a composition comprises amodiaquine and CoV HR2 peptide. In some embodiments, a composition comprises amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide. In some embodiments, a composition comprises verapamil and umifenovir. In some embodiments, a composition comprises verapamil and amiodarone. In some embodiments, a composition comprises verapamil and chloroquine. In some embodiments, a composition comprises verapamil and baicalein. In some embodiments, a composition comprises verapamil and CoV HR2 peptide. In some embodiments, a composition comprises umifenovir and amiodarone. In some embodiments, a composition comprises umifenovir and chloroquine. In some embodiments, a composition comprises umifenovir and baicalein. In some embodiments, a composition comprises umifenovir and CoV HR2 peptide. In some embodiments, a composition comprises amiodarone and chloroquine. In some embodiments, a composition comprises amiodarone and baicalein. In some embodiments, a composition comprises amiodarone and CoV HR2 peptide. In some embodiments, a composition comprises chloroquine and baicalein. In some embodiments, a composition comprises chloroquine and CoV HR2 peptide. In some embodiments, a composition comprises baicalein and CoV HR2 peptide.
Subject
The composition and methods of the present disclosure may be used to prevent or treat coronavirus (e.g., SARS-CoV2) infection in any subject, such as a human subject. In some embodiments, a subject is a child, who is between the ages of 1 year and 5 years. In some embodiments, a subject is an elderly person, who is 65 years old, or older.
In some embodiments, a subject is immunocompromised (having an impaired or weakened immune system). In some embodiments, a subject has another comorbidity, such as heart disease or liver disease. Routes of Administration and Dosing Schedule
The route of administration of the compositions provided herein may vary depending on the specific agents. In some embodiments, a composition comprising an agent is administered orally (e.g., as a liquid suspension or tablet). In some embodiments, a composition comprising an agent is administered nasally, intravenously, intramuscularly, subcutaneously, or intraperitoneally. In some embodiments, the composition is administered via inhalation. In some embodiments, a composition is administered to the lung airway, for example, via aerosol, nebulizer, or tracheal wash.
The compositions herein may be administered as a single dose or as multiple doses (e.g., a booster dose or multiple booster doses). For example, a composition may be administered weekly, monthly, every six months, yearly, every 5 years, or every 10 years. In some embodiments, a composition is administered as a therapeutic intervention after diagnosis of viral infection. In some embodiments, a composition is administered as a prophylactic treatment before diagnosis of viral infection.
Additional Embodiments
1. A method for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of an agent selected from the group consisting of (Z)-toremifene, clomiphene (e.g., clomiphene citrate), amodiaquine (e.g., amodiaquin dihydrochloride dehydrate), verapamil (e.g., verapamil hydrochloride), umifenovir (e.g., arbidol hydrochloride), amiodarone (e.g., amiodarone hydrochloride), chloroquine (e.g., chloroquine diphosphate), baicalein, and CoV HR2 peptide.
2. A method for preventing coronavirus infection, comprising administering to a subject at risk of coronavirus a composition comprising a therapeutically effective amount of an agent selected from the group consisting of (Z)-toremifene, clomiphene (e.g., clomiphene citrate), amodiaquine (e.g., amodiaquin dihydrochloride dehydrate), verapamil (e.g., verapamil hydrochloride), umifenovir (e.g., arbidol hydrochloride), amiodarone (e.g., amiodarone hydrochloride), chloroquine (e.g., chloroquine diphosphate), baicalein, and CoV HR2 peptide.
3. The method of paragraph 1 or 2, wherein the coronavirus is a beta coronavirus.
4. The method of paragraph 3, wherein the beta coronavirus is SARS-CoV2.
5. The method of any one of the preceding paragraphs, wherein the composition comprises two, three, four, five, six, seven, eight, or nine agents selected from the group consisting of (Z)- toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide. 6. The method of any one of the preceding paragraphs, wherein the composition comprises (Z)-toremifene.
7. The method of any one of the preceding paragraphs, wherein the composition comprises clomiphene.
8. The method of any one of the preceding paragraphs, wherein the composition comprises amodiaquine.
9. The method of any one of the preceding paragraphs, wherein the composition comprises verapamil.
10. The method of any one of the preceding paragraphs, wherein the composition comprises umifenovir.
11. The method of any one of the preceding paragraphs, wherein the composition comprises amiodarone.
12. The method of any one of the preceding paragraphs, wherein the composition comprises chloroquine.
13. The method of any one of the preceding paragraphs, wherein the composition comprises baicalein.
14. The method of any one of the preceding paragraphs, wherein the composition comprises a CoV HR2 peptide or a variant CoV HR2 peptide.
15. The method of paragraph 14, wherein the composition comprises a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
16. The method of paragraph 14, wherein the composition comprises a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
17. The method of paragraph 16, wherein the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
18. The method of any one of the preceding paragraphs, wherein the therapeutically effective amount is an amount effective for decreasing coronavirus infectivity.
19. The method of paragraph 18, wherein the therapeutically effective amount is an amount effective for decreasing viral infectivity by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control.
20. The method of any one of the preceding paragraphs, wherein the therapeutically effective amount is an amount effective for increasing the rate of inhibition of coronavirus infection, relative to a control. 21. The method of paragraph 20, wherein the therapeutically effective amount is an amount effective for increasing the rate of inhibition of coronavirus infection by at least 20%, at least 40%, at least 60%, or at least 80% relative to a control.
22. The method of any one of the preceding paragraphs, wherein the composition further comprises a pharmaceutically-acceptable excipient.
23. The method of any one of the preceding paragraphs, wherein the subject is a human subject.
24. The method of paragraph 23, wherein the subject is immunocompromised.
25. The method of paragraph 23 or 24, wherein the subject is a child or an elderly subject.
26. The method of any one of the preceding paragraphs, wherein the composition is administered nasally, orally, intravenously, intramuscularly, subcutaneously, or intraperitoneally.
27. The method of any one of the preceding paragraphs, wherein the composition is administered to the lung airway of the subject.
28. The method of paragraph 27, wherein the composition is administered to the lung airway via an aerosol, a nebulizer, or a tracheal wash.
29. The method of any one of the preceding paragraphs, wherein the composition is administered as a single dose.
30. The method of any one of the preceding paragraphs, wherein the composition is administered as multiple doses.
31. The method of paragraph 29, wherein the composition is administered at multiple time points.
32. The method of paragraph 31, wherein the composition is administered weekly, monthly, every six months, yearly, every 5 years, or every 10 years.
33. A composition comprising at least two agents selected from the group consisting of (Z)- toremifene, clomiphene, amodiaquine, verapamil, umifenovir, amiodarone, chloroquine, baicalein, and CoV HR2 peptide.
34. A composition comprising a therapeutically effective amount of a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
35. A composition comprising a therapeutically effective amount of a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
36. The composition of paragraph 35, wherein the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1. 37. The composition of any one of paragraphs 33-35, further comprising a pharmaceutically- acceptable excipient.
38. A method for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of a SARS-CoV2 HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1, wherein the composition is administered to a lung airway of the subject.
39. The method of paragraph 38, wherein the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
40. The method of paragraph 38 or 39, wherein the composition is administered via an aerosol, a nebulizer, or a tracheal wash.
EXAMPLES
Example 1. Generation of pseudotyped SARS-CoV2 virus and infection assay
The lifecycle of viruses includes three main steps: entry into host cells, replicate in host cells, and release from host cells. Inhibition of the first step (viral entry) should reduce virus propagation and dampen infection-related inflammatory responses. Therefore, entry inhibitors represent a new generation of antiviral drugs.6 To test potential SARS-CoV2 entry inhibitors, we generated SARS-CoV2 pseudoparticles (CoVpp) by packaging the SARS-CoV2 spike protein into a retroviral core (FIG. 1). This pseudovirus allows us to develop potential inhibitors for viral entry while alleviating biosafety concerns.
The generation of CoVpp was conducted as described previously.7 Briefly, HEK293T cells (5 x 105') were seeded into six- well plates 24 hours before transfection. Then, HEK293T cells were cotransfected with pNL4-3 plasmid, a pCMV3 plasmid that encodes the viral spike protein of SARS-CoV2, and pAdvantage plasmid using the transfection reagent MegaTran 1.0 (OriGene) according to the manufacturer's instructions. Supernatant was collected 72 hours later to obtain SARS-CoV2 spike protein pseudotyped virions (CoVpp). infections were initially performed in 96-well plates by adding diluted CoVpp to 5 x 103 Huh-7 cells (a human liver cell line) per well in the presence or absence of the test drugs or compounds. The mixtures were then incubated for 72 hours at 37°C. Luciferase activity, which reflected the number of pseudopartieles in the host cells, was measured 3 days after infection using the Bright-Glo reagent (Promega) and an FB 15 lurninometer (Berihold Detection Systems) according to the manufacturer's protocol, lest drugs were serially diluted to a final concentration of 5 μM or 1 μM in 1% DMSO. The maximum infeetivity (100%) was derived from the control wells containing DMSO control; background (0%) from uninfected wells. To calculate the inhibition values, the luciferase intensity in each of the compound testing wells were subtracted by the background signals, divided by the average signals of the control values (DMSO control), and multiplied by 100%.
Example 2. Identification of entry inhibitors of SARS-CoV2 by CoVpp entry assay
To test whether CoVpp can be used to test SARS-CoV2 entry inhibitors, we first confirmed its infection in Huh-7 cell line (FIG. 2). Using this model, we found that (Z)- toremifene (FARESTON®), clomiphene (e.g., CLOMID®), amodiaquine (e.g., CAMOQUIN®), verapamil (e.g., CALAN®), amiodarone (e.g., NEXTERONE®), chloroquine (e.g.,
ARALEN®), hydroxychloroquine, and umifenovir (e.g., ARB IDOL®) demonstrated dose- dependent inhibition of SARS-CoV-2pp entry in Huh-7 cells when added at 1 and 5 mM simultaneously with the virus and culturing for 72 hours (FIG. 3, data not shown for hydroxychloroquine, which was similar to the chloroquine data), without producing any detectable cell toxicity in this model (FIGs. 7A-7B). Among the drugs tested, chloroquine has been reported to inhibit SARS-CoV2 in vitro. Surprisingly, at the higher dose, umifenovir inhibited viral entry. This is in contrast to recent preliminary data reporting that umifenovir, at the particular dose used, is “no better than placebo in expediting recovery” of patents infected with SARS-CoV2 (DOI: 10.3760/cma.j.cn311365-20200210-00050). We have not found reports that any of the other five FDA-approved drugs exhibit anti-SARS-CoV2 activity.
In addition to these FDA-approved drugs, we also found that CoV HR2 peptide, which is derived from the HR2 domain of the spike protein of S ARS-CoV2, and baicalein, which is a natural product isolated from Chinese herbs, such as roots of Scutellaria baicalensis and Scutellaria lateriflora, exhibited inhibitory activity on the entry of SARS-CoV2 in a dose- dependent manner (FIG. 4).
Example 3. Evaluation of repurposed entry inhibitors of SARS-CoV2 in human Lung Airway Chips.
Next, we tested these potential inhibitors of SARS-CoV2 within the more clinically relevant human Airway Chip. We first confirmed that the host cell receptor Angiotension Converting Enzyme-2 (ACE2) was highly expressed in differentiated human Airway chip (FIG. 5A). Then we tested whether CoVpp can infect clinically relevant target epithelial cells within the human Airway Chip by delivering viral particles into the lumen of the channel lined by human bronchiolar epithelial cells, and found that high viral gene levels were detected in infected cells (FIG. 5B), demonstrating CoVpp can infect differentiated human airway cells in vitro as in human lung in vivo. To explore the potential of using these drugs as COVID19 prophylactic therapies, we then pretreated the human Airway Chips by perfusing their vascular channel with nafamostat mesylate, chloroquine diphosphate, hydroxychloroquine, arbidol hydrochloride, toremifene, clomiphene citrate, amodiaquine, verapamil hydrochloride, amiodarone, HR2 protein, and imatinib at their maximum concentration (Cmax) in blood reported in humans (with the exception of nafamostat) (Table 1) to mimic systemic distribution after oral administration for 24 hours, before adding the CoV-2pp into the airway channel, and then continuously flowing the drug through the vascular channel for 48 additional hours.
Table 1. The Cmax of drugs according to clinical reports.
Figure imgf000030_0001
These studies revealed that three of these drugs - nafamostat, amodiaquine and toremifene - displayed statistically significant inhibition of viral infection under these more clinically relevant experimental conditions (FIGs. 6A-6B). qPCR quantitation of viral mRNA revealed that three of these drugs - amodiaquine, toremiphene, and clomiphene - significantly reduced viral entry (by 59.1%, 51.1% and 28.1%, respectively) (FIG. 6B) without producing detectable cytotoxicity (FIG. 7). The finding that umifenovir did not significantly inhibit the entry of CoV-2pp in the human Airway Chip when administered at a clinically relevant dose (FIGs. 6A-6B), whereas it was an effective inhibitor in Huh-7 cells under static conditions (FIG. 3), is consistent with the observation that umifenovir did not relieve symptoms or accelerate clearance of native SARS-CoV2 (also abbreviated as SARS-CoV-2) virus in a human clinical trial (Chin J Infect Dis 38, E008-E008, doi:10.3760/cma.j.cn311365-20200210-00050 (2020)). Interestingly, chloroquine did not produce statistically significant inhibition effects when administered at its Cmax in the human Airway Chip (FIGs. 6A-6B). The lack of efficacy of chloroquine seen here might be explained by the observation that chloroquine can concentrate to higher levels in lung compared to blood in vivo , or that it exerts its therapeutic effects in humans through mechanisms other than blocking viral entry. However, our results in the human Airway Chip are reminiscent of the findings indicating that while chloroquine was reported to show mild therapeutic effects in preliminary clinical studies with a small numbers of patients, it was not found to demonstrate any significant anti- viral activity in other studies, and that significant toxicity was observed, including in preliminary findings from a larger randomized, double- blinded, phase lib clinical trial.
When administered to patients, the most potent drug amodiaquine is rapidly transformed (half life ~ 5 hr) into its active metabolite, desethylamodiaquine, which has a much longer half- life (~ 9-18 days). When desethylamodiaquine was administered at a clinically relevant dose (1 mM; Table 1) in the human Airway Chips, it also reduced entry of the pseudotyped SARS-CoV-2 viral particles by -60% (FIG. 9) suggesting that both amodiaquine and its metabolite are active inhibitors of SARS-CoV-2 S protein-dependent viral entry.
Given that amodiaquine was an effective inhibitor of SARS-CoV-2 viral entry while the related anti-malarial drugs hydroxychloroquine and chloroquine were not, quantitative mass spectrometry was performed to compare the effects of these three drugs on the proteome of airway epithelial cells. Proteomics analysis revealed that amodiaquine triggered distinct and broader pertubations in host proteome compared to the other related anti-malarial drugs (data not shown) with the most differentially affected proteins being related to regulation of cilia and expression of lysosomal proteins, which may be responsible for it having greater effects against viral entry.
Statistical analysis of viral entry measurements in Huh-7 cells and airway chips.
Tests for statistically significant differences between drug-treated and untreated groups were performed using a two-tailed Student's t-test and the Bonferroni correction for multiple hypothesis testing (Bonferroni, C. E., Teoria statistica delle classi e calcolo delle probabilità, Pubblicazioni del R Istituto Superiore di Scienze Economiche e Commerciali di Firenze. 8, 3-62. 1936). Differences were considered significant when the P value was less than 0.05 (*, P<0-05; **, P<0-01; ***, P<0-001; n.s., not significant). All results are expressed as means ± standard deviation (SD); N > 3 in all studies. See Table 2.
Table 2
Figure imgf000031_0001
Figure imgf000032_0001
Example 4. Amodiaquine and desethylamodiaquine inhibit SARS-CoV-2 infection in vitro and in vivo
We tested the ability of the most potent drug identified in the Airway Chip, amodiaquine, and its metabolite desethylamodiaquine to inhibit infection by GFP-labeled SARS-CoV-2 vims at a multiplicity of infection (MOI = 0.1) in Vero E6 cells. We found that both compounds inhibited infection by SARS-CoV-2 in a dose-dependent manner (FIG. 8A) with half maximal inhibitory concentrations (IC50) of 10.3 + 1.6 and 8.5 + 3.0 mM for amodiaquine and desethylamodiaquine, respectively. Amodiaquine and desethylamodiaquine also inhibited infection by wild type SARS-CoV-2 vims when administered under less stringent conditions (MOI = 0.01), with both compounds exhibiting IC50 < 5 mM (FIG. 81). In addition, amodiaquine reduced viral load by ~ 3 logs in ACE2-expressing human lung A549 cells infected with native SARS-CoV-2 when administered at 10 mM (FIG. 8B).
Given this potent inhibitory activity against native SARS-CoV-2, we then evaluated amodiaquine in a hamster COVID-19 prevention model in which the drug was first administered subcutaneously (50 mg/kg) one day before the animals were infected intranasally with SARS- CoV-2 vims (103 PFU), and then treated daily with the same dose for 3 additional days. The animals were treated once a day for 4 days with amodiaquine (50 mg/kg via subcutaneous injection) beginning one day prior to SARS-CoV-2 infection. The dosing regimen was selected based on a PK study for amodiaquine that was carried out in healthy hamsters in parallel. A single dose of amodiaquine (50 mg/kg) injected subcutaneously revealed that the Crnax for amodiaquine and its active metabolite desethylamodiaquine were ~ 3.2 and 0.7 mM, respectively; while the Tl/2 for amodiaquine was 18.1 hours, that of its active metabolite was significantly greater than the 1 day time course analyzed (FIG. 10, Table 3), which is consistent with human clinical data. Analysis of drug concentrations in lung, kidney, intestine, and heart revealed that both drugs became concentrated in these organs relative to plasma (data not shown). Analysis of drug concentrations 24 hours after dosing revealed significant exposures of amodiaquine and desethylamodiaquine in lung, kidney, and intestine (Table 4), with levels in tissues relative to plasma enhanced 21- to 138-fold for amodiaquine and 8- to 45-fold for desethylamodiaquine. These PK results, including the extended half lives and tissue concentration for both parent compound and metabolite compounds are consistent with results of past PK studies in humans.
Table 3, PK parameters for amodiaquine and desethylamodiaquine in plasma
Figure imgf000033_0001
Table 4, Concentration of amodiaquine and desethylamodiaquine in tissues (lung, kidney, intestine, heart) and plasma 24 hours after subcutaneous dosing of 50 mg/kg amodiaquine
Figure imgf000033_0002
Prophylaxis of infected hamsters with amodiaquine beginning 1 day before infection and treating daily over the following 3 days using this dosing regimen resulted in -70% reduction in SARS-CoV-2 viral load measured by RT-qPCR of the viral N transcript when measured on the third day after the viral challenge (FIG. 8C), which is the day at which peak viral loads are observed in this model. Immunohistochemical analysis of lungs from these animals confirmed that amodiaquine treatment resulted in a significant reduction in expression of SARS-CoV-2 N protein in these tissues (FIG. 8D). To explore the potential use of using amodiaquine to prevent the spread of COVID- 19 within populations, we then carried out studies using a SARS-CoV-2 anirnal-to-anirnal transmission model in which vehicle or amodiaquine-lreaied healthy animals treated with amodiaquine for 1 day were placed in the same cage with animals that had been infected with SARS-CoV-2 virus (1(F PFU) one day earlier, In vehicle controls, this experimental setup results in a 100% transmission of infection within two days of exposure to the infected animals, in contrast, the same amodiaquine treatment regimen as described above resulted in a 90% inhibition of SARS-CoV-2 infection as measured by quantifying N transcript levels (FIG. 8E). These results were further corroborated in an independent experiment where amodiaquine-treated animals showed a greater than one log decrease in viral titers measured by plaque assays when compared to vehicle (FIG. 8F).
As amodiaquine is commonly administered as an oral dose clinically, we repeated these studies with drug administered through oral gavage at a dose (75 mg/kg) that produced similar PK parameters to those observed with the subcutaneous administration (Cmax for amodiaquine and desethylamodiaquine of ~ 1.8 and 4.5 mM, respectively; Tl/2 for amodiaquine was 12.8 hours and again greater than 24 hours for its active metabolite (FIG. 11, Tables 5). However, oral administration resulted in even higher levels in tissues compared to plasma (enhanced 29- to 331-fold for amodiaquine and 8- to 119-fold for desethylamodiaquine) (Table 6). Importantly, pretreatment for 1 day with oral amodiaquine prevented infection of native SARS-CoV-2 (FIG. 8G) to a similar degree as when administered subcutaneously (FIG. 8C), whereas when we use the same prevention model to test the closely related anti-malarial drug, hydroxychloroquine at a dose (50 mg/kg) previously shown to produce a clinically relevant level of lung exposure in hamsters41, it had no inhibitory activity (FIG. 8G), much as we observed in our human Organ Chip model (FIG. 6A-6B). Furthermore, RNA seq analysis of infected hamsters treated with oral amodiaquine revealed that drug treatment resulted in a significant down regulation of genes associated with the inflammatory response, including those involved in signaling through TNF-a and NF-kB, IL-6 JAK STAT3, and interferon-g (FIG. 12, Table 7).
Table 5. PK parameters for amodiaquine and desethylamodiaquine in plasma
Figure imgf000034_0001
Table 6. Concentration of amodiaquine and desethylamodiaquine in tissues (lung, kidney, intestine, heart) and plasma 24 hours after oral dosing of 75 mg/kg amodiaquine
Figure imgf000035_0001
Table 7. Top significant enriched pathways identified by Gene Set Enrichment Analysis (GSEA)
Figure imgf000035_0002
Finally, given the robustness of these responses, we tested amodiaquine's ability to inhibit SARS-CoV-2 infection when administered in a treatment mode, beginning one day after infection. These studies revealed that post-infection treatment with amodiaquine produces similar inhibition of infection by -70% at day 3, with complete clearance of detectable viral N transcripts in lung by day 7 (FIG. 8H). These findings confirm that the effects we observed were due to effective resolution of infection rather than to a time delay in the response. Taken together, these results confirm that the antiviral activities exhibited by amodiaquine in the human Lung Airway Chips translate to the in vivo setting and suggest that oral amodiaquine may provide significant protection when administered prophylactically or therapeutically. Table 8. Summary of estimated ratio of free drug in target tissue and plasma over EC50 and EC90
Figure imgf000035_0003
Figure imgf000036_0001
*Based on drug concentration at 24 hours post-dosing and estimated 92.5% protein binding from published studies in human plasma **Based on concentration at CMax and estimated 92.5% protein binding from published studies in human plasma
Methods
3-5 week-old Syrian hamsters were acclimated to the CDC/USDA-approved BSL-3 facility of the Global Health and Emerging Pathogens Institute at the Icahn School of Medicine at Mount Sinai for 2-4 days. Hamsters were given a subcutaneous injection posteriorly with drug treatment within two hours of drug reconstitution the day before SARS-CoV-2 infection and every day thereafter until terminal lung harvest. Amodiaquine was reconstituted in 12% sulfobutylether-β-cyclodextrin (Selleckchem) in water(w/w) (with HC1/NaOH) at pH 5.0. Animals were anesthetized by intraperitoneal injection of 200 μl of ketamine and xylazine (4:1) and provided thermal support while unconscious. Hamsters were intranasally infected with 1 * 103 PFU of passage 3 SARS-CoV-2 USA-WA1/2020 in 100 μl of PBS. The animals were weighed daily and on day 3 of infection were sacrificed. Whole lungs were harvested and homogenized in lmL of PBS. Homogenates were then spun down at 10,000rcf for 5 minutes. Supernatant was subsequently titered by plaque assay and the lung pellet was resuspended in Trizol.
Lung RNA was extracted by phenol chloroform extraction and DNase treated using DNA-free DNA removal kit (Invitrogen). After cDNA synthesis of RNA samples by reverse transcription using Superscript II Reverse Transcriptase (invitrogen) with oligo d(T) primers, quantitative RT-PCR was performed using KAPA SYBR FAST qPCR Master Mix Kit (Kapa Biosystems) on a LightCycler 480 Instrument II (Roche) for subgenomic nucleocapsid (N) RNA (sgRNA) and actin using the following primers: Actin forward primer: 5’- CC AAGGCC AACCGTGAAAAG-3 ’ , Actin reverse primer 5’- ATGGCTACGTAC ATGGCTGG-3 ’ , N sgRNA forward primer: 5’- CTCTTGTAGATCTGTTCTCTAAACGAAC-3’, N sgRNA reverse primer: 5’- GGTCCACCAAACGTAATGCG-3’ Relative sgRNA levels were quantified by normalizing sgRNA to actin expression and normalizing drug-treated infected lung RNA to vehicle-treated infected controls. Significance was determined using an unpaired one-tailed student's t-test.
Virus and plaque assay
SARS -related coronavirus 2 (SARS-CoV-2), Isolate USA-WA1/ 2020 (NR-52281) was deposited by the Center for Disease Control and Prevention and obtained through BEI Resources, NIAID, NIH. SARS-CoV-2 was propagated in Vero E6 cells in DMEM supplemented with 2% FBS, 4.5 g/L D-glucose, 4 mM L-glutamine, 10 mM Non-Essential Amino Acids, 1 mM Sodium Pyruvate and 10 mM HEPES and filtered through an Amicon Ultracel 15 (100kDa) centrifugal filter. Flow through was discarded and virus resuspended in DMEM supplemented as above. Infectious titers of SARS-CoV-2 stock and hamster lung homogenates were determined by plaque assay in Vero E6 cells in Minimum Essential Media supplemented with 2% FBS, 4 mM L-glutamine, 0.2% BSA, 10 mM HEPES and 0.12% NaHCO3 and 0.7% agar.
The efficacy of the agents described herein may be farther increased by increasing drug levels, either via intravenous administration, through delivery of drug directly to the lung airway (e.g., via aerosol, nebulizer, or tracheal wash), or both. The efficacy may increase further when used against native SARS-CoV2, as inhibition of viral entry will effectively block the propagation of many progeny virus, and thus the untreated virus will increase many more fold in number in controls whereas the drug treated should show a similar total low number of progeny.
The efficacy of these drugs against entry of SARS-CoV2 are currently under further validation by varying the concentrations and delivery routes in human Lung Airway Chips, Lung Alveolus Chips, and lntestine Chips.
These inhibitors of viral entry should synergize with inhibitors of viral infection and response to infection that work via distinct mechanisms, such as inhibitors of viral budding, inhibitors of proteases required for activation of entry' (e.g,, nafarnostat, casmoslat, and aprotinin), inhibitors of viral replication, and inhibitors of membrane fusion, as well as modulators of the host inflammatory response.
References
1. Benam KH, Villenave R, Lucchesi C, et al. Small airway-on-a-chip enables analysis of human lung inflammation and drug responses in vitro. Nat Methods 2016; 13(2): 151-7.
2. Benam KH, Novak R, Nawroth J, et al. Matched-Comparative Modeling of Normal and Diseased Human Airway Responses Using a Microengineered Breathing Lung Chip. Cell Syst 2016; 3(5): 456-66 e4.
3. Huh D, Matthews BD, Mammoto A, Montoya-Zavala M, Hsin HY, Ingber DE. Reconstituting organ-level lung functions on a chip. Science 2010; 328(5986): 1662-8.
4. Huh D, Leslie DC, Matthews BD, et al. A human disease model of drug toxicity-induced pulmonary edema in a lung-on-a-chip microdevice. Sci Transl Med 2012; 4(159): 159ra47.
5. Longlong Si RP-B, Kambez H Benam, Haiqing Bai, Melissa Rodas, Morgan Burt, Donald E. Ingber. Discovery of influenza drug resistance mutations and host therapeutic targets using a human airway chip. bioRxiv 2019. 6. Yu M, Si L, Wang Y, et al. Discovery of pentacyclic triterpenoids as potential entry inhibitors of influenza viruses. J Med Chem 2014; 57(23): 10058-71.
7. Si L, Meng K, Tian Z, et al. Triterpenoids manipulate a broad range of virus-host fusion via wrapping the HR2 domain prevalent in viral envelopes. Sci Adv 2018; 4(11): eaau8408.
All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.
The indefinite articles “a” and “an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean “at least one.”
It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,”
“composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of’ and “consisting essentially of’ shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03. The terms “about” and “substantially” preceding a numerical value mean ±10% of the recited numerical value.
Where a range of values is provided, each value between the upper and lower ends of the range are specifically contemplated and described herein.

Claims

What is claimed is: CLAIMS
1. A method for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of an agent selected from the group consisting of amodiaquine, (Z)-toremifene, clomiphene, verapamil, amiodarone, baicalein, and CoV HR2 peptide.
2. A method for preventing coronavirus infection, comprising administering to a subject at risk of coronavirus a composition comprising a therapeutically effective amount of an agent selected from the group consisting of amodiaquine, (Z)-toremifene, clomiphene, verapamil, amiodarone, baicalein, and CoV HR2 peptide.
3. The method of claim 1 or 2, wherein the coronavirus is a beta coronavirus.
4. The method of claim 3, wherein the beta coronavirus is SARS-CoV2.
5. The method of any one of the preceding claims, wherein the composition comprises two, three, four, five, six, or seven agents selected from the group consisting of amodiaquine, (Z)- toremifene, clomiphene, verapamil, amiodarone, baicalein, and CoV HR2 peptide.
6. The method of any one of the preceding claims, wherein the composition comprises amodiaquine.
7. The method of any one of the preceding claims, wherein the composition comprises (Z)- toremifene.
8. The method of any one of the preceding claims, wherein the composition comprises clomiphene.
9. The method of any one of the preceding claims, wherein the composition comprises verapamil.
10. The method of any one of the preceding claims, wherein the composition comprises amiodarone.
11. The method of any one of the preceding claims, wherein the composition comprises baicalein.
12. The method of any one of the preceding claims, wherein the composition comprises a CoV HR2 peptide or a variant CoV HR2 peptide.
13. The method of claim 12, wherein the composition comprises a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
14. The method of claim 12, wherein the composition comprises a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
15. The method of claim 14, wherein the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
16. The method of any one of the preceding claims, wherein the therapeutically effective amount is an amount effective for decreasing coronavirus infectivity.
17. The method of claim 16, wherein the therapeutically effective amount is an amount effective for decreasing viral infectivity by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, or at least 80% relative to a control.
18. The method of any one of the preceding claims, wherein the therapeutically effective amount is an amount effective for increasing the rate of inhibition of coronavirus infection, relative to a control.
19. The method of claim 18, wherein the therapeutically effective amount is an amount effective for increasing the rate of inhibition of coronavirus infection by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, or at least 80% relative to a control.
20. The method of any one of the preceding claims, wherein the composition further comprises a pharmaceutically-acceptable excipient.
21. The method of any one of the preceding claims, wherein the subject is a human subject.
22. The method of claim 21, wherein the subject is immunocompromised.
23. The method of claim 21 or 22, wherein the subject is a child or an elderly subject.
24. The method of any one of the preceding claims, wherein the composition is administered nasally, orally, intravenously, intramuscularly, subcutaneously, or intraperitoneally.
25. The method of any one of the preceding claims, wherein the composition is administered to the lung airway of the subject.
26. The method of claim 25, wherein the composition is administered to the lung airway via an aerosol, a nebulizer, or a tracheal wash.
27. The method of any one of the preceding claims, wherein the composition is administered as a single dose.
28. The method of any one of the preceding claims, wherein the composition is administered as multiple doses.
29. The method of claim 30, wherein the composition is administered at multiple time points.
30. The method of claim 29, wherein the composition is administered weekly, monthly, every six months, yearly, every 5 years, or every 10 years.
31. A composition comprising at least two agents selected from the group consisting of amodiaquine, (Z)-toremifene, clomiphene, verapamil, amiodarone, baicalein, and CoV HR2 peptide.
32. A composition comprising a therapeutically effective amount of a variant CoV HR2 peptide that comprises a CoV HR2 peptide having a single amino acid modification relative to a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
33. A composition comprising a therapeutically effective amount of a CoV HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1.
34. The composition of claim 33, wherein the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
35. The composition of any one of claims 31-34, further comprising a pharmaceutically- acceptable excipient.
36. A method for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of a SARS-CoV2 HR2 peptide that comprises an amino acid sequence having at least 95% identity to the amino acid sequence of SEQ ID NO: 1, wherein the composition is administered to a lung airway of the subject.
37. The method of claim 36, wherein the composition comprises a CoV HR2 peptide that comprises the amino acid sequence of SEQ ID NO: 1.
38. The method of claim 36 or 37, wherein the composition is administered via an aerosol, a nebulizer, or a tracheal wash.
39. A method for treating coronavirus infection, comprising administering to a subject infected with coronavirus a composition comprising a therapeutically effective amount of amodiaquine, wherein viral load in the subject is reduced by at least 40% relative to a control.
40. The method of claim 39, wherein viral load in the subject is reduced by at least 50%, by at least 60%, or by at least 70% relative to a control.
41. The method of claim 39 or 40, wherein the coronavirus is a beta coronavirus.
42. The method of claim 41, wherein the beta coronavirus is SARS-CoV2.
43. The method of any one of the preceding claims, further comprising administering to the subject an agent selected from antiviral agents, antibacterial agents, and anti-inflammatory agents.
PCT/US2021/021427 2020-03-09 2021-03-09 Compositions and methods for treating coronavirus WO2021183463A1 (en)

Applications Claiming Priority (12)

Application Number Priority Date Filing Date Title
US202062987024P 2020-03-09 2020-03-09
US62/987,024 2020-03-09
US202062993816P 2020-03-24 2020-03-24
US62/993,816 2020-03-24
US202063000772P 2020-03-27 2020-03-27
US63/000,772 2020-03-27
US202063028312P 2020-05-21 2020-05-21
US63/028,312 2020-05-21
US202063051601P 2020-07-14 2020-07-14
US63/051,601 2020-07-14
US202063069583P 2020-08-24 2020-08-24
US63/069,583 2020-08-24

Publications (1)

Publication Number Publication Date
WO2021183463A1 true WO2021183463A1 (en) 2021-09-16

Family

ID=77671937

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/021427 WO2021183463A1 (en) 2020-03-09 2021-03-09 Compositions and methods for treating coronavirus

Country Status (1)

Country Link
WO (1) WO2021183463A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023085392A1 (en) * 2021-11-12 2023-05-19 国立大学法人鹿児島大学 Anti-sars-cov-2 drug
US20230255924A1 (en) * 2020-04-28 2023-08-17 Manoj P. Jadhav Use of verapamil for the treatment of sars-cov-2 induced covid-19

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005002500A2 (en) * 2003-04-14 2005-01-13 Merck & Co., Inc. Inhibitors of coronavirus
US20090149545A1 (en) * 2003-05-28 2009-06-11 Tsu-An Hsu Treatment of coronavirus infection
US20090304683A1 (en) * 2006-01-19 2009-12-10 Dimitrov Dimiter S Soluble Fragments of The Sars-Cov Spike Glycoprotein
US20160113920A1 (en) * 2014-10-24 2016-04-28 Keck Graduate Institute Of Applied Life Sciences Compositions and methods for inhibiting bacterial and viral pathogens
US20160237123A1 (en) * 2008-01-23 2016-08-18 Dana-Farber Cancer Institute, Inc. Compositions and methods for the treatment of viral infections
US20170027975A1 (en) * 2014-04-07 2017-02-02 University Of Maryland, Baltimore Methods of Treating Coronavirus Infection

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2005002500A2 (en) * 2003-04-14 2005-01-13 Merck & Co., Inc. Inhibitors of coronavirus
US20090149545A1 (en) * 2003-05-28 2009-06-11 Tsu-An Hsu Treatment of coronavirus infection
US20090304683A1 (en) * 2006-01-19 2009-12-10 Dimitrov Dimiter S Soluble Fragments of The Sars-Cov Spike Glycoprotein
US20160237123A1 (en) * 2008-01-23 2016-08-18 Dana-Farber Cancer Institute, Inc. Compositions and methods for the treatment of viral infections
US20170027975A1 (en) * 2014-04-07 2017-02-02 University Of Maryland, Baltimore Methods of Treating Coronavirus Infection
US20160113920A1 (en) * 2014-10-24 2016-04-28 Keck Graduate Institute Of Applied Life Sciences Compositions and methods for inhibiting bacterial and viral pathogens

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20230255924A1 (en) * 2020-04-28 2023-08-17 Manoj P. Jadhav Use of verapamil for the treatment of sars-cov-2 induced covid-19
US11938110B2 (en) * 2020-04-28 2024-03-26 Jadhav Manoj P Use of verapamil for the treatment of SARS-Cov-2 induced COVID-19
WO2023085392A1 (en) * 2021-11-12 2023-05-19 国立大学法人鹿児島大学 Anti-sars-cov-2 drug

Similar Documents

Publication Publication Date Title
Choy et al. Remdesivir, lopinavir, emetine, and homoharringtonine inhibit SARS-CoV-2 replication in vitro
Hashem et al. Therapeutic use of chloroquine and hydroxychloroquine in COVID-19 and other viral infections: A narrative review
Klimke et al. Hydroxychloroquine as an aerosol might markedly reduce and even prevent severe clinical symptoms after SARS-CoV-2 infection
KR102174576B1 (en) Mek inhibitors in the treatment of virus diseases
WO2021183463A1 (en) Compositions and methods for treating coronavirus
US11376232B2 (en) Vidofludimus for use in the treatment or prevention of viral diseases
US20230346820A1 (en) Methods and compositions for the treatment of sars-cov-2
CN113679726A (en) Application of salvia miltiorrhiza extract and quinone compounds in resisting coronavirus
CN112912074A (en) EGCG-palmitate compositions and methods of use thereof
WO2020241759A1 (en) Prophylactic and/or therapeutic agent for influenza virus infection or corona virus infection
US20090318379A1 (en) Statins for the Treatment of Viral Influenza Infections
JP2023519277A (en) Pharmaceutical composition for preventing or treating infectious RNA virus infection
US7341988B2 (en) Method of treating influenza with geranyl-geranyl acetone
WO2017083971A1 (en) Compositions and methods for treatment of influenza
RU2346692C2 (en) Application of 9-oxoacridine-10-acetic acid, its salts and esters in combined therapy for ovarian carcinoma, method of treatment and sets
AU2021317737A1 (en) Use of Wnt/Beta-catenin pathway inhibitors to block replication of SARS-CoV-2 and other pathogenic viruses
JP2023517639A (en) Compositions containing diltiazem for treating viral infections caused by SARS-CoV-2 virus
CN114786659A (en) MEK inhibitors for the treatment of hantavirus infection
US20230210816A1 (en) Inhibition of arenaviruses by combinations of approved therapeutic drugs
CN114377015B (en) Application of naftopidil in preparation of anti-influenza virus drugs
RU2794315C1 (en) Method for prevention or treatment of coronavirus and other acute respiratory viral infections
US20230277524A1 (en) Combination therapy for treatment of viral infections
WO2022232337A2 (en) Composition and method for treating covid-19
AU2022246073A1 (en) Compositions and methods for inhibiting and treating viral infections
JP2024514770A (en) Compounds for use in the treatment and prevention of COVID-19

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: 21767185

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21767185

Country of ref document: EP

Kind code of ref document: A1