WO2022123513A1 - Composition and method for managing coronavirus infection - Google Patents

Composition and method for managing coronavirus infection Download PDF

Info

Publication number
WO2022123513A1
WO2022123513A1 PCT/IB2021/061564 IB2021061564W WO2022123513A1 WO 2022123513 A1 WO2022123513 A1 WO 2022123513A1 IB 2021061564 W IB2021061564 W IB 2021061564W WO 2022123513 A1 WO2022123513 A1 WO 2022123513A1
Authority
WO
WIPO (PCT)
Prior art keywords
composition
procyanidin
type
coronavirus
agents
Prior art date
Application number
PCT/IB2021/061564
Other languages
French (fr)
Inventor
Sunil Bhaskaran
Original Assignee
Indus Biotech Private Limited
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Indus Biotech Private Limited filed Critical Indus Biotech Private Limited
Publication of WO2022123513A1 publication Critical patent/WO2022123513A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/18Magnoliophyta (angiosperms)
    • A61K36/185Magnoliopsida (dicotyledons)
    • A61K36/54Lauraceae (Laurel family), e.g. cinnamon or sassafras
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2236/00Isolation or extraction methods of medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicine

Definitions

  • the present disclosure relates to the field of immunology, more specifically, to the field of virology, biotechnology and medicine.
  • the present disclosure provides a botanical composition effective against viruses such as but not limited to strains of Coronavirus and mutants or variants thereof.
  • the present disclosure provides a composition comprising procyanidins selected from a group comprising pentameric procyanidin, tetrameric procyanidin and trimeric procyanidin or any combination thereof, optionally along with pharmaceutically acceptable excipient(s) for use in the management, treatment and/or prevention of diseases or infections caused by the different strains of Coronavirus and complications arising from such infections.
  • the present disclosure thus also provides a method for the management, treatment and/or prevention of Coronavirus infections and/or associated complications.
  • Catechins are polyphenolic plant metabolites which belong to the flavonoid family. The molecular formula and weight of catechins are C isH Or, and 290 g/mol, respectively. Catechin and epicatechin are epimers, with (-)-epicatechin and (+)-catechin being the most common optical isomers found in nature.
  • Procyanidins or condensed tannins are flavonoid oligomers whose building blocks are (+) - catechin and (-) - epicatechin. They are present abundantly in the plant kingdom in fruits, barks, leaves and seeds where they provide protection against light, oxidation and predators. Procyanidins are found in many plants, mainly apples, pine bark, cinnamon bark, litchi pericarp, peanuts, grape seed, cocoa, grape skin, bilberry, cranberry, black currant, green tea and black tea.
  • procyanidins are classified as Types A, B or C polyphenols.
  • the linkage between successive monomeric units of procyanidins is between the 4th position of the ‘upper’ unit and the 8th position of the ‘lower’ unit, leading to a Type B procyanidin.
  • the linkage can occur between C4 of the ‘upper’ unit and C6 of the lower unit, leading to a Type C procyanidin.
  • Type B and C polyphenols are abundantly seen in many botanical sources.
  • Coronaviruses are a group of related viruses known to cause diseases in mammals and birds. Infection by said viruses, in humans, is known to lead to respiratory tract infections that can range from mild to lethal.
  • Coronavirus infections in the form of SARS, and COVID-19 have had alarming implications.
  • Coronavirus disease 2019 (COVID-19) which took the form of a pandemic in early 2020 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2).
  • SARS-CoV-2 severe acute respiratory syndrome coronavirus 2
  • the virus is known to cause lung pathology, sepsis and renal failure.
  • a robust cure/treatment regime for Coronavirus is however yet to be found.
  • COVID-19 coronavirus disease 2019 (COVID-19) may appear 2 to 14 days after exposure. This time after exposure and before having symptoms is called the incubation period. The subject can still spread COVID- 19 before he has symptoms (Pre-symptomatic transmission).
  • COVID- 19 Common signs and symptoms are Fever, Cough, Tiredness, Shortness of breath or difficulty breathing, Muscle aches, Chills, Sore throat, Runny nose, Headache, Chest pain, Pink eye (conjunctivitis), Nausea, Vomiting, and Diarrhea.
  • early symptoms of COVID- 19 may include a loss of taste or smell.
  • Certain medical conditions that may increase the risk of serious illness from COVID- 19 may be Serious heart diseases, such as heart failure, coronary artery disease or cardiomyopathy, Chronic obstructive pulmonary disease (COPD), Type 1 or type 2 diabetes, Overweight, obesity or severe obesity, High blood pressure and Asthma.
  • Serious heart diseases such as heart failure, coronary artery disease or cardiomyopathy, Chronic obstructive pulmonary disease (COPD), Type 1 or type 2 diabetes, Overweight, obesity or severe obesity, High blood pressure and Asthma.
  • COVID-19 symptoms can range from very mild to severe. Some people may have only a few or mild symptoms. Some people may experience more severe symptoms, such as worsened shortness of breath and pneumonia, about a week after symptoms start. Chronic lung diseases such as cystic fibrosis or pulmonary hypertension. Although most people with COVID-19 have mild to moderate symptoms, the disease can cause severe medical complications and lead to death in some people. Older adults or people with existing medical conditions are at greater risk of becoming seriously ill with COVID-19.
  • the Complications can include Pneumonia and trouble breathing, Organ failure in several organs, cardiac problems and blood clots. And a severe lung condition that causes a low amount of oxygen to go through your bloodstream to your organs (acute respiratory distress syndrome).
  • SPO2 Oxygen Saturation
  • the viral persistence carries even after the symptoms goes by and the syndrome is called Long Covid with symptoms such as fatigue and decrease in quality of life.
  • the U.S. Food and Drug Administration has given emergency use authorization to some COVID- 19 vaccines in the United States.
  • the FDA has approved the Pfizer-BioNTech COVID- 19 vaccine, now called Comimaty, to prevent COVID-19 in people age 16 and older.
  • the FDA has given emergency use authorization to Pfizer-BioNTech COVID- 19 vaccines for ages 5 through 15.
  • the CDC recommends additional doses and booster doses of COVID- 19 vaccines in specific instances for some people with weakened immune systems, such as those who have had an organ transplant. People with weakened immune systems might not develop enough immunity after vaccination with two doses of an mRNA COVID-19 vaccine. An additional dose might improve their protection against COVID- 19.
  • An additional dose might improve their protection against COVID- 19.
  • Coronavirus entry into the host cell is mediated by a completely different process compared to HIV and H1N1.
  • the spike protein S has a Receptor Binding Domain (RBD). This binds to the Angiotensin Converting Enzyme 2 (ACE2) expressed on the host cell.
  • ACE2 Angiotensin Converting Enzyme 2
  • a protease enzyme expressed near the ACE2 TMPRSS2 and/or Furin, depending on the cell
  • TMPRSS2 and/or Furin cleaves the spike protein and facilitates the fusion of the virus into the host cell.
  • antiviral drugs known for treatment of HIV and H IN 1 are not effective in preventing and/or treating Coronavirus infections.
  • the present invention addresses the need for a drug and a method directed towards Coronavirus infections and provides a composition and a method that targets the infection mechanism of the virus to inhibit the entry of the virus, improves oxygen saturation levels, prevents further progression of the disease by blocking the entry of the virus and improves quality of life of people suffering from the disease.
  • the present disclosure provides a suitable composition fit for said application and methods employing the same.
  • the present invention provides a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in preventing, managing or treating Coronavirus infection and/or associated complication(s).
  • the Coronavirus infection is caused by SARS-CoV-2 or mutants or variants thereof.
  • the associated complication is selected from a group comprising reduction in quality of life, reduction in oxygen saturation (SpCh) levels, pneumonia, troubled breathing, blood clot formation, cardiac problems and organ failure or any combination thereof.
  • SpCh reduction in oxygen saturation
  • the associated complication is selected from a group comprising reduction in quality of life and/or reduction in oxygen saturation (SpCh) levels.
  • SpCh oxygen saturation
  • the pentameric type A procyanidin is at concentration ranging from about 55 % w/w to about 99% w/w
  • the tetrameric type A procyanidin and the trimeric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 35 % w/w
  • the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
  • the pentameric type A procyanidin is at concentration ranging from about 80 % w/w to about 90% w/w
  • the tetrameric type A procyanidin and the trimeric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 20 % w/w
  • the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
  • the pharmaceutically acceptable excipient(s) as referred to above is selected from a group comprising gums, granulating agents, binding agents, lubricating agents, disintegrating agents, sweetening agents, coloring agents, flavoring agents, coating agents, plasticizers, preservatives, suspending agents, emulsifying agents, antistatic agents, and spheronization agents or any combination thereof.
  • the composition is formulated into dosage forms selected from group comprising solid formulation, liquid formulation, parenteral formulation, aerosol or spray, phytoceutical, nutraceutical and medicinal foods or any combinations thereof.
  • the solid oral formulation is selected from group comprising tablet, capsule, troche, lozenge, dispersible powder, dispersible granule or any combinations thereof;
  • the liquid oral formulation is selected from group comprising aqueous or oily suspension, emulsion, drops, emulsion in hard or soft gel capsule, syrup, elixir or any combinations thereof;
  • the parenteral formulation is selected from group comprising intravenous injection, intramuscular injection, intramuscular depot, subcutaneous injection, percutaneous injection or any combinations thereof;
  • the aerosol formulation is selected from a group comprising oral spray, nasal spray and inhaler.
  • the composition is formulated into a nasal spray.
  • the aforesaid use is as a medicament for preventing, managing, or treating Coronavirus infection and/or associated complication(s).
  • a method for preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell comprising contacting the host cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s).
  • the composition inhibits formation of a complex between ACE2 receptor of a host cell and Spike protein of the Coronavirus.
  • the composition binds to the ACE2 receptor of the host cell.
  • the composition binds to Receptor Binding Domain (RBD) of spike protein of the Coronavirus.
  • RBD Receptor Binding Domain
  • Also envisaged herein is a method of preventing, treating and/or managing Coronavirus infection and/or associated complication(s) comprising administering a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) to a subject in need thereof, at a pharmaceutically effective dosage.
  • the pharmaceutically effective dosage ranges from about 1 mg/kg to about 100 mg/kg body weight of the subject.
  • the subject is a mammal, including human beings.
  • the method improves oxygen saturation in an infected subject to a value from about 89% to about 100%.
  • kits comprising a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s), along with a pamphlet or brochure indicating suitable dosage of the composition.
  • the kit further comprises means for administration of the composition.
  • Figure 1 depicts the reduction in binding of ACE2 receptor of host cell (hereinafter referred to as ACE2) to COVID spike 1 protein caused by the composition of the present disclosure.
  • ACE2 ACE2 receptor of host cell
  • Figure 2 depicts the standard curve of ACE2 binding with coated spike 1 protein.
  • Figure 3 depicts blocking of the ACE2-spike 1 protein interaction by the composition of the present disclosure.
  • Figure 4 depicts blocking of the ACE2-spike 1 protein interaction by the composition of the present disclosure
  • Figure 5 depicts binding capacity of the composition of the present disclosure to ACE2 receptor as measured by Bio-Layer Interferometry (BLI) using Octet Red 96.
  • Figure 6 depicts binding capacity of the composition of the present disclosure to SARS-CoV2 RBD as measured by Bio-Layer Interferometry (BLI) using Octet Red 96.
  • Figure 7 depicts neutralization of pseudotyped virus with spike protein of SARS-CoV-2 by the composition of the present disclosure.
  • the dotted line indicates the 50% inhibition rate.
  • Data are shown as mean ⁇ SD (error bar) from three different experiments.
  • Figure 8 depicts inhibitory activity of the composition against authentic SARS-CoV-2 as observed in a plaque-reduction assay
  • A) depicts observation in the plaque inhibition assay
  • B) depicts analysis of IC50 of the composition
  • C) depicts cell cytotoxicity analysis of the composition .
  • Figure 9 depicts inhibitory activity of the composition against SARS-CoV-2 at the stage of viral entry and post viral entry A) depicts observation in the plaque inhibition assay; B) depicts % reduction as observed in the plaque reduction assay. DETAILED DESCRIPTION OF THE DISCLOSURE
  • Coronaviruses are typically characterized by the presence of an outer envelope, wherein said envelope consists of a lipid bilayer. Membrane, envelope and spike proteins are embedded within said lipid bilayer. As mentioned above, the mechanism of viral entry into the host cell is primarily governed by spike proteins, wherein the viral membrane fuses with the cell membrane of the host upon binding of the spike protein the host cell receptor. Once said fusion occurs, it leads to entry of the viral genome into the host cells which causes the infection.
  • the present disclosure provides a composition fit for application in the management of said infections.
  • the present disclosure further provides a method for the management of infections caused by Coronavirus.
  • reference to ‘subject’ throughout the present disclosure implies reference to a subject having or suspected of having Coronavirus infection i.e. a subject infected or suspected to be infected with Coronavirus.
  • the subject may be mammal such as an animal or a human being.
  • composition implies reference to the composition of the present disclosure as per any of the embodiments defining the composition, in all possible permutations and combinations between each of said embodiments.
  • coronavirus throughout the present disclosure envisages reference to any virus/viral strain belonging to the family of Coronavirus and all possible mutants thereof.
  • the coronavirus is selected from a group comprising 229E (alpha coronavirus), NL63 (alpha coronavirus), OC43 (beta coronavirus), HKU 1 (beta coronavirus), MERS-CoV, SARS-CoV. SARS-CoV-2 and any mutants or variants thereof.
  • Coronavirus infection in the context of the present invention, may be simultaneously caused by one or more of the above defined strains or variants of coronavirus.
  • coronavirus infection in the context of the present disclosure envisages active infection as well as associated complications during or after the infection.
  • Associated complications include but are not limited to reduction in quality of life, reduction in oxygen saturation (SpCh) levels, pneumonia, troubled breathing, blood clot formation, cardiac problems, organ failure and other identified as well as unidentified complications that bear any association with coronavirus infection.
  • SpCh oxygen saturation
  • ACE2 or ‘ACE2 receptor’ in the present disclosure is with respect to the functional receptor on host cell surfaces through which SARS-CoV-2 enters the host cells.
  • the present disclosure relates to the management, mitigation and resolution of infection caused by the Coronavirus family that are typically known to cause systematic acute respiratory syndrome and forms thereof.
  • the present invention provides a composition and its applications in the treatment, management and/or prevention of Coronavirus infection.
  • the present disclosure provides a composition comprising a mixture of procyanidin compounds for preventing, managing or treating Coronavirus infection and associated complications.
  • composition comprising pentameric type A procyanidin, trimeric type A procyanidin and tetrameric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in preventing, managing, or treating Coronavirus infection and/or associated complications.
  • the Coronavirus infection is caused by SARS-CoV2, including different variants and/or mutants thereof.
  • Associated complications include those that arise during or after the infection.
  • associated complications include but are not limited to reduction in quality of life, reduction in oxygen saturation levels, and other identified as well as unidentified complications that bear an association with coronavirus infection.
  • the associated complications are selected from a group comprising reduction in quality of life, reduction in oxygen saturation levels, pneumonia, troubled breathing, blood clot formation, cardiac problems, organ failure or any combination thereof.
  • composition comprising one or more of pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the prevention of Coronavirus infections and/or associated complications.
  • composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the treatment and/or management of Coronavirus infections and/or associated complications.
  • the pentameric type A procyanidin is at concentration ranging from about 55 % w/w to about 99% w/w
  • the trimeric type A procyanidin and the tetrameric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 35 % w/w
  • the pharmaceutically acceptable excipient is at concentration ranging from about 0.5% to about 99.9%.
  • the pentameric type A procyanidin is at concentration ranging from about 80 % w/w to about 90% w/w
  • the trimeric procyanidin and the tetrameric procyanidin are each at concentration ranging from about 0.5 % w/w to about 20 % w/w
  • the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
  • the procyanidins in any of the above defined compositions of the present disclosure are derived from Cinnamon.
  • pharmaceutically acceptable excipients in the above defined composition are selected from a group comprising gums, granulating agents, binding agents, lubricating agents, disintegrating agents, sweetening agents, coloring agents, flavoring agents, coating agents, plasticizers, preservatives, suspending agents, emulsifying agents, antistatic agents, and spheronization agents or any combination thereof.
  • the composition is formulated into dosage forms selected from group comprising solid formulation, liquid formulation, parenteral formulation, aerosol or spray, phytoceutical, nutraceutical and medicinal foods or any combinations thereof.
  • the solid oral formulation is selected from group comprising tablet, capsule, troche, lozenge, dispersible powder, dispersible granule or any combinations thereof.
  • the liquid oral formulation is selected from group comprising aqueous or oily suspension, emulsion, drops, emulsion in hard or soft gel capsule, syrup, elixir, and oral sprays or any combinations thereof.
  • the parenteral formulation is selected from group comprising intravenous injection, intramuscular injection, intramuscular depot, subcutaneous injection, percutaneous injection or any combinations thereof.
  • the aerosol formulation is selected from a group comprising oral spray, nasal spray and inhaler.
  • the composition is formulated into aerosols or sprays such as but not limited to oral sprays, nasal sprays or inhalers.
  • composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) in the prevention of Coronavirus infections and/or associated complications.
  • the present disclosure provides use of the composition comprising the pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) in the treatment and/or management of Coronavirus infections and/or associated complications.
  • the use is as a medicament for the prevention, management or treatment of Coronavirus infections and/or associated complications.
  • the present disclosure further provides the composition, as per any of the above embodiments, for use in the manufacture of medicaments directed towards prevention, management or treatment of Coronavirus infections and/or associated complications.
  • composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the manufacture of a medicament for the prevention of Coronavirus infections and/or associated complications.
  • composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the manufacture of a medicament for the treatment and/or management of Coronavirus infections and/or associated complications.
  • the present disclosure further provides use of a composition pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) for the manufacture of a medicament for prevention, management or treatment of Coronavirus infections and/or associated complications.
  • the present disclosure provides use of a composition pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) for the manufacture of a medicament for the prevention of Coronavirus infections and/or associated complications.
  • the present disclosure provides use of a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) for the manufacture of a medicament for the management or treatment of Coronavirus infections and/or associated complications.
  • each of the above applications of the composition of the present disclosure may be in-vitro, in-vivo or ex-vivo applications.
  • the present disclosure further relates to the above-described composition for use in preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell.
  • said use is an in-vitro use.
  • said use is an ex-vivo use.
  • the present disclosure in some embodiments, provides a composition comprising pentameric type A procyanidin, trimeric procyanidin and tetrameric procyanidin, optionally along with pharmaceutically acceptable excipient for use in preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell in vitro.
  • the present disclosure provides a method for preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell, comprising contacting the host cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s).
  • said method is an in-vitro method. In some embodiments, said method is an ex-vivo method.
  • the present disclosure provides a method of preventing entry of Coronavirus into a host cell, said method comprising the step of contacting the cell with a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s).
  • the present disclosure further provides an in-vitro method of preventing entry of Coronavirus into a host cell, said method comprising the step of contacting the cell with a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s).
  • the present disclosure also provides a method of inhibiting survival and/or propagation of live Coronavirus in a host cell, said method comprising the step of contacting the cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally in combination with pharmaceutically acceptable excipient(s). Said inhibition has been studied and observed by plaque reduction assay.
  • said method is an in-vitro method.
  • said method is an ex- vivo method.
  • the present disclosure provides an in-vitro method of inhibiting survival and/or propagation of live Coronavirus in a host cell, said method comprising the step of contacting the cell with a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) as per any of the above-mentioned embodiments.
  • concentration of components in the composition in each of the above embodiments are the same as that defined for the composition as defined in the initial part of the specification and repetition has been avoided for reasons of brevity.
  • ACE2 cell surface angiotensin-converting enzyme 2
  • the composition of the present disclosure blocks binding of coronavirus Spike protein to the ACE receptor by binding to the ACE2 receptor. In some embodiments, the composition binds to ACE2 receptor through hydrogen bond interactions.
  • composition of the present disclosure renders it effective against different variants and/or mutants of coronaviruses such as but not limited to SARS-CoV2, since the target of the composition is on the host cell which remains unchanged despite the mutations in the virus.
  • binding of the composition to the ACE2 receptor facilitates inhibition of the ACE2-Spike protein complex formation at the entry level across different variants and/or mutants of coronaviruses.
  • the spike protein as referred to above is selected from a group comprising Spike 1 and Spike 2 protein or a combination thereof.
  • the composition of the present disclosure blocks entry of the virus into a host cell by binding to the receptor binding domain (RBD) of the coronavirus. In some embodiments, the composition binds to the RBD through hydrogen bond interactions.
  • RBD receptor binding domain
  • the present disclosure provides a method of inhibiting formation of a complex between ACE2 of a cell and Spike protein of Coronavirus by contacting the cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally in combination with pharmaceutically acceptable excipient(s).
  • the above method is facilitated in-vivo. In some embodiments, the above method is an in-vitro method. In some embodiments, said method is an ex-vivo method.
  • the present disclosure provides an in-vitro method of inhibiting formation of a complex between ACE2 of a cell and Spike protein of Coronavirus comprising contacting the cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally in combination with pharmaceutically acceptable excipient(s).
  • the composition employed in the above methods comprises concentration of pentameric type A procyanidin, tetrameric type A procyanidin, and trimeric type A procyanidin as defined in the embodiments that define the composition and said information has not been repeated herein for reasons of brevity.
  • Coronavirus infections commonly manifest in pulmonary complications. Excess inflammation is hall mark of this infection caused by recruitment of Neutrophils to Lungs. It is observed that in severe Corona infections such as COVID-19 infections caused by SARS-CoV-2, there is a dysfunctional delayed excess inflammation causing damage to lungs causing severe hypoxia and microthrombus in the capillaries of lung.
  • the present disclosure provides a method of preventing, managing and/or treating Coronavirus infection and/or associated complications comprising administering the composition of the present disclosure to a subject in need thereof, at a pharmaceutically effective dosage.
  • the present disclosure provides a method of preventing, managing and/or treating Coronavirus infection and/or associated complications comprising step of administering composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient, to a subject in need thereof.
  • the present disclosure provides a method of preventing Coronavirus infection and/or associated complications comprising administering the composition of the present disclosure comprising pentameric type A, trimeric type A procyanidins and tetrameric type A procyanidins, optionally along with pharmaceutically acceptable excipients to a subject in need thereof, at a pharmaceutically effective dosage.
  • the present disclosure provides a method of treating and/or managing Coronavirus infection and/or associated complications comprising administering a composition comprising pentameric type A procyanidins, trimeric type A procyanidins and tetrameric type A procyanidins, optionally along with pharmaceutically acceptable excipients to a subject in need thereof, at a pharmaceutically effective dosage.
  • the composition employed for the treatment, management and/or prevention of Coronavirus infections or for the preparation of a medicament for the treatment, management and/or prevention of Coronavirus infections comprises concentration of components as defined in any of the above embodiments defining the composition and said information has not been repeated for the reasons of brevity.
  • the composition is administered to the subject before entry of virus particles i.e. before infection. In some embodiments, the composition is administered to the subject after entry of virus particles i.e. after infection.
  • the pharmaceutically effective amount of the composition ranges from about 1 mg/kg to about 100 mg/kg body weight of the subject.
  • the application of the composition in the prevention and/or treatment or management of infections caused by viruses of the Coronavirus family such as but not limited to SARS-CoV-2 and mutants or variants thereof.
  • the Coronavirus infection and/or associated complications is treated, managed and/or prevented by administration of the composition of the present disclosure as per any of the above embodiments, in pharmaceutically acceptable amounts, by any commonly adopted route(s) of administration such as but not limited to oral, intravenous, sublingual, buccal, nasal, ocular, topical, otic, rectal and vaginal routes.
  • a particularly preferred route of administration is the nasal route of administration.
  • the aforesaid methods employ the composition formulated as a nasal spray or inhaler.
  • the aforesaid method may be carried out in combination with other treatments for coronavirus infection.
  • the above defined methods of the present disclosure inhibits the survival and propagation of live Coronavirus at the cellular level and thus shows anti-viral efficacy.
  • the median time required for a negative RT-PCR post treatment ranges between about 13 days to about 28 days.
  • the aforesaid method improves oxygen saturation in an infected subject to a value from about 89% to about 100%.
  • the aforesaid method(s) improves the quality of life of the subject, wherein the subjects include but are not limited to non-hospitalized and hospitalized covid patients.
  • hospitalized patients are discharged in 9-10 days, therefore showing the reduction in severity of the infection.
  • the present disclosure further provides a kit comprising the composition as per any of the aforementioned embodiments, along with a pamphlet or brochure indicating suitable dosage of the composition.
  • the present disclosure provides a kit comprising the composition composed pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin or any combination thereof optionally in combination with pharmaceutically acceptable excipient(s), along with a pamphlet or brochure indicating suitable dosage of the composition.
  • the kit further comprises means for administration of the composition such as but not limited to inhalers, pumps and nebulizers.
  • means for administration of the composition such as but not limited to inhalers, pumps and nebulizers.
  • the extract was filtered through a two-stage chromatographic column to obtain the composition having about 80% of Type A procyanidin pentamer of flavanoid of molecular weight.
  • the extract was passed through the first column to extract the relatively less polar molecules of the composition and the second stage of chromatographic separation was for the relatively more polar molecules of the composition.
  • the resins used were the equivalent of an XAD-1180 and an XAD-7HP resin respectively.
  • the column was thoroughly washed with D.M. water free of adhering substances and the eluent is neutral.
  • the column was further eluted with about 175 ml pure iso propyl alcohol and the collected eluent was concentrated under vacuum below about 40° C. and diluted with water and spray dried under following conditions:
  • Inlet temperature 140° C.
  • the final weight was 5gms.
  • composition comprising type A pentameric procyanidin flavonoid of concentration ranging from about 55 % w/w to about 99% w/w, trimers and tetramers of procyanidin flavonoid each at concentration ranging from about 0.5 % w/w to about 35 % w/w as obtained above was formulated into capsules by blending with about 2% w/w of micro crystalline cellulose, about 0.5% w/w of crospovidone and about 0.2% w/w of magnesium stearate. This mixture was filled in capsules.
  • compositions were prepared by addition of appropriate excipient(s) selected from list comprising: granulating agent, binding agent, lubricating agent, disintegrating agent, sweetening agent, glidant, anti-adherent, anti-static agent, surfactant, antioxidant, gum, coating agent, coloring agent, flavouring agent, coating agent, plasticizer, preservative, suspending agent, emulsifying agent, plant cellulosic material and spheronization agent or any combination thereof.
  • excipient(s) selected from list comprising: granulating agent, binding agent, lubricating agent, disintegrating agent, sweetening agent, glidant, anti-adherent, anti-static agent, surfactant, antioxidant, gum, coating agent, coloring agent, flavouring agent, coating agent, plasticizer, preservative, suspending agent, emulsifying agent, plant cellulosic material and spheronization agent or any combination thereof.
  • the type of formulation was selected from group comprising solid oral formulation, liquid oral formulation, parenteral formulation, aerosols or spray, phytoceutical, nutraceutical and medicinal foods or any combinations thereof.
  • the solid oral formulation was selected from group comprising tablet, capsule, troche, lozenge, dispersible powder, dispersible granule or any combinations thereof.
  • the liquid oral formulation was selected from group comprising aqueous or oily suspension, emulsion, drop, emulsion in hard or soft gel capsule, syrup, elixir or any combinations thereof.
  • the parenteral formulation was selected from group comprising intravenous injection, intramuscular injection, intramuscular depot, subcutaneous injection, percutaneous injection or any combinations thereof.
  • the aerosol or spray based formulations include but are not limited to nasal sprays or inhalers.
  • a suitable formulation of the instant composition for managing coronavirus infection using a daily dose range from about 1 mg/kg to about 100 mg/kg of body weight of subject or preferably, a daily dose range from about 10 mg/kg to about 75 mg/kg of body weight of subject.
  • Example 1 About 20 pg/mL of the composition of Example 1 was incubated with spike protein 1 coated wells for about 1 hour. Lung tissue lysates were added into the well and incubated for about 2 hours after wash. The unbound ACE2 was removed by washing and the bound ACE2 was detected by adding human ACE2-specific rabbit antibody and HRP -conjugated anti-rabbit IgG antibody. The reaction was then visualized by adding substrate and quenched by adding H2SO4. The binding was measured by an ELISA reader (OD450).
  • OD450 ELISA reader
  • composition was found to reduce the binding of ACE2 to COVID spike 1 protein by about 9% (Figure 1).
  • Figure 2 the standard curve of ACE2 binding with coated spike 1 protein was showed in Figure 2.
  • Example 1 the composition of Example 1 at different concentrations (5, 10, 20, 40, 80, 100, 150, 200 pg/mL) was added into recombinant ACE2 coated 96-wells and incubated for about 30 minutes. After wash, spike 1 protein (Figure 3, 1.0 pg/well; Figure 4, 0.1 pg/well) was added into the well and incubated at about 25 °C for about 1.5 hours. The unbound spike 1 protein was removed by washing and the bound spike 1 protein was detected by adding goat anti-sheep Fc antibody for about 2 hours, followed by three times of wash and incubated with HRP -conjugated anti-goat IgG antibody for about 1 hour.
  • spike 1 protein Figure 3, 1.0 pg/well; Figure 4, 0.1 pg/well
  • the reaction was then visualized by adding substrate and quenched by adding H2SO4.
  • the binding was measured by an ELISA reader (OD450).
  • the calculated ICso of the composition to ACE2-spike 1 protein interaction were about 21.82 pg/mL and about 9.949 pg/mL, respectively.
  • composition of the present disclosure composed of a combination of pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin inhibited the binding of the ACE2 receptor to the spike protein.
  • EXAMPLE 3 Binding kinetics and affinity (KD) of compositions of the present disclosure to human ACE2
  • the ACE2-His at about 10 pg/mL in running buffer was immobilized onto Ni-NT A biosensors, and then incubated with serial dilutions of the composition (about 33.3 pg/mL, about 11.1 pg/mL, about 3.7 pg/mL, about 1.2 pg/mL, about 0.4 pg/mL) in running buffer.
  • the curves were fitted by a 1: 1 binding model using ForteBio Data Analysis software 10.0 ( Figure 7).
  • the KD of the composition of Example 1 for ACE-2 was about 0.65 pM.
  • composition of the present disclosure showed significant binding efficacy to ACE2.
  • EXAMPLE 4 Effect of the composition on interaction of coronavirus RBD with ACE2 in a competition binding assay
  • the recombinant receptor binding domain (RBD) sequence encoding amino acids R319- F541 of SARS-CoV -2 was cloned into pSecTag2B expression vector with an additional C terminal Fc fragment of human IgGl and AviTag in tandem and transiently expressed in Expi293 HEK cells as a secreted protein. After 3 days, supernatants were harvested by centrifuging the culture at about 2500 g for about 15 minutes and filtering the supernatant with a 0.22 mm vacuum filter. The protein was purified using protein G resin (GE healthcare).
  • biotinylated proteins used in bio-panning and BLI experiment were prepared by the BirA biotin-protein ligase in PBS for about 30 minutes at about 30 °C, which adds biotin covalently to AviTag in a highly specific manner, according to the manufacturer's instructions (Avidity).
  • Binding kinetics and affinity (Kp) of the composition to SARS-CoV-2 RBD Binding kinetics and affinity (Kp) of the composition to SARS-CoV-2 RBD
  • the biotinylated RBD-Fc of SARS-CoV-2 at about 10 pg/mL in running buffer was immobilized onto streptavidin-coated biosensors, and then incubated with serial dilutions of the compositions of Example 1 and 3 (about 33.3 pg/mL, about 11.1 pg/mL, about 3.7 pg/mL, about 1.2 pg/mL) in running buffer.
  • KD of the composition of Example 1 for SARS-CoV-2 RBD was about 1.43 pM.
  • composition of the present disclosure effectively binds to SARS- CoV-2 RBD.
  • EXAMPLE 5 Neutralizing activity of the composition against pseudotyped SARS-CoV- 2 infection
  • the neutralization activity of the Example 1 composition was measured with a SARS-CoV-2 spike (S) protein-mediated, luciferase -encoding pseudovirus neutralizing assay.
  • S SARS-CoV-2 spike
  • 293 T cells were cotransfected with expression vectors of pcDNA3.
  • l-SARS-CoV-2-S encoding SARS-CoV-2 S protein
  • pNL4-3.luc.RE bearing the luciferase reporter-expressing HIV -1 backbone.
  • the supernatants containing SARS-CoV-2 pseudotyped virus were harvested.
  • Serial dilutions of single-domain antibodies in DMEM supplemented with 10% fetal calf serum were incubated with pseudo viruses at 37°C for 1 h and then the mixtures were added to monolayer Huh-7 cells (104 per well in 96-well plates). Twelve hours after infection, culture medium was refreshed and then incubated for an additional 48 hours.
  • the luciferase activity was calculated for the detection of relative light units using the Bright-GloTM Luciferase Assay System (Promega). A nonlinear regression analysis was performed on the resulting curves using Prism (GraphPad) to calculate half-maximal inhibitory concentration (IC50) values ( Figure 7).
  • the composition showed potent neutralization activity, inhibiting SARS-CoV-2 pseudovirus infection with a half-maximal inhibitory concentration (ICso) values of about 9.232 pg/mL.
  • ICso half-maximal inhibitory concentration
  • Huh-7 cells in the logarithmic phase of growth were collected and seeded in 96-well plates at a density of about 1 x 10 4 cells/well and cultured overnight.
  • the culture medium was removed and serial dilution of the composition of claim 1 was added into wells of 96-well plates in 200pl medium. Cells were treated for about 48 hours at about 37 °C. After treatment, the culture medium was removed and lOOpl CCK8 solution (lOpl CCK8 + 90pl medium) (Dojindo Molecular Technologies, Gaithersburg, MD) was added into each well, then incubated at 37°C for 2-4 h. Absorbance was measured at 450nm using a 96-well microplate reader. Each concentration was repeated in triplicate.
  • Plaque assay was performed in triplicate in 24-well tissue culture plates.
  • the VeroE6 cells were seeded in DMEM with 10% FCS and antibiotics one day before infection.
  • SARS-CoV-2 was added to the cell monolayer for 1 hour at 37°C.
  • viruses were removed and the cell monolayer was washed once with PBS before covering with media containing agarose or methylcellulose for 5-7 days.
  • the cells were fixed with formaldehyde overnight. After removal of overlay media, the cells were stained with crystal violet and the plaques were counted.
  • the percentage of inhibition was calculated as [1 -(VD/ VC)] X 100%, where VD and Vc refer to the virus titer in the presence and absence of the compound, respectively.
  • Example 1 could inhibit SARS-CoV-2 efficiently.
  • ICso inhibition concentration
  • Figure 8C The cell toxicity of the compositions was also determined by ACP assay and the cell cytotoxicity values for the composition was estimated to be more than 80 pg/mL ( Figure 8C).
  • the derived SI index for the composition of Example 1 was >22.1.
  • Vero E6 cells were seeded to the 24-well culture plate in DMEM with 10% FBS and antibiotics one day before infection. Vero E6 cells were infected by SARS-CoV-2 virus (100 pfu/well) for about 1 hour at about 37°C. After removal of virus inoculum, the cells were washed once with PBS and overlaid with 1 mL overlay medium containing 1% methylcellulose for 5 days at 37°C. After about 5 days, the cells were fixed with about 10% formalin overnight. After removal of overlay media, the cells were stained with about 0.5% crystal violet and the plaques were counted. For “Entry” treatment, the cells were first pre-treated with the compound for about 1 hour, then infected with virus in the presence of the compound.
  • the compound was added to the cells at 1 hpi (Hours post infection), and maintained in the overlay medium until the end of the experiment.
  • the percentage of inhibition was calculated as [ 1 -(VD / Vc)] x W0%, where VD and Vc refer to the virus titer in the presence and absence of the composition, respectively.
  • the minimal concentration of composition required to reduce 50% of plaque numbers (ECso) was calculated by regression analysis of the dose-response curves generated from plaque assays. To elucidate the mechanism of action of the composition of Example 1 on SARS-CoV2, time of addition assays were performed.
  • the cells were first pre-treated with the compound for about 1 hour, followed by infection of the virus in the presence of the compound.
  • the composition was added to the cells at 1-hour post infection (hpi).
  • the composition showed inhibition of SARS CoV2 mainly at the entry level, and also some inhibitory effect at the post entry stage of the virus infection (Figure 9).
  • Example 1 This was an interventional, randomized, double-blind, placebo-controlled clinical study to evaluate the safety and effectiveness of the composition of Example 1, formulated as capsules, in preventing the progression of disease severity in COVID-19 patients.
  • test composition placebo capsules (1:1) for about 15 days in two regimens, along with standard of care treatment: For initial 3 days: the composition or placebo was administered as about 3 g/day (2 capsules of 500 mg X 3 times) as a loading dose.
  • test composition or placebo was administered as 1.5 g/day (1 capsule of 500 mg X 3 times) as a maintenance dose.
  • the screening and enrolment procedures were done at the study centre if the patient was being treated either in hospital or quarantine setting.
  • a study team member performed a home visit during the time of randomization of the patient into the study, to ensure that all study related procedures were completed appropriately.
  • the other study visits were performed either at the study centre (if the patient was hospitalized) or the study team needed to perform cither a home or telephonic visit (if the patient was at home) to ensure study-related activities were completed.
  • everyone involved followed social distancing to the extent possible and wore masks throughout the interaction.
  • the treatment period consisted of about 15 days for all the patients: unless the treatment ended earlier because of withdrawal of consent by the patient or in case the patient progressed to a more severe stage of the disease or in case of death of the patient, or as per the discretion of the investigator.
  • the primary endpoint of the study - time to clinical recovery - defined as the time required to attain a score of either 1 (asymptomatic; viral RNA detected) or 0 (uninfected; no vims detected) on the WHO Clinical Progression Scale was comparable between the composition and Placebo groups with no statistically significant difference.
  • the median (SE) time to recovery for the composition treated group was about 8 (0.678) days, while that for the Placebo group was about 7 (0.419) days.
  • the treatment group was associated with significant improvement in the mean total score of WHO-QOL-BREF Questionnaire from Day I to EOT, comparable to the Placebo group.
  • the mean scores for the physical health domain were significantly higher on Day 8 and Day 14 for the composition treated group as compared to the placebo group.
  • the mean scores for the psychological health domain were higher in the composition treated group as compared to the Placebo group at each day.
  • the mean scores for the social relationship domain were significantly higher on day 6 as compared to the Placebo group, and continued to be higher for all other days.
  • the mean scores for the environmental domain were significantly higher on days 6 to 9. day 11. and EOT in the composition treated group as compared to the Placebo group.
  • Example 1 Overall, the investigational composition of Example 1 was reported to have the potential of therapeutic activity in patients with mild to moderate covid- 19 infection.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Molecular Biology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Communicable Diseases (AREA)
  • Engineering & Computer Science (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Biotechnology (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present disclosure provides a composition comprising procyanidins selected from a group comprising pentameric procyanidin, tetrameric procyanidin and trimeric procyanidin or any combination thereof, optionally along with pharmaceutically acceptable excipient(s) for use in preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell, and in the management, treatment and/or prevention of diseases or infections caused by the different strains of Coronavirus including SARS-CoV2 and mutants and/or variants thereof. The present disclosure thus also provides a method for the management, treatment and/or prevention of Coronavirus infections and/or associated complications. The composition reduces severity of coronavirus infections and significantly improves oxygen saturation levels in subjects.

Description

COMPOSITION AND METHOD FOR MANAGING CORONAVIRUS INFECTION
TECHNICAL FIELD
The present disclosure relates to the field of immunology, more specifically, to the field of virology, biotechnology and medicine. Particularly, the present disclosure provides a botanical composition effective against viruses such as but not limited to strains of Coronavirus and mutants or variants thereof. More particularly, the present disclosure provides a composition comprising procyanidins selected from a group comprising pentameric procyanidin, tetrameric procyanidin and trimeric procyanidin or any combination thereof, optionally along with pharmaceutically acceptable excipient(s) for use in the management, treatment and/or prevention of diseases or infections caused by the different strains of Coronavirus and complications arising from such infections. The present disclosure thus also provides a method for the management, treatment and/or prevention of Coronavirus infections and/or associated complications.
BACKGROUND OF THE DISCLOSURE
Catechins are polyphenolic plant metabolites which belong to the flavonoid family. The molecular formula and weight of catechins are C isH Or, and 290 g/mol, respectively. Catechin and epicatechin are epimers, with (-)-epicatechin and (+)-catechin being the most common optical isomers found in nature. Procyanidins or condensed tannins are flavonoid oligomers whose building blocks are (+) - catechin and (-) - epicatechin. They are present abundantly in the plant kingdom in fruits, barks, leaves and seeds where they provide protection against light, oxidation and predators. Procyanidins are found in many plants, mainly apples, pine bark, cinnamon bark, litchi pericarp, peanuts, grape seed, cocoa, grape skin, bilberry, cranberry, black currant, green tea and black tea.
Based on the linkage between the successive monomeric units, procyanidins are classified as Types A, B or C polyphenols. Generally, the linkage between successive monomeric units of procyanidins is between the 4th position of the ‘upper’ unit and the 8th position of the ‘lower’ unit, leading to a Type B procyanidin. Alternatively, the linkage can occur between C4 of the ‘upper’ unit and C6 of the lower unit, leading to a Type C procyanidin. Type B and C polyphenols are abundantly seen in many botanical sources. When successive monomeric units are linked by an ether linkage between the C2 and C4 of the ‘upper’ unit and the oxygen at the C7 position and the C6/C8 positions (respectively) of the lower unit, a Type A procyanidin is formed.
Coronaviruses are a group of related viruses known to cause diseases in mammals and birds. Infection by said viruses, in humans, is known to lead to respiratory tract infections that can range from mild to lethal.
Coronavirus infections in the form of SARS, and COVID-19 have had alarming implications. Coronavirus disease 2019 (COVID-19) which took the form of a pandemic in early 2020 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Coronavirus infects cells through its spike protein which binds to specific cell receptors of the host particularly ACE2. The virus is known to cause lung pathology, sepsis and renal failure. A robust cure/treatment regime for Coronavirus is however yet to be found.
Signs and symptoms of coronavirus disease 2019 (COVID-19) may appear 2 to 14 days after exposure. This time after exposure and before having symptoms is called the incubation period. The subject can still spread COVID- 19 before he has symptoms (Pre-symptomatic transmission).
Common signs and symptoms are Fever, Cough, Tiredness, Shortness of breath or difficulty breathing, Muscle aches, Chills, Sore throat, Runny nose, Headache, Chest pain, Pink eye (conjunctivitis), Nausea, Vomiting, and Diarrhea. However early symptoms of COVID- 19 may include a loss of taste or smell.
People who are older have a higher risk of serious illness from COVID-19, and the risk increases with age. People who have existing medical conditions also may have a higher risk of serious illness. Certain medical conditions that may increase the risk of serious illness from COVID- 19 may be Serious heart diseases, such as heart failure, coronary artery disease or cardiomyopathy, Chronic obstructive pulmonary disease (COPD), Type 1 or type 2 diabetes, Overweight, obesity or severe obesity, High blood pressure and Asthma.
The severity of COVID-19 symptoms can range from very mild to severe. Some people may have only a few or mild symptoms. Some people may experience more severe symptoms, such as worsened shortness of breath and pneumonia, about a week after symptoms start. Chronic lung diseases such as cystic fibrosis or pulmonary hypertension. Although most people with COVID-19 have mild to moderate symptoms, the disease can cause severe medical complications and lead to death in some people. Older adults or people with existing medical conditions are at greater risk of becoming seriously ill with COVID-19. The Complications can include Pneumonia and trouble breathing, Organ failure in several organs, cardiac problems and blood clots. And a severe lung condition that causes a low amount of oxygen to go through your bloodstream to your organs (acute respiratory distress syndrome).
Major symptom leading to disease severity is decline in Oxygen Saturation (SPO2) due to the lung pathology of the disease.
Also, it is seen that the viral persistence carries even after the symptoms goes by and the syndrome is called Long Covid with symptoms such as fatigue and decrease in quality of life.
The U.S. Food and Drug Administration (FDA) has given emergency use authorization to some COVID- 19 vaccines in the United States. The FDA has approved the Pfizer-BioNTech COVID- 19 vaccine, now called Comimaty, to prevent COVID-19 in people age 16 and older. The FDA has given emergency use authorization to Pfizer-BioNTech COVID- 19 vaccines for ages 5 through 15.
The CDC recommends additional doses and booster doses of COVID- 19 vaccines in specific instances for some people with weakened immune systems, such as those who have had an organ transplant. People with weakened immune systems might not develop enough immunity after vaccination with two doses of an mRNA COVID-19 vaccine. An additional dose might improve their protection against COVID- 19. Given the high rate at which the Coronavirus infections spread, and the severe complications caused by the disease such as difficulty in breathing, pneumonia, blood clot formation and severe lung infection, there exists a need for a compositions and methods exclusively directed towards management of the infections caused by different strains of the Coronavirus by targeting the underlying cause, and alongside management of the symptoms.
Coronavirus entry into the host cell is mediated by a completely different process compared to HIV and H1N1. The spike protein S has a Receptor Binding Domain (RBD). This binds to the Angiotensin Converting Enzyme 2 (ACE2) expressed on the host cell. A protease enzyme expressed near the ACE2 (TMPRSS2 and/or Furin, depending on the cell) cleaves the spike protein and facilitates the fusion of the virus into the host cell. Given said difference in mechanism of infection, antiviral drugs known for treatment of HIV and H IN 1 are not effective in preventing and/or treating Coronavirus infections.
The present invention addresses the need for a drug and a method directed towards Coronavirus infections and provides a composition and a method that targets the infection mechanism of the virus to inhibit the entry of the virus, improves oxygen saturation levels, prevents further progression of the disease by blocking the entry of the virus and improves quality of life of people suffering from the disease.
STATEMENT OF THE DISCLOSURE
In order to address the need in the art for a composition and a method directed towards the treatment/management and/or prevention of Coronavirus infection and/or associated complications, the present disclosure provides a suitable composition fit for said application and methods employing the same.
Accordingly, the present invention provides a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in preventing, managing or treating Coronavirus infection and/or associated complication(s).
In some embodiments, the Coronavirus infection is caused by SARS-CoV-2 or mutants or variants thereof.
In some embodiments, the associated complication is selected from a group comprising reduction in quality of life, reduction in oxygen saturation (SpCh) levels, pneumonia, troubled breathing, blood clot formation, cardiac problems and organ failure or any combination thereof.
Preferably, the associated complication is selected from a group comprising reduction in quality of life and/or reduction in oxygen saturation (SpCh) levels.
In some embodiments, in the above referred composition, the pentameric type A procyanidin is at concentration ranging from about 55 % w/w to about 99% w/w, the tetrameric type A procyanidin and the trimeric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 35 % w/w; and the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
In some embodiments, in the above referred composition, the pentameric type A procyanidin is at concentration ranging from about 80 % w/w to about 90% w/w, the tetrameric type A procyanidin and the trimeric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 20 % w/w; and the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
In some embodiments, the pharmaceutically acceptable excipient(s) as referred to above, is selected from a group comprising gums, granulating agents, binding agents, lubricating agents, disintegrating agents, sweetening agents, coloring agents, flavoring agents, coating agents, plasticizers, preservatives, suspending agents, emulsifying agents, antistatic agents, and spheronization agents or any combination thereof.
In some embodiments, the composition is formulated into dosage forms selected from group comprising solid formulation, liquid formulation, parenteral formulation, aerosol or spray, phytoceutical, nutraceutical and medicinal foods or any combinations thereof.
In some embodiments, the solid oral formulation is selected from group comprising tablet, capsule, troche, lozenge, dispersible powder, dispersible granule or any combinations thereof; the liquid oral formulation is selected from group comprising aqueous or oily suspension, emulsion, drops, emulsion in hard or soft gel capsule, syrup, elixir or any combinations thereof; the parenteral formulation is selected from group comprising intravenous injection, intramuscular injection, intramuscular depot, subcutaneous injection, percutaneous injection or any combinations thereof; and the aerosol formulation is selected from a group comprising oral spray, nasal spray and inhaler.
In exemplary embodiments, the composition is formulated into a nasal spray.
In some embodiments, the aforesaid use is as a medicament for preventing, managing, or treating Coronavirus infection and/or associated complication(s). Further provided herein is a method for preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell, comprising contacting the host cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s).
In some embodiments, the composition inhibits formation of a complex between ACE2 receptor of a host cell and Spike protein of the Coronavirus.
In some embodiments, the composition binds to the ACE2 receptor of the host cell.
In some embodiments, the composition binds to Receptor Binding Domain (RBD) of spike protein of the Coronavirus.
Also envisaged herein is a method of preventing, treating and/or managing Coronavirus infection and/or associated complication(s) comprising administering a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) to a subject in need thereof, at a pharmaceutically effective dosage.
In some embodiments, the pharmaceutically effective dosage ranges from about 1 mg/kg to about 100 mg/kg body weight of the subject.
In some embodiments, the subject is a mammal, including human beings.
In some embodiments, the method improves oxygen saturation in an infected subject to a value from about 89% to about 100%.
Further provided herein is a kit comprising a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s), along with a pamphlet or brochure indicating suitable dosage of the composition. In some embodiments, the kit further comprises means for administration of the composition.
BRIEF DESCRIPTION OF THE ACCOMPANYING FIGURES
In order that the disclosure may be readily understood and put into practical effect, reference will now be made to exemplary embodiments as illustrated with reference to the accompanying figures. The figures together with detailed description below, are incorporated in and form part of the specification, and serve to further illustrate the embodiments and explain various principles and advantages, where:
Figure 1 depicts the reduction in binding of ACE2 receptor of host cell (hereinafter referred to as ACE2) to COVID spike 1 protein caused by the composition of the present disclosure.
Figure 2 depicts the standard curve of ACE2 binding with coated spike 1 protein.
Figure 3 depicts blocking of the ACE2-spike 1 protein interaction by the composition of the present disclosure.
Figure 4 depicts blocking of the ACE2-spike 1 protein interaction by the composition of the present disclosure
Figure 5 depicts binding capacity of the composition of the present disclosure to ACE2 receptor as measured by Bio-Layer Interferometry (BLI) using Octet Red 96.
Figure 6 depicts binding capacity of the composition of the present disclosure to SARS-CoV2 RBD as measured by Bio-Layer Interferometry (BLI) using Octet Red 96.
Figure 7 depicts neutralization of pseudotyped virus with spike protein of SARS-CoV-2 by the composition of the present disclosure. The dotted line indicates the 50% inhibition rate. Data are shown as mean± SD (error bar) from three different experiments.
Figure 8 depicts inhibitory activity of the composition against authentic SARS-CoV-2 as observed in a plaque-reduction assay A) depicts observation in the plaque inhibition assay; B) depicts analysis of IC50 of the composition; C) depicts cell cytotoxicity analysis of the composition .
Figure 9 depicts inhibitory activity of the composition against SARS-CoV-2 at the stage of viral entry and post viral entry A) depicts observation in the plaque inhibition assay; B) depicts % reduction as observed in the plaque reduction assay. DETAILED DESCRIPTION OF THE DISCLOSURE
Coronaviruses are typically characterized by the presence of an outer envelope, wherein said envelope consists of a lipid bilayer. Membrane, envelope and spike proteins are embedded within said lipid bilayer. As mentioned above, the mechanism of viral entry into the host cell is primarily governed by spike proteins, wherein the viral membrane fuses with the cell membrane of the host upon binding of the spike protein the host cell receptor. Once said fusion occurs, it leads to entry of the viral genome into the host cells which causes the infection.
In view of the limitations discussed above, and to remedy the requirement in the art for compositions and methods directed towards management of Coronavirus infections, the present disclosure provides a composition fit for application in the management of said infections. The present disclosure further provides a method for the management of infections caused by Coronavirus.
However, before providing a detailed description of the invention, the below paragraphs provide definitions of terms regularly used in the present disclosure for the purposes of clarity and ease of understanding.
The particulars shown herein are for purposes of illustrative discussion of the preferred embodiments of the present invention only and are presented in the cause of providing what is believed to be the most useful and readily understood description of the principles and conceptual aspects of various embodiments of the invention. In this regard, no attempt is made to show structural details of the invention in more detail than is necessary for the fundamental understanding of the invention, the description making apparent to those skilled in the art how the several forms of the invention may be embodied in practice.
The following definitions and explanations are meant and intended to be controlling in any future construction unless clearly and unambiguously modified in the following examples or when application of the meaning renders any construction meaningless or essentially meaningless. In cases where the construction of the term would render it meaningless or essentially meaningless, the definition should be taken from Webster’s Dictionary, 3rd Edition or a dictionary known to those of skill in the art, such as the Oxford Dictionary of Biochemistry and Molecular Biology (Ed. Anthony Smith, Oxford University Press, Oxford, 2004). The use of the term “or” in the claims is used to mean “and/or” unless expressly stated to refer only to alternatives, or alternatives are mutually exclusive.
As used herein and in the claims, the terms “include” (any form of “include”, such as “include”), “have” (and “have”, etc. Any form of “having”, “including” (and any form of “including” such as “including”), or “including” (and “including” Any form of “containing” is inclusive or variable and does not exclude further uncited elements or method steps.
With respect to the use of substantially any plural and/or singular terms herein, unless otherwise required by context, singular terms used herein shall include pluralities and plural terms shall include the singular. The various singular/plural permutations may be expressly set forth herein for sake of clarity. As used herein and unless otherwise indicated, the terms “a” and “an” are taken to mean “one”, “at least one” or “one or more”. The use of the expression “at least” or “at least one” suggests the use of one or more elements or ingredients or quantities, as the use may be in the embodiment of the disclosure to achieve one or more of the desired objects or results. Throughout this specification, the word “comprise”, or variations such as “comprises” or “comprising” wherever used, will be understood to imply the inclusion of a stated element, integer or step, or group of elements, integers or steps, but not the exclusion of any other element, integer or step, or group of elements, integers or steps. Further, use of the term ‘about’ before values defined in the present disclosure envisages values of about ±10%. Furthermore, ranges defined in the present disclosure provide basis for employment of any specific value or narrower range lying within said defined range, including the extreme values.
As used herein, reference to ‘subject’ throughout the present disclosure, unless otherwise mentioned, implies reference to a subject having or suspected of having Coronavirus infection i.e. a subject infected or suspected to be infected with Coronavirus. The subject may be mammal such as an animal or a human being.
As used throughout the present disclosure, reference to ‘composition’, implies reference to the composition of the present disclosure as per any of the embodiments defining the composition, in all possible permutations and combinations between each of said embodiments.
As used herein, reference to ‘Coronavirus’ throughout the present disclosure envisages reference to any virus/viral strain belonging to the family of Coronavirus and all possible mutants thereof. In a non-limiting embodiment, the coronavirus is selected from a group comprising 229E (alpha coronavirus), NL63 (alpha coronavirus), OC43 (beta coronavirus), HKU 1 (beta coronavirus), MERS-CoV, SARS-CoV. SARS-CoV-2 and any mutants or variants thereof. In an embodiment, Coronavirus infection, in the context of the present invention, may be simultaneously caused by one or more of the above defined strains or variants of coronavirus.
Reference to ‘coronavirus infection’ in the context of the present disclosure envisages active infection as well as associated complications during or after the infection. Associated complications include but are not limited to reduction in quality of life, reduction in oxygen saturation (SpCh) levels, pneumonia, troubled breathing, blood clot formation, cardiac problems, organ failure and other identified as well as unidentified complications that bear any association with coronavirus infection.
Reference to ‘ACE2’ or ‘ACE2 receptor’ in the present disclosure is with respect to the functional receptor on host cell surfaces through which SARS-CoV-2 enters the host cells.
To provide an overview, the present disclosure relates to the management, mitigation and resolution of infection caused by the Coronavirus family that are typically known to cause systematic acute respiratory syndrome and forms thereof. The present invention provides a composition and its applications in the treatment, management and/or prevention of Coronavirus infection.
Particularly, the present disclosure provides a composition comprising a mixture of procyanidin compounds for preventing, managing or treating Coronavirus infection and associated complications.
In some embodiments, provided herein is a composition comprising pentameric type A procyanidin, trimeric type A procyanidin and tetrameric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in preventing, managing, or treating Coronavirus infection and/or associated complications.
In some embodiments, the Coronavirus infection is caused by SARS-CoV2, including different variants and/or mutants thereof.
Associated complications include those that arise during or after the infection. In some embodiments, associated complications include but are not limited to reduction in quality of life, reduction in oxygen saturation levels, and other identified as well as unidentified complications that bear an association with coronavirus infection. In some embodiments, the associated complications are selected from a group comprising reduction in quality of life, reduction in oxygen saturation levels, pneumonia, troubled breathing, blood clot formation, cardiac problems, organ failure or any combination thereof.
In some embodiments, provided herein is a composition comprising one or more of pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the prevention of Coronavirus infections and/or associated complications.
Further provided herein is a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the treatment and/or management of Coronavirus infections and/or associated complications.
In an embodiment of the present disclosure, the pentameric type A procyanidin is at concentration ranging from about 55 % w/w to about 99% w/w, the trimeric type A procyanidin and the tetrameric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 35 % w/w; and the pharmaceutically acceptable excipient is at concentration ranging from about 0.5% to about 99.9%.
In another embodiment of the present disclosure, the pentameric type A procyanidin is at concentration ranging from about 80 % w/w to about 90% w/w, the trimeric procyanidin and the tetrameric procyanidin are each at concentration ranging from about 0.5 % w/w to about 20 % w/w; and the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
In a non-limiting embodiment, the procyanidins in any of the above defined compositions of the present disclosure, in a non-limiting embodiment, are derived from Cinnamon.
In some embodiments, pharmaceutically acceptable excipients in the above defined composition are selected from a group comprising gums, granulating agents, binding agents, lubricating agents, disintegrating agents, sweetening agents, coloring agents, flavoring agents, coating agents, plasticizers, preservatives, suspending agents, emulsifying agents, antistatic agents, and spheronization agents or any combination thereof. In some embodiments of the present disclosure, the composition is formulated into dosage forms selected from group comprising solid formulation, liquid formulation, parenteral formulation, aerosol or spray, phytoceutical, nutraceutical and medicinal foods or any combinations thereof.
In some embodiments, the solid oral formulation is selected from group comprising tablet, capsule, troche, lozenge, dispersible powder, dispersible granule or any combinations thereof.
In some embodiments, the liquid oral formulation is selected from group comprising aqueous or oily suspension, emulsion, drops, emulsion in hard or soft gel capsule, syrup, elixir, and oral sprays or any combinations thereof.
In some embodiments, the parenteral formulation is selected from group comprising intravenous injection, intramuscular injection, intramuscular depot, subcutaneous injection, percutaneous injection or any combinations thereof.
In some embodiments, the aerosol formulation is selected from a group comprising oral spray, nasal spray and inhaler.
In an exemplary embodiment, the composition is formulated into aerosols or sprays such as but not limited to oral sprays, nasal sprays or inhalers.
Further envisaged herein is use of the aforesaid composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) in the prevention of Coronavirus infections and/or associated complications.
In some embodiments, the present disclosure provides use of the composition comprising the pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) in the treatment and/or management of Coronavirus infections and/or associated complications.
In some embodiments, the use, as referred to above, is as a medicament for the prevention, management or treatment of Coronavirus infections and/or associated complications. Accordingly, the present disclosure further provides the composition, as per any of the above embodiments, for use in the manufacture of medicaments directed towards prevention, management or treatment of Coronavirus infections and/or associated complications.
Accordingly, provided in the present disclosure is a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the manufacture of a medicament for the prevention of Coronavirus infections and/or associated complications.
Further provided herein is a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in the manufacture of a medicament for the treatment and/or management of Coronavirus infections and/or associated complications.
The present disclosure further provides use of a composition pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) for the manufacture of a medicament for prevention, management or treatment of Coronavirus infections and/or associated complications.
In some embodiments, the present disclosure provides use of a composition pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) for the manufacture of a medicament for the prevention of Coronavirus infections and/or associated complications.
In some embodiments, the present disclosure provides use of a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) for the manufacture of a medicament for the management or treatment of Coronavirus infections and/or associated complications.
In some embodiments, each of the above applications of the composition of the present disclosure may be in-vitro, in-vivo or ex-vivo applications.
In some embodiments, the present disclosure further relates to the above-described composition for use in preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell. In some embodiments, said use is an in-vitro use. In some embodiments, said use is an ex-vivo use.
Accordingly, the present disclosure, in some embodiments, provides a composition comprising pentameric type A procyanidin, trimeric procyanidin and tetrameric procyanidin, optionally along with pharmaceutically acceptable excipient for use in preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell in vitro.
In some embodiments, the present disclosure provides a method for preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell, comprising contacting the host cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s). In some embodiments, said method is an in-vitro method. In some embodiments, said method is an ex-vivo method.
The present disclosure provides a method of preventing entry of Coronavirus into a host cell, said method comprising the step of contacting the cell with a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s).
In exemplary embodiments, the present disclosure further provides an in-vitro method of preventing entry of Coronavirus into a host cell, said method comprising the step of contacting the cell with a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s).
The present disclosure also provides a method of inhibiting survival and/or propagation of live Coronavirus in a host cell, said method comprising the step of contacting the cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally in combination with pharmaceutically acceptable excipient(s). Said inhibition has been studied and observed by plaque reduction assay. In some embodiments, said method is an in-vitro method. In some embodiments, said method is an ex- vivo method. In exemplary embodiments, the present disclosure provides an in-vitro method of inhibiting survival and/or propagation of live Coronavirus in a host cell, said method comprising the step of contacting the cell with a composition comprising trimeric type A procyanidin, tetrameric type A procyanidin and pentameric type A procyanidin optionally along with pharmaceutically acceptable excipient(s) as per any of the above-mentioned embodiments.
The concentration of components in the composition in each of the above embodiments are the same as that defined for the composition as defined in the initial part of the specification and repetition has been avoided for reasons of brevity.
The spike proteins of Coronavirus recognize cell surface angiotensin-converting enzyme 2 (ACE2) as its cellular receptor for binding and entry. In silico binding studies show that the composition of the present disclosure has high affinity to ACE2 receptor.
In some embodiments, the composition of the present disclosure blocks binding of coronavirus Spike protein to the ACE receptor by binding to the ACE2 receptor. In some embodiments, the composition binds to ACE2 receptor through hydrogen bond interactions.
Said mechanism of the composition of the present disclosure renders it effective against different variants and/or mutants of coronaviruses such as but not limited to SARS-CoV2, since the target of the composition is on the host cell which remains unchanged despite the mutations in the virus. Thus, binding of the composition to the ACE2 receptor facilitates inhibition of the ACE2-Spike protein complex formation at the entry level across different variants and/or mutants of coronaviruses.
In some embodiments, the spike protein as referred to above is selected from a group comprising Spike 1 and Spike 2 protein or a combination thereof.
In some embodiments, the composition of the present disclosure blocks entry of the virus into a host cell by binding to the receptor binding domain (RBD) of the coronavirus. In some embodiments, the composition binds to the RBD through hydrogen bond interactions.
Accordingly, the present disclosure, in some embodiments, provides a method of inhibiting formation of a complex between ACE2 of a cell and Spike protein of Coronavirus by contacting the cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally in combination with pharmaceutically acceptable excipient(s).
In some embodiments, the above method is facilitated in-vivo. In some embodiments, the above method is an in-vitro method. In some embodiments, said method is an ex-vivo method.
In some embodiments, the present disclosure provides an in-vitro method of inhibiting formation of a complex between ACE2 of a cell and Spike protein of Coronavirus comprising contacting the cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin optionally in combination with pharmaceutically acceptable excipient(s).
In some embodiments, the composition employed in the above methods comprises concentration of pentameric type A procyanidin, tetrameric type A procyanidin, and trimeric type A procyanidin as defined in the embodiments that define the composition and said information has not been repeated herein for reasons of brevity.
Coronavirus infections commonly manifest in pulmonary complications. Excess inflammation is hall mark of this infection caused by recruitment of Neutrophils to Lungs. It is observed that in severe Corona infections such as COVID-19 infections caused by SARS-CoV-2, there is a dysfunctional delayed excess inflammation causing damage to lungs causing severe hypoxia and microthrombus in the capillaries of lung.
Accordingly, the present disclosure provides a method of preventing, managing and/or treating Coronavirus infection and/or associated complications comprising administering the composition of the present disclosure to a subject in need thereof, at a pharmaceutically effective dosage.
Particularly, the present disclosure provides a method of preventing, managing and/or treating Coronavirus infection and/or associated complications comprising step of administering composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient, to a subject in need thereof.
The present disclosure provides a method of preventing Coronavirus infection and/or associated complications comprising administering the composition of the present disclosure comprising pentameric type A, trimeric type A procyanidins and tetrameric type A procyanidins, optionally along with pharmaceutically acceptable excipients to a subject in need thereof, at a pharmaceutically effective dosage.
The present disclosure provides a method of treating and/or managing Coronavirus infection and/or associated complications comprising administering a composition comprising pentameric type A procyanidins, trimeric type A procyanidins and tetrameric type A procyanidins, optionally along with pharmaceutically acceptable excipients to a subject in need thereof, at a pharmaceutically effective dosage.
In some embodiments, the composition employed for the treatment, management and/or prevention of Coronavirus infections or for the preparation of a medicament for the treatment, management and/or prevention of Coronavirus infections, as per the abovementioned embodiments, comprises concentration of components as defined in any of the above embodiments defining the composition and said information has not been repeated for the reasons of brevity.
In some embodiments, the composition is administered to the subject before entry of virus particles i.e. before infection. In some embodiments, the composition is administered to the subject after entry of virus particles i.e. after infection.
In an embodiment, the pharmaceutically effective amount of the composition ranges from about 1 mg/kg to about 100 mg/kg body weight of the subject.
In some embodiments, the application of the composition in the prevention and/or treatment or management of infections caused by viruses of the Coronavirus family such as but not limited to SARS-CoV-2 and mutants or variants thereof.
In some embodiments of the present disclosure, the Coronavirus infection and/or associated complications is treated, managed and/or prevented by administration of the composition of the present disclosure as per any of the above embodiments, in pharmaceutically acceptable amounts, by any commonly adopted route(s) of administration such as but not limited to oral, intravenous, sublingual, buccal, nasal, ocular, topical, otic, rectal and vaginal routes. In a non-limiting embodiment, a particularly preferred route of administration is the nasal route of administration. In some embodiments, the aforesaid methods employ the composition formulated as a nasal spray or inhaler.
In a non-limiting embodiment, the aforesaid method may be carried out in combination with other treatments for coronavirus infection.
The above defined methods of the present disclosure inhibits the survival and propagation of live Coronavirus at the cellular level and thus shows anti-viral efficacy.
In a non-limiting embodiment, the median time required for a negative RT-PCR post treatment ranges between about 13 days to about 28 days.
In another non-limiting embodiment, the aforesaid method improves oxygen saturation in an infected subject to a value from about 89% to about 100%.
In some embodiments, the aforesaid method(s) improves the quality of life of the subject, wherein the subjects include but are not limited to non-hospitalized and hospitalized covid patients.
In exemplary embodiments, hospitalized patients are discharged in 9-10 days, therefore showing the reduction in severity of the infection.
In order to help facilitate the treatment, management and/or prevention of Coronavirus infection and/or associated complications, the present disclosure further provides a kit comprising the composition as per any of the aforementioned embodiments, along with a pamphlet or brochure indicating suitable dosage of the composition.
In some embodiments, the present disclosure provides a kit comprising the composition composed pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin or any combination thereof optionally in combination with pharmaceutically acceptable excipient(s), along with a pamphlet or brochure indicating suitable dosage of the composition.
In some embodiments, the kit further comprises means for administration of the composition such as but not limited to inhalers, pumps and nebulizers. It is to be understood that the foregoing descriptive matter is illustrative of the disclosure and not a limitation. While considerable emphasis has been placed herein on the particular features of this disclosure, it will be appreciated that various modifications can be made, and that many changes can be made in the preferred embodiments without departing from the principles of the disclosure. Those skilled in the art will recognize that the embodiments herein can be practiced with modification within the spirit and scope of the embodiments as described herein. Similarly, additional embodiments and features of the present disclosure will be apparent to one of ordinary skill in art based upon description provided herein.
Descriptions of well-known/conventional methods/steps and techniques are omitted so as to not unnecessarily obscure the embodiments herein. Further, the disclosure herein provides for examples illustrating the above-described embodiments, and in order to illustrate the embodiments of the present disclosure certain aspects have been employed. The examples used herein for such illustration are intended merely to facilitate an understanding of ways in which the embodiments herein may be practiced and to further enable those of skill in the art to practice the embodiments herein. Accordingly, the following examples should not be construed as limiting the scope of the embodiments herein.
EXAMPLES
EXAMPLE 1 - Preparation of the composition comprising the pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin
1000 gms of pulverized cinnamon powder (prepared using Cinnamon sourced from Sri Lanka) with an average size ranging from 16 mesh size was soaked in 3000 ml of ethyl acetate and poured into an extractor havin g a perforated bottom sieve of the 200 mesh sieve. The bottom eluent was recycled again and again over the packed mass to achieve effective extraction for a period of about 8 hrs. The eluent was discarded and the mass was removed out of the extractor and dried in a forced draft oven at about 30°C. After removal of solvent by drying, the mass was again packed in the extractor. The packed mass was extracted with about 5000 ml of acidified deionised water at pH of about 4.0 and the extract was recycled over the bed for about 8 hrs at about 35° C. to achieve efficient extraction.
The extract was filtered through a two-stage chromatographic column to obtain the composition having about 80% of Type A procyanidin pentamer of flavanoid of molecular weight. The extract was passed through the first column to extract the relatively less polar molecules of the composition and the second stage of chromatographic separation was for the relatively more polar molecules of the composition. The resins used were the equivalent of an XAD-1180 and an XAD-7HP resin respectively. The column was thoroughly washed with D.M. water free of adhering substances and the eluent is neutral. The column was further eluted with about 175 ml pure iso propyl alcohol and the collected eluent was concentrated under vacuum below about 40° C. and diluted with water and spray dried under following conditions:
Spray drier: Co current airflow
Inlet temperature: 140° C.
Outlet temperature 60° C.
Atomizer RPM 14000
The final weight was 5gms.
Preparing the formulation -
The composition comprising type A pentameric procyanidin flavonoid of concentration ranging from about 55 % w/w to about 99% w/w, trimers and tetramers of procyanidin flavonoid each at concentration ranging from about 0.5 % w/w to about 35 % w/w as obtained above was formulated into capsules by blending with about 2% w/w of micro crystalline cellulose, about 0.5% w/w of crospovidone and about 0.2% w/w of magnesium stearate. This mixture was filled in capsules.
Similar formulations of the instant composition were prepared by addition of appropriate excipient(s) selected from list comprising: granulating agent, binding agent, lubricating agent, disintegrating agent, sweetening agent, glidant, anti-adherent, anti-static agent, surfactant, antioxidant, gum, coating agent, coloring agent, flavouring agent, coating agent, plasticizer, preservative, suspending agent, emulsifying agent, plant cellulosic material and spheronization agent or any combination thereof.
The type of formulation was selected from group comprising solid oral formulation, liquid oral formulation, parenteral formulation, aerosols or spray, phytoceutical, nutraceutical and medicinal foods or any combinations thereof. The solid oral formulation was selected from group comprising tablet, capsule, troche, lozenge, dispersible powder, dispersible granule or any combinations thereof. The liquid oral formulation was selected from group comprising aqueous or oily suspension, emulsion, drop, emulsion in hard or soft gel capsule, syrup, elixir or any combinations thereof. The parenteral formulation was selected from group comprising intravenous injection, intramuscular injection, intramuscular depot, subcutaneous injection, percutaneous injection or any combinations thereof. The aerosol or spray based formulations include but are not limited to nasal sprays or inhalers.
Depending on the route of administration, different excipients/carriers were used.
Those skilled in art will know to choose a suitable formulation of the instant composition for managing coronavirus infection, using a daily dose range from about 1 mg/kg to about 100 mg/kg of body weight of subject or preferably, a daily dose range from about 10 mg/kg to about 75 mg/kg of body weight of subject.
EXAMPLE 2: Effect of the composition on ACE2-spike 1 interaction
About 20 pg/mL of the composition of Example 1 was incubated with spike protein 1 coated wells for about 1 hour. Lung tissue lysates were added into the well and incubated for about 2 hours after wash. The unbound ACE2 was removed by washing and the bound ACE2 was detected by adding human ACE2-specific rabbit antibody and HRP -conjugated anti-rabbit IgG antibody. The reaction was then visualized by adding substrate and quenched by adding H2SO4. The binding was measured by an ELISA reader (OD450).
The composition was found to reduce the binding of ACE2 to COVID spike 1 protein by about 9% (Figure 1). In addition, the standard curve of ACE2 binding with coated spike 1 protein was showed in Figure 2.
Further, in another experiment the composition of Example 1 at different concentrations (5, 10, 20, 40, 80, 100, 150, 200 pg/mL) was added into recombinant ACE2 coated 96-wells and incubated for about 30 minutes. After wash, spike 1 protein (Figure 3, 1.0 pg/well; Figure 4, 0.1 pg/well) was added into the well and incubated at about 25 °C for about 1.5 hours. The unbound spike 1 protein was removed by washing and the bound spike 1 protein was detected by adding goat anti-sheep Fc antibody for about 2 hours, followed by three times of wash and incubated with HRP -conjugated anti-goat IgG antibody for about 1 hour. The reaction was then visualized by adding substrate and quenched by adding H2SO4. The binding was measured by an ELISA reader (OD450). The calculated ICso of the composition to ACE2-spike 1 protein interaction were about 21.82 pg/mL and about 9.949 pg/mL, respectively.
The above shows that the composition of the present disclosure composed of a combination of pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin inhibited the binding of the ACE2 receptor to the spike protein.
EXAMPLE 3: Binding kinetics and affinity (KD) of compositions of the present disclosure to human ACE2
For studying the binding affinities of the composition of Example 1 with human ACE2, the ACE2-His at about 10 pg/mL in running buffer (PBS buffer supplemented with about 0.02% Tween 20 and about 0. 1% BSA) was immobilized onto Ni-NT A biosensors, and then incubated with serial dilutions of the composition (about 33.3 pg/mL, about 11.1 pg/mL, about 3.7 pg/mL, about 1.2 pg/mL, about 0.4 pg/mL) in running buffer.
The experiments included the following steps at about 37 °C: (1) baseline (water, 60 s); (2)immobilization of ACE2-His onto sensors (300 s); (3) baseline (running buffer, 120 s); (4)association of the composition for measurement of kOn (serial-diluted composition as prepared in Example 1, 300 s); (5) dissociation of the composition for measurement of kos (running buffer, 300 s). The curves were fitted by a 1: 1 binding model using ForteBio Data Analysis software 10.0 (Figure 7).
The KD of the composition of Example 1 for ACE-2 was about 0.65 pM.
It was therefore found that the composition of the present disclosure showed significant binding efficacy to ACE2.
EXAMPLE 4: Effect of the composition on interaction of coronavirus RBD with ACE2 in a competition binding assay
Preparation of RBD-Fc (SARS-CoV-2)
The recombinant receptor binding domain (RBD) sequence encoding amino acids R319- F541 of SARS-CoV -2 was cloned into pSecTag2B expression vector with an additional C terminal Fc fragment of human IgGl and AviTag in tandem and transiently expressed in Expi293 HEK cells as a secreted protein. After 3 days, supernatants were harvested by centrifuging the culture at about 2500 g for about 15 minutes and filtering the supernatant with a 0.22 mm vacuum filter. The protein was purified using protein G resin (GE healthcare). Equilibration and wash steps were performed with phosphate buffered saline solution (PBS) (Hyclone) and protein were eluted in 0.1 M glycine pH 2.7. The eluates were pH equilibrated to 7.4 using 1.0 M Tris HCI pH 9.0 and immediately buffer-exchanged into PBS und concentrated using an Amicon ultra centrifugal concentrator (Millipore) with n molecular weight cut-off of 10 kDa. Purity was estimated to be about 95% by SDS- polyacrylamide gel electrophoresis, and protein concentration was measured using the NanoDrop spectrophotometer. The biotinylated proteins used in bio-panning and BLI experiment were prepared by the BirA biotin-protein ligase in PBS for about 30 minutes at about 30 °C, which adds biotin covalently to AviTag in a highly specific manner, according to the manufacturer's instructions (Avidity).
Binding kinetics and affinity (Kp) of the composition to SARS-CoV-2 RBD,
For measuring the binding affinities of the composition with RBD of SARS-CoV-2, the biotinylated RBD-Fc of SARS-CoV-2 at about 10 pg/mL in running buffer (PBS buffer supplemented with about 0.02% Tween 20 and about 0.1% BSA) was immobilized onto streptavidin-coated biosensors, and then incubated with serial dilutions of the compositions of Example 1 and 3 (about 33.3 pg/mL, about 11.1 pg/mL, about 3.7 pg/mL, about 1.2 pg/mL) in running buffer. The experiments included the following steps at 37 °C: (1) baseline (water, 60s); (2) immobilization of RBD-Fc onto sensors (150 s); (3) baseline (running buffer, 120 s);
(4) association of the composition for measurement of kon (serial-diluted composition, 300 s);
(5) dissociation of the composition for measurement of kott (running buffer, 300 s). The curves were fitted by a 1: 1 binding model using ForteBio Data Analysis software 10.0 (Figure 6).
KD of the composition of Example 1 for SARS-CoV-2 RBD was about 1.43 pM.
It was therefore found that the composition of the present disclosure effectively binds to SARS- CoV-2 RBD.
EXAMPLE 5: Neutralizing activity of the composition against pseudotyped SARS-CoV- 2 infection The neutralization activity of the Example 1 composition was measured with a SARS-CoV-2 spike (S) protein-mediated, luciferase -encoding pseudovirus neutralizing assay.
To determine the inhibition activity in pseudotyped virus assay, 293 T cells were cotransfected with expression vectors of pcDNA3. l-SARS-CoV-2-S (encoding SARS-CoV-2 S protein) and pNL4-3.luc.RE bearing the luciferase reporter-expressing HIV -1 backbone. The supernatants containing SARS-CoV-2 pseudotyped virus were harvested. Serial dilutions of single-domain antibodies in DMEM supplemented with 10% fetal calf serum were incubated with pseudo viruses at 37°C for 1 h and then the mixtures were added to monolayer Huh-7 cells (104 per well in 96-well plates). Twelve hours after infection, culture medium was refreshed and then incubated for an additional 48 hours. The luciferase activity was calculated for the detection of relative light units using the Bright-Glo™ Luciferase Assay System (Promega). A nonlinear regression analysis was performed on the resulting curves using Prism (GraphPad) to calculate half-maximal inhibitory concentration (IC50) values (Figure 7).
The composition showed potent neutralization activity, inhibiting SARS-CoV-2 pseudovirus infection with a half-maximal inhibitory concentration (ICso) values of about 9.232 pg/mL. The composition therefore was found to have the potential to inhibit the Pseudovirus.
EXAMPLE 6: Cytotoxicity assay
Cell cytotoxicity in Vero cells was determined following standard ACP assay protocol.
Huh-7 cells in the logarithmic phase of growth were collected and seeded in 96-well plates at a density of about 1 x 104 cells/well and cultured overnight. The culture medium was removed and serial dilution of the composition of claim 1 was added into wells of 96-well plates in 200pl medium. Cells were treated for about 48 hours at about 37 °C. After treatment, the culture medium was removed and lOOpl CCK8 solution (lOpl CCK8 + 90pl medium) (Dojindo Molecular Technologies, Gaithersburg, MD) was added into each well, then incubated at 37°C for 2-4 h. Absorbance was measured at 450nm using a 96-well microplate reader. Each concentration was repeated in triplicate.
It was observed that the composition showed no cytotoxicity against human cells (Figure 7). EXAMPLE 7: Plaque reduction assay
Plaque assay was performed in triplicate in 24-well tissue culture plates. The VeroE6 cells were seeded in DMEM with 10% FCS and antibiotics one day before infection. SARS-CoV-2 was added to the cell monolayer for 1 hour at 37°C. Subsequently, viruses were removed and the cell monolayer was washed once with PBS before covering with media containing agarose or methylcellulose for 5-7 days. The cells were fixed with formaldehyde overnight. After removal of overlay media, the cells were stained with crystal violet and the plaques were counted. The percentage of inhibition was calculated as [1 -(VD/ VC)] X 100%, where VD and Vc refer to the virus titer in the presence and absence of the compound, respectively.
As shown in Figure 8A the composition of Example 1 could inhibit SARS-CoV-2 efficiently. This demonstrated inhibitory activity of the compositions against authentic SARS-CoV-2 with 50% inhibition concentration (ICso) values of 3.62 ± 0.04 pg/mL (Figure 8B). The cell toxicity of the compositions was also determined by ACP assay and the cell cytotoxicity values for the composition was estimated to be more than 80 pg/mL (Figure 8C). The derived SI index for the composition of Example 1 was >22.1.
EXAMPLE 8: Time of addition assay
Vero E6 cells were seeded to the 24-well culture plate in DMEM with 10% FBS and antibiotics one day before infection. Vero E6 cells were infected by SARS-CoV-2 virus (100 pfu/well) for about 1 hour at about 37°C. After removal of virus inoculum, the cells were washed once with PBS and overlaid with 1 mL overlay medium containing 1% methylcellulose for 5 days at 37°C. After about 5 days, the cells were fixed with about 10% formalin overnight. After removal of overlay media, the cells were stained with about 0.5% crystal violet and the plaques were counted. For “Entry” treatment, the cells were first pre-treated with the compound for about 1 hour, then infected with virus in the presence of the compound. For “Post-entry” treatment, the compound was added to the cells at 1 hpi (Hours post infection), and maintained in the overlay medium until the end of the experiment. The percentage of inhibition was calculated as [ 1 -(VD / Vc)] x W0%, where VD and Vc refer to the virus titer in the presence and absence of the composition, respectively. The minimal concentration of composition required to reduce 50% of plaque numbers (ECso) was calculated by regression analysis of the dose-response curves generated from plaque assays. To elucidate the mechanism of action of the composition of Example 1 on SARS-CoV2, time of addition assays were performed. For “Entry” treatment, the cells were first pre-treated with the compound for about 1 hour, followed by infection of the virus in the presence of the compound. For “Post-entry” treatment, the composition was added to the cells at 1-hour post infection (hpi). The composition showed inhibition of SARS CoV2 mainly at the entry level, and also some inhibitory effect at the post entry stage of the virus infection (Figure 9).
EXAMPLE 9: Clinical study of effect of the composition on Coronavirus infected patients
This was an interventional, randomized, double-blind, placebo-controlled clinical study to evaluate the safety and effectiveness of the composition of Example 1, formulated as capsules, in preventing the progression of disease severity in COVID-19 patients.
SARS-CoV2 positive patients with mild to moderate COVID- 19 manageable as per the guidelines from the Government of India at CO VID-19 management centres capable of being integrated with Ayurvedic care were eligible for participation in this strictly, after obtaining their written informed consent.
All patients were pre-screened for COVID- 19 symptoms and RT-PCR test prior to screening procedures for the study. Following the baseline screening evaluations, patients were randomized into the study to receive either the test composition or placebo capsules (1:1) for about 15 days in two regimens, along with standard of care treatment: For initial 3 days: the composition or placebo was administered as about 3 g/day (2 capsules of 500 mg X 3 times) as a loading dose.
For 12 subsequent days, the test composition or placebo was administered as 1.5 g/day (1 capsule of 500 mg X 3 times) as a maintenance dose.
The screening and enrolment procedures were done at the study centre if the patient was being treated either in hospital or quarantine setting. In case the patient chose, or the Investigator recommended self/home quarantine, a study team member performed a home visit during the time of randomization of the patient into the study, to ensure that all study related procedures were completed appropriately. The other study visits were performed either at the study centre (if the patient was hospitalized) or the study team needed to perform cither a home or telephonic visit (if the patient was at home) to ensure study-related activities were completed. At all times, everyone involved followed social distancing to the extent possible and wore masks throughout the interaction.
The treatment period consisted of about 15 days for all the patients: unless the treatment ended earlier because of withdrawal of consent by the patient or in case the patient progressed to a more severe stage of the disease or in case of death of the patient, or as per the discretion of the investigator.
In case of hospitalized patients, if the patient was discharged from the hospital earlier than 15 days from enrolment in the study, the study treatment was provided to the patient and the patient was required to continue and complete the study treatment of 15 days on an OPD basis at his/her home.
For patients who tested negative on RT-PCR at the End of Treatment (EoT) and the End of Study (EoS), visit was completed as per the Investigator's discretion, either telephonically if the patient was already at home: or at the study centre, if the patient was still in the hospital.
Efficacy results:
The primary endpoint of the study - time to clinical recovery - defined as the time required to attain a score of either 1 (asymptomatic; viral RNA detected) or 0 (uninfected; no vims detected) on the WHO Clinical Progression Scale was comparable between the composition and Placebo groups with no statistically significant difference. The median (SE) time to recovery for the composition treated group was about 8 (0.678) days, while that for the Placebo group was about 7 (0.419) days.
All the 59 (100%) patients who tested covid positive in the composition treatment group and 57 (96.6%) patients in the Placebo group showed clinical recovery and clinical improvement at end of treatment. The median (SE) time to clinical improvement of 7.0 (0.147) days in the composition treated group was comparable to that of 7.0 (0.212) days in the Placebo. The median duration of hospital stay was 9.5 days in the Composition treatment group and 10 days in placebo group. The difference in the proportion of patients with negative RT-PCR finding between the composition treated group (56 patients; 94.9%) and placebo groups (49 patients, 83.1%) was statistically significant (p-0.039). This shows that the test composition had stopped the further progression of the disease which might have lead to severe complications. Thus the test composition helped the patients to recover without causing severe complications
A significantly (p-=0.0496) higher number of patients showed improvement in oxygen saturation (SpCh) by pulse oximetry in the composition treated group (21 i.e. 35.59%) as compared to the placebo group (11 i.e. 19.29%). Also, none of the patients in the composition group required ventilation support during the study period. This shows the test composition helped the patients in significant improvement of oxygen levels further preventing the severe complications such as difficulty in breathing, pneumonia or blood clots.
The treatment group was associated with significant improvement in the mean total score of WHO-QOL-BREF Questionnaire from Day I to EOT, comparable to the Placebo group. The mean scores for the physical health domain were significantly higher on Day 8 and Day 14 for the composition treated group as compared to the placebo group. The mean scores for the psychological health domain were higher in the composition treated group as compared to the Placebo group at each day. The mean scores for the social relationship domain were significantly higher on day 6 as compared to the Placebo group, and continued to be higher for all other days. The mean scores for the environmental domain were significantly higher on days 6 to 9. day 11. and EOT in the composition treated group as compared to the Placebo group. The within-group comparison of WHO-QOL-BREF scores for all the four domains in the subset of hospitalized patients revealed that the scores for the psychological health domain in the composition treated group differed significantly between Day 1 and end of hospital stay. Further, in the composition treated group, the overall total score at the end of hospital stay patients was also significantly higher as compared to Day 1.
Overall. 5 (4.3%) patients reported at least one adverse event during the study: 2 (3.4%) patients in the composition treatment group had skin rash while 3 (5.3%) patients in the Placebo group had skin rash (2 patients) and elevated triglycerides (1 patient). None of the adverse events were considered related to the study treatment. This shows that the test composition was safe and was not associated with any adverse events. Further, treatment with the composition was not associated with prolonged hospitalization as compared with placebo.
Overall, the investigational composition of Example 1 was reported to have the potential of therapeutic activity in patients with mild to moderate covid- 19 infection.

Claims

WE CLAIM:
1. A composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) for use in preventing, managing or treating Coronavirus infection and/or associated complication s).
2. The composition for use as claimed in claim 1, wherein the Coronavirus infection is caused by SARS-CoV-2 or mutants or variants thereof.
3. The composition as claimed in claim 1, wherein the associated complication is selected from a group comprising reduction in quality of life, reduction in oxygen saturation (SpCh) levels, pneumonia, troubled breathing, blood clot formation, cardiac problems and organ failure or any combination thereof.
4. The composition as claimed in claim 1, wherein the associated complication is selected from a group comprising reduction in quality of life and/or reduction in oxygen saturation (SpCh) levels.
5. The composition for use as claimed in claim 1, wherein the pentameric type A procyanidin is at concentration ranging from about 55 % w/w to about 99% w/w, the tetrameric type A procyanidin and the trimeric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 35 % w/w; and the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
6. The composition for use as claimed in claim 1, wherein the pentameric type A procyanidin is at concentration ranging from about 80 % w/w to about 90% w/w, the tetrameric type A procyanidin and the trimeric type A procyanidin are each at concentration ranging from about 0.5 % w/w to about 20 % w/w; and the pharmaceutically acceptable excipient(s) is at concentration ranging from about 0.5% to about 99.9%.
7. The composition for use as claimed in claim 1 , wherein the pharmaceutically acceptable excipient(s) is selected from a group comprising gums, granulating agents, binding agents, lubricating agents, disintegrating agents, sweetening agents, coloring agents, flavoring agents, coating agents, plasticizers, preservatives, suspending agents, emulsifying agents, antistatic agents, and spheronization agents or any combination thereof.
8. The composition for use as claimed in claim 1, wherein composition is formulated into dosage forms selected from group comprising solid formulation, liquid formulation, parenteral formulation, aerosol or spray, phytoceutical, nutraceutical and medicinal foods or any combinations thereof.
9. The composition for use as claimed in claim 8, wherein the solid oral formulation is selected from group comprising tablet, capsule, troche, lozenge, dispersible powder, dispersible granule or any combinations thereof; wherein the liquid oral formulation is selected from group comprising aqueous or oily suspension, emulsion, drops, emulsion in hard or soft gel capsule, syrup, elixir or any combinations thereof; wherein the parenteral formulation is selected from group comprising intravenous injection, intramuscular injection, intramuscular depot, subcutaneous injection, percutaneous injection or any combinations thereof; and wherein the aerosol formulation is selected from a group comprising oral spray, nasal spray and inhaler.
10. The composition for use as claimed in claim 1, wherein composition is formulated into a nasal spray.
11. The composition for use as claimed in claim 1, wherein the use is as a medicament for preventing, managing, or treating Coronavirus infection and/or associated complication(s).
12. A method for preventing entry of Coronavirus into a host cell or inhibiting survival and/or propagation of live Coronavirus in a host cell, comprising contacting the host cell with a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s).
13. The method as claimed in claim 12, wherein the composition inhibits formation of a complex between ACE2 receptor of a host cell and Spike protein of the Coronavirus.
14. The method as claimed in claim 13 , wherein the composition binds to the ACE2 receptor ofa host cell.
15. The method as claimed in claim 14, wherein the composition binds to Receptor Binding Domain (RBD) of spike protein of the Coronavirus.
16. A method of preventing, treating and/or managing Coronavirus infection and/or associated complication(s) comprising administering a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s) to a subject in need thereof, at a pharmaceutically effective dosage.
17. The method as claimed in any of claim 16, wherein the pharmaceutically effective dosage ranges from about 1 mg/kg to about 100 mg/kg body weight of the subject.
18. The method as claimed in claim 16, wherein the subject is a mammal, including human beings.
19. The method as claimed in in claim 16, wherein the method improves oxygen saturation in an infected subject to a value from about 89% to about 100%. 0. A kit comprising a composition comprising pentameric type A procyanidin, tetrameric type A procyanidin and trimeric type A procyanidin, optionally along with pharmaceutically acceptable excipient(s), along with a pamphlet or brochure indicating suitable dosage of the composition. 1. The kit as claimed in claim 20, wherein the kit further comprises means for administration of the composition.
PCT/IB2021/061564 2020-12-10 2021-12-10 Composition and method for managing coronavirus infection WO2022123513A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
IN202021024421 2020-12-10
IN202021024421 2020-12-10

Publications (1)

Publication Number Publication Date
WO2022123513A1 true WO2022123513A1 (en) 2022-06-16

Family

ID=81973157

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2021/061564 WO2022123513A1 (en) 2020-12-10 2021-12-10 Composition and method for managing coronavirus infection

Country Status (1)

Country Link
WO (1) WO2022123513A1 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060275515A1 (en) * 2003-12-24 2006-12-07 Michael Ovadia Antiviral preparations obtained from a natural cinnamon extract
US20110039923A1 (en) * 2009-08-11 2011-02-17 Indus Biotech Private Limited Novel standardized composition, method of manufacture and use in the resolution of rna virus infection
US20120190736A1 (en) * 2010-07-28 2012-07-26 Sunil Bhaskaran Method of managing broncho-constrictive condition
CN111789880A (en) * 2020-05-07 2020-10-20 郑州航空港百桥生物科技有限公司 Application of cinnamon extract in inhibiting entry and replication of novel COVID-19 coronavirus into cells

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060275515A1 (en) * 2003-12-24 2006-12-07 Michael Ovadia Antiviral preparations obtained from a natural cinnamon extract
US20110039923A1 (en) * 2009-08-11 2011-02-17 Indus Biotech Private Limited Novel standardized composition, method of manufacture and use in the resolution of rna virus infection
US20120190736A1 (en) * 2010-07-28 2012-07-26 Sunil Bhaskaran Method of managing broncho-constrictive condition
CN111789880A (en) * 2020-05-07 2020-10-20 郑州航空港百桥生物科技有限公司 Application of cinnamon extract in inhibiting entry and replication of novel COVID-19 coronavirus into cells

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
IHERB.COM, NATURAL FACTORS, PYCNOGENOL, 25 MG, 60 VEGETARIAN CAPSULES, Retrieved from the Internet <URL:https://au.iherb.com/pr/natural-factors-pycnogenol-25-mg-60-vegetarian-capsules/2630> [retrieved on 20220127] *
PRASANTH D. S. N. B. K., MURAHARI MANIKANTA, CHANDRAMOHAN VIVEK, PANDA SIVA PRASAD, ATMAKURI LAKSHMANA RAO, GUNTUPALLI CHAKRAVARTH: "In silico identification of potential inhibitors from Cinnamon against main protease and spike glycoprotein of SARS CoV-2", JOURNAL OF BIOMOLECULAR STRUCTURE & DYNAMICS, ADENINE PRESS, NEW YORK, NY, US, vol. 39, no. 13, 22 June 2020 (2020-06-22), US , pages 4618 - 4632, XP055815066, ISSN: 0739-1102, DOI: 10.1080/07391102.2020.1779129 *
THAKUR A.: "Benefits of Cinnamon in COVID-19: How it Fights Lung Infections And Boosts Immunity", INDIA.COM, 4 September 2020 (2020-09-04), XP055943895, Retrieved from the Internet <URL:https://www.india.com/lifestyle/benefits-of-cinnamon-in-covid-19-how-it-fights-lung-infections-and-boosts-immunity-4131409> [retrieved on 20220719] *
WEICHMANN, F ET AL.: "Projected supportive effects of Pycnogenol® in patients suffering from multi-dimensional health impairments after a SARS-CoV2 infection", INTERNATIONAL JOURNAL OF ANTIMICROBIAL AGENTS, vol. 56, 2020 - 9 October 2020 (2020-10-09), pages 106191, XP086367965, DOI: 10.1016/j.ijantimicag.2020.106191 *

Similar Documents

Publication Publication Date Title
CA2721040C (en) Procyanidin extracts of cinnamon, litchi and arachis useful in the treatment of retroviral infections
CA2817787C (en) Composition comprising a peptide and an inhibitor of viral neuraminidase
US20210196673A1 (en) Compositions and methods for treating and preventing respiratory viral infections using green tree extract
RU2505306C2 (en) Composition for preventing and treating viral infections
US20210315857A1 (en) Pharmaceutical micronutrient composition and its use to simultaneously inhibit multiple cellular mechanisms of infectivity caused by coronavirus, its variants and mutants
WO2022036774A1 (en) Application of tannic acid in preparation of anti-respiratory virus drug
CN112386595A (en) Pharmaceutical composition for treating viral infection of respiratory system
WO2020037095A1 (en) Egcg-palmitate compositions and methods of use thereof
CN116018130A (en) Catechin-containing composition and use
AU2011284357B2 (en) A method of managing broncho-constrictive condition
US20220047545A1 (en) Micronutrient combination to inhibit coronavirus cell infection
WO2022123513A1 (en) Composition and method for managing coronavirus infection
CN108014102A (en) The micromolecular inhibitor of Ebola&#39;s pseudovirus
WO2021191904A1 (en) Methods for preventing and treating viral infection
US20090076132A1 (en) Antiviral compositions and methods of treatment
US20230190848A1 (en) P2et reduces covid severity by inhibition of viral replication, reduction of pulmonar fibrosis markers and modulation of inmune response
US20200299331A1 (en) Composition Comprising a Peptide and an Inhibitor of Viral Neuraminidase
WO2024096743A1 (en) Sars-cov-2 binding antibody
TWI421072B (en) A method of managing broncho-constrictive condition
GB2622661A (en) Composition
JP2003155230A (en) Anti-influenza medicine
JP5525376B2 (en) Anti-influenza virus agent
RR et al. Effect of" Deep Block®" in the short and long term in patients with COVID-19: Prospective Phase II study.

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: 21902854

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: 21902854

Country of ref document: EP

Kind code of ref document: A1