COMPOSITIONS AND METHODS FOR TREATING AND PREVENTING NON-MALIGNANT RESPIRATORY DISEASE

Information

  • Patent Application
  • 20230346866
  • Publication Number
    20230346866
  • Date Filed
    March 19, 2021
    3 years ago
  • Date Published
    November 02, 2023
    5 months ago
  • Inventors
    • ALKALAY; Rachel
  • Original Assignees
    • NOVEL CONCEPTS MEDICAL LTD
Abstract
A method of preventing or treating non-malignant respiratory disease (NMRD) in a subject in need thereof is provided. The method comprising administering to the subject an effective amount of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma polyphyllum, Boswellia sacra and Panax ginseng preventing or treating NMRD in the subject.
Description
FIELD AND BACKGROUND OF THE INVENTION

The present invention, in some embodiments thereof, relates to compositions and methods for treating and preventing non-malignant respiratory disease (NMRD).


Non-malignant respiratory disease (“NMRD”, also known as non-malignant pulmonary disease or “NMPD”) is a diverse group of acute and chronic conditions affecting the respiratory tract and interfering with gas exchange which constitute a common and significant cause of illness and death worldwide, and make up the vast majority of ICU utilization throughout all hospital systems. Chronic lung disease of some form, including, inter alia, Interstitial Lung Disease (ILD), Bronchiectastis and Chronic Obstructive Pulmonary Disease (COPD), affects greater than 10% of individuals in both highly industrialized and less highly industrialized nations, with chronic obstructive pulmonary disease being the most common.


Chronic respiratory disease is of particular importance in Environmental and Occupational Medicine, with environmental (e.g. pollution) and workplace exposures contributing to the burden of disease across a range of nonmalignant lung conditions in the young, in adults and in the elderly (in addition to the 100% burden for the classic occupational pneumoconioses). This burden has important clinical, research, and policy implications. Current treatment for chronic respiratory disease focuses mostly on palliative care.


According to the World Health Organization (WHO) study, in low-income countries pneumonia leads the top five death causing diseases followed by heart disease, diarrhea, HIV/AIDS and stroke, in high-income countries, pneumonia, and asthma/bronchitis are closing the top five death causing diseases topped only by heart disease, stroke and lung cancer. (see who.int/news/item/27-10-2008-new-study-presents-state-of-the-world-s-health).


Thus, there is great need for a new therapeutic and preventive treatment for non-malignant respiratory disease.


SUMMARY OF THE INVENTION

According to an aspect of the invention there is provided a method of preventing or treating non-malignant respiratory disease (“NMRD”) in a subject in need thereof, the method comprising administering to the subject an effective amount of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng, preventing or treating NMRD in the subject.


According to an aspect of the invention there is provided a vaccine against a NMRD comprising an effective amount of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.


According to an aspect of the invention there is provided a pharmaceutical composition comprising an effective amount of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng for use in preventing or treating NMRD.


According to an aspect of the invention there is provided a composition of matter comprising at least 2 of a plant species or genus thereof-derived components selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.


According to an aspect of the invention there is provided a food supplement comprising a combination of at least 2 of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.


According to an aspect of the present invention there is provided compositions or food supplements comprising Bromelain or pineapple extracts comprising Bromelain.


According to some embodiments of the invention the NMRD is selected from the group pulmonary hypertension, interstitial lung disease, acute lung injury (ALI), acute respiratory distress syndrome (ARDS), sarcoidosis of the lung, bronchiectasis, asbestosis, berylliosis, silicosis, anthracosis, Histiocytosis X, pneumotitis, smoker's lung, bronchiolitis obliterans, lung scarring due to tuberculosis and pulmonary fibrosis, chronic obstructive pulmonary disease, pneumoconiosis, traumatic pulmonary injury, pulmonary infections and pulmonary manifestations of connective tissue diseases, including systemic lupus erythematosus, rheumatoid arthritis, scleroderma, polymyositis and dermatomyositis.


According to some embodiments of the invention the NMRD is Chronic Obstructive Pulmonary Disease (COPD), Idiopathic Pulmonary Fibrosis (IPF) or Asthma.


According to some embodiments of the invention the symptoms are selected from the group consisting of coughing, shortness of breath, wheezing, difficulty in breathing, inflammation of the airways, chest tightness and exercise intolerance.


According to some embodiments of the invention, the component comprises at least 2 components.


According to some embodiments of the invention, the component comprises at least 3 components.


According to some embodiments of the invention, the component comprises at least 4 components.


According to some embodiments of the invention, the component comprises at least 5 components.


According to some embodiments of the invention, the component comprises 5-10 components.


According to some embodiments of the invention, the component comprises thymoquinone or an analog thereof.


According to some embodiments of the invention, the component comprises thymol or an analog thereof.


According to some embodiments of the invention, the component comprises carvacrol or an analog thereof.


According to some embodiments of the invention, the component comprises Tryptophan or an analog thereof.


According to an aspect of the invention there is provided a food supplement, composition or extracts further including “Beduin Tea” comprising


Rose Leaves Micromeria fruticose, Salvia, cymbopgon (Citral) Aloysia, Verbena officinalis, Origanum majorana, menthe

According to an aspect of the invention there is provided a food supplement, composition or extracts further including “Beduin Tea” comprising


Thyme, sage, cardamom, cinnamon, black tea, habuk, Marmaya.


Further details of components of Thyme Vulgaris are included in APPENDIX1.


Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.





BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWING(S)

Some embodiments of the invention are herein described, by way of example only, with reference to the accompanying drawings. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of embodiments of the invention. In this regard, the description taken with the drawings makes apparent to those skilled in the art how embodiments of the invention may be practiced.


In the drawings:



FIGS. 1A-C shows embodiments in plant extraction methods as taken from berkem(dot)com. FIG. 1A—scheme describing the general principle of plant extraction; FIG. 1B—scheme describing the main separation process according to some embodiments; FIG. 1C—scheme describing parameters that may influence the process.



FIG. 2—depicting SDS-page of the SARS-CoV-2 S1 subunit protein digestion assay with the tested extracts following an incubation time of 6 h at 37° c.



FIG. 3—depicting SDS-page of the SARS-CoV-2 S2 subunit protein digestion assay with the tested extracts following an incubation time of 6 h at 37° c.



FIG. 4—depicting SDS-page of the SARS-CoV-2 Nucleocapsid digestion assay with the tested extracts following an incubation time of 6 h at 37° c.



FIG. 5—depicting a graphic representation of the densitometry test of the SARS-CoV-2 S1 subunit protein digestion assay with the tested extracts following an incubation time of 6 h at 37° c.



FIG. 6—depicting a graphic representation of the densitometry test of the SARS-CoV-2 S2 subunit protein digestion assay with the tested extracts following an incubation time of 6 h at 37° c.



FIG. 7—depicting a graphic representation of the densitometry test of the SARS-CoV-2 Nucleocapsid digestion assay with the tested extracts following an incubation time of 6 h at 37° c.





DESCRIPTION OF SPECIFIC EMBODIMENTS OF THE INVENTION

The present invention, in some embodiments thereof, relates to compositions and methods for treating and preventing non-malignant respiratory disease (NMRD).


Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not necessarily limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways.


Non-malignant respiratory disease (NMRD) is a group of acute and chronic conditions affecting the respiratory tract and interfering with gas exchange, exclusive of cancerous and malignant proliferative pulmonary disease. In mild forms (e.g. the common cold, mild bronchitis, pharyngitis, mild allergic respiratory distress), symptoms of NMRD may include coughing, wheezing, pulmonary and/or nasal congestion, copious mucus and mild shortness of breath. Most often, patients with mild NMRD will experience mild to moderate symptoms and recover without requiring special treatment. Moderate to severe cases of NMRD, and particularly in older individuals, those with underlying medical problems and individuals frequently exposed to common triggers of respiratory disease such as particulate pollution (e.g. coal dust, asbestos, silica, etc), dust mites and allergens are more likely to develop into serious, chronic illness, in many cases requiring intensive care with ventilation and life-threatening disease. At this time, there are no specific vaccines or effective preventative treatments for many of the non-malignant respiratory conditions.


Thus, according to an aspect of the invention there is provided a method of preventing or treating a non-malignant respiratory disease or condition (NMRD) in a subject in need thereof, the method comprising administering to the subject an effective amount of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng, preventing or treating NMRD in the subject.


According to an alternative or an additional aspect of the invention there is provided a vaccine against a NMRD comprising an effective amount of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.


Plant derived component, synthetic analogs and the like are well established compositions used to treat a wide spectrum of diseases and syndromes. From the Aspirin (acetyl salicylic acid) historically discovered as a component of Salix alba to the Vinca derived chemotherapeutic drugs vinblastine and vincristine. It stands to reason that plant derived components and substances are capable of attenuating a wide spectrum of viral infections and ameliorating the same. Moreover, the aforementioned plant substances maybe used as a profilaxis for human but for livestock as well, thus lowering the rate of zoonosis. Some representative viruses, viral families and viral genus are depict in table A.












TABLE A





FAMILY
GENUS
DISEASE/VIRUS
ABBREVIATION







Adenoviridae

Respiratory
HAdV-B/C




Adenovirus


Arenaviridae

Mammarenavirus

Lassa Virus
LASV


Astroviridae

Mamstrovirus

Mamstrovirus
MAstV


Caliciviridae

Norovirus


Coronaviridae

Severe Acute
SARS-CoV




Respiratory




Syndrome




Middle East
MERS-CoV




Respiratory




Syndrome




Covid-19
SARS-CoV-2


Filoviridae

Ebolavirus

Ebola Virus
EVD




Marburgvirus

Marburg Virus
marv


Flaviviridae

Flavivirus

Dengue Fever
DENV




Flavivirus

Japanese Encephalitis
JEV




Flavivirus

Powassan Virus
POWV




Flavivirus

West Nile Virus
WNV




Flavivirus

Yellow Fever




Flavivirus

Zika Virus
ZIKV


Hantaviridae

Orthohantavirus

Hanta Virus
HPS




Pulmonary Syndrome


Herpesviridae

Herpes Simplex
HSV




Varicella Zoster
VZV




Cytomegalovirus
CMV




Epstein-Barr Virus
EBV




Roseola Virus
HHV-6


Matonaviridae

Rubivirus

Rubella (German
RUBV




Measles)


Nairoviridae

Orthornavirus

Crimean-Congo
CCHFV




Hemorrhagic Fever


Papillomaviridae

Human Papilloma
HPV




Virus


Paramyxoviridae

Morobilivirus

Measles
MeV




Orthobulavirus

Mumps
MuV




Henipavirus

Nipah Virus
NiV


Peribunyviridae

Orthobunyavirus

Cache Valley
CVV




Orthobunyavirus


Picornaviridae

Poliomyelitis
Polio




Coxsackie Virus
CV




Echovirus
EV




Hepatitis A, B, C, D, E
HAV, HBV, HCV, HDV, HEV


Pneomoviridae

Orthopneomovirus

Respiratory Syncytial Virus
RSV




Metapneumovirus

Human
HMPV




Metapneumovirus


Poxviridae

Orthopoxviros

Variola (Small Pox)
VARV


Reoaviridae

Rotavirus

Rota Virus


Rhabdoviridae

Lyssavirus

Rabies
RARV


Retroviridae

Lentivirus

Human
HIV 1/HIV 2/AIDS




immunodeficiency




virus


Togaviridae

Alphavirus

Chinkungunya
CHIKV









According to an alternative or an additional aspect of the invention there is provided a pharmaceutical composition comprising an effective amount of a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng for use in preventing or treating a NMRD.


According to an alternative or an additional aspect of the invention there is provided a composition of matter comprising at least 2 of a plant species or genus thereof-derived components selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein the component is capable of ameliorating symptoms of NMRD and wherein the plant species is selected from the group consisting of Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.


The term “plant” as used herein encompasses whole plants, a grafted plant, ancestors and progeny of the plants and plant parts, including seeds, flowers, bark, shoots, stems, roots (including tubers), fruit, rootstock, scion, and plant cells, tissues and organs.


According to a specific embodiment, the plant part is a seed.


According to a specific embodiment, the plant part is a fruit.


According to a specific embodiment, the plant part is a leaf.


According to a specific embodiment, the plant part is a stem.


According to a specific embodiment, the plant part is a flower.


The plant part can be a solid part or a non-solid part such as oil or aqueous portions of the plant.


The plant may be in any form including suspension cultures, embryos, meristematic regions, callus tissue, leaves, gametophytes, sporophytes, pollen, and microspores.


The term plant refers to a wild plant or a cultivated variety thereof.


As used herein the term “plant species” refers to a sub-group of one or more plants within the genus. These plants will share similar characteristics with each other. There may be a single plant within a species, or there may be many hundreds of plants. The term intends to include subspecies, such as grown or can be found in different geographical location, e.g., Lebanese Sumac and Syrian Sumac or Korean ginseng and American ginseng.


As used herein “plant genus” refers to a taxonomic rank below family and above species.


It will be appreciated that the relevant species and genera and listed below and each option or combination thereof represents a different embodiment of the invention.


The term “extraction” refers to a separation process which relies on the separation of one or more analytes from the components of a sample other than the one or more analytes. Extractions are processes that typically use two immiscible phases to separate one or more solutes from one phase into the other. The distribution of a solute between two phases is an equilibrium condition described by partition theory. For example, boiling tea leaves in water extracts the tannins, theobromine, and caffeine out of the leaves and into the water. More typical extractions preformed typically but not only in a laboratory are settings of organic compounds out of an aqueous phase and into an organic phase. Common extractants are arranged from ethyl acetate to water (ethyl acetate<acetone<ethanol<methanol<acetone:water (7:3)<ethanol:water (8:2)<methanol:water (8:2)<water) in increasing order of polarity according to the Hildebrand solubility parameter. Procedures for plant extraction are provided in FIGS. 1A-C.


The term “extract” as used herein refers to the result of such process of separation that can take the form of a solution formulation or other chemical form depending on the extraction process. In particular, the term extract can relate to a substance made by extracting a part of a sample (e.g. a raw material), such as by using a solvent such as ethanol or water. In various instances an extract relates to a solvent that is enriched in one or more solute. In particular, a “plant extract” in the sense of the present disclosure typically comprises a concentrated preparation of a plant material obtained by isolating or purifying desired active constituents with one or more extraction processes.


The choice of the solvent depends on the desired component to be obtained. For example, to extract polar components in an extraction process suggested solvents include, but are not limited to, water, ethanol methanol or butanol while for non polar compounds diethyl ether, hexane or chloroform depending on the use of the extract. For midpolar one may choose Ethyl acetate but other solvents can be used as well.


The general procedure of solid/liquid extraction can be scaled in five different ways:


Maceration: the contact stage is maintained at room temperature.


Decoction or reflux: the contact stage is maintained at the boiling point of the solvent.


Digestion: the contact stage is maintained at a temperature in between those of the previous two cases.


Infusion: the boiling solvent is poured over the solid, then left to cool for a set time.


Leaching or percolation: the solvent passes through the biomass.


It is also possible to combine these methods with each other or with other processes such as distillation, steam distillation, rectification, etc.


According to another embodiment, the use of various solvents, either successively or in combination is contemplated and the ordinary skilled of organic chemistry will know which to choose according to the active ingredient as described below.


Extraction may be further assisted by other means such as ultrafiltration, reverse osmosis, high pressure (supercritical CO2), microwaves, ultrasound, etc.


In some embodiments, the plant part is contacted with a polar solvent (e.g. ethanol) or nonpolar solvent (e.g., hexane or pentane) for several minutes, e.g., 15 minutes or more, about 30 minutes or more, about 1 hour or more, about 2 hours or more, or about 5 hours or more.


Temperature can also be controlled during the contacting.


According to specific embodiments, the plant part is contacted with the solvent (e.g. ethanol) while being constantly mixed e.g. on a shaker.


It will be appreciated that the extraction process can also be solvent-free.


For example, solvent-free microwave extraction (SFME) has been proposed as a green method for the extraction of essential oil from aromatic herbs that are extensively used in the food industry. This technique is a combination of microwave heating and dry distillation performed at atmospheric pressure without any added solvent or water. The isolation and concentration of volatile compounds is performed in a single stage. In some embodiments, SFME and/or hydro-distillation (HD)), are used for the extraction of essential oil from the plants of the invention.


In some embodiments, the process of the present invention comprises isolating a liquid extract (i.e. filtered extract) from the mixture (i.e. crude extract) comprising the liquid extract and solids. Suitable means for isolating the liquid extract (i.e. filtered extract) include those known in the art of organic synthesis and include, but are not limited to, gravity filtration, suction and/or vacuum filtration, centrifuging, setting and decanting, and the like. In some embodiments, the isolating comprises filtering a liquid extract through a porous membrane, syringe, sponge, zeolite, paper, or the like having a pore size of about 1-5 μm, about 0.5-5 μm, about 0.1-5 μm, about 1-2 μm, about 0.5-2 μm, about 0.1-2 μm, about 0.5-1 μm, about 0.1-1 μm, about 0.25-0.45 μm, or about 0.1-0.5 μm (e.g. about 2 μm, about 1 μm, about 0.45 μm, or about 0.25 μm).


According specific embodiments, the present invention contemplates drying (i.e. removal of the polar/non-polar solvent) and/or freezing the filtered extract following generation thereof.


The method for drying the filtered extract (i.e. removing the polar solvent) is not particularly limited, and can include solvent evaporation at a reduced pressure (e.g., sub-atmospheric pressure) and/or an elevated temperature (e.g., above about 25° C.). In some embodiments, it can be difficult to completely remove a solvent from a liquid extract by standard solvent removal procedures such as evaporation. In some embodiments, processes such as co-evaporation, lyophilization, and the like can be used to completely remove the polar solvent from a liquid fraction to form a dry powder, dry pellet, dry granulate, paste, and the like. According to a specific embodiment the polar solvent is evaporated with a vacuum evaporator.


The selection of the extraction process much depends on the component to be isolated.


It will be appreciated that following generation of the extract, specific embodiments of the present invention further contemplate additional purification steps so as to further isolate/purify active agents from the extract, for example, by fractionating the filtered extract.


As used herein “a fraction” refers to a portion of the extract that contains only certain chemical ingredients of the extract but not all.


Fractionating can be performed by processes such as, but not limited to: column chromatography, preparative high performance liquid chromatography (“HPLC”), reduced pressure distillation, and combinations thereof.


According to a specific embodiment, fractionating is performed by HPLC.


In some embodiments, fractionating comprises re-suspending the filtered extract in a polar solvent (such as methanol, as discussed above), applying the polar extract to a separation column, and isolating the extract having the anti-respiratory disease (e.g. anti-fibrotic, anti-inflammatory) activity by column chromatography (preparative HPLC).


An eluting solvent is applied to the separation column with the polar extract to elute fractions from the polar extract. Suitable eluting solvents for use include, but are not limited to, methanol, ethanol, propanol, acetone, acetic acid, carbon dioxide, methylethyl ketone, acetonitrile, butyronitrile, carbon dioxide, ethyl acetate, tetrahydrofuran, di-iso-propylether, ammonia, triethylamine, N,N-dimethylformamide, N,N-dimethylacetamide, and the like, and combinations thereof.


According to an alternative or an additional embodiment, liquid chromatography comprises high performance liquid chromatography (HPLC).


According to an alternative or an additional embodiment, liquid chromatography is performed on a reverse stationary phase.


The fractions may be characterized by analytical methods such as, but not limited to, spectroscopic methods such as, but not limited to, ultraviolet-visible spectroscopy (“UV-Vis”), infrared spectroscopy (“IR”), and the like; mass-spectrometry (“MS”) methods such as, but not limited to, time-of-flight MS; quadrupole MS; electrospray MS, Fourier-transform MS, Matrix-Assisted Laser Desorption/Ionization (“MALDI”), and the like; chromatographic methods such as, but not limited to, gas-chromatography (“GC”), liquid chromatograph (“LC”), high-performance liquid chromatography (“HPLC”), and the like; and combinations thereof (e.g., GC/MS, LC/MS, HPLC/UV-Vis, and the like), and other analytical methods known to persons of ordinary skill in the art.


The component (active ingredients, extract and/or fractions) obtained may be tested for reducing NMRD or symptoms thereof. Exemplary methods for testing the effect are further described herein below as well as in the Examples section which follows.


The active ingredients, extract and/or fraction described herein may be immediately used or stored until further used.


According to specific embodiments, the active ingredients, extract and/or fractions is kept frozen, e.g. in a freezer, until further use (e.g. at about −20° C. to −90° C., at about −70° C. to −90° C., e.g. at −80° C.), for any required length of time.


According to other specific embodiments, the active ingredients, extract and/or fractions is immediately used (e.g. within a few minutes e.g., up to 30 minutes).


The active ingredients, extract and/or fractions may be used separately. Alternatively, different active ingredients, extract and/or fractions (e.g. from different plants or from separate extraction procedures) may be pooled together. Likewise, different active ingredients, extract and/or fractions (from the same extract, from different extracts, from different plants and/or from separate extraction procedures) may be pooled together.


Using the present teachings, the present inventor was able to identify not only plants and extracts that can be used to effectively treat or prevent NMRD, but also active ingredients thereof.


“Active ingredient” refers to a defined chemical composition which is responsible for the anti (preventive or therapeutic) effect against NMRD.


The active ingredient can be purified from a plant or chemically synthesized (artificial, man-made).


Also contemplated herein are analogs and derivatives of the active ingredients as long as the anti (preventive or therapeutic) effect against NMRD is maintained (see e.g., Examples section which follows), which are also referred to as mimetics.


Following are some non-limiting examples for extraction of active ingredients from selected plants of the present invention.


Extraction from leaves of T. capitatus—The Aerial parts of T. capitatus (leaves) samples are collected. Leaves separated from branches are dehydrated at room temperature for 7 days and slightly blended into fine powders for extractions.


Essential oil (EO) extraction—hydro-distillation is used to extract EO from the plant, e.g., dried aerial parts of T. capitatus. In brief, the extraction is conducted for several hours for example, 3 h, by mixing 100 g of plants in 500 mL of distilled water. The extract is dried and concentrated using sodium sulphate and rotatory evaporator under reduced pressure. The EO yield is established by quantity of the obtained oil in mL for 100 g of dried plant. Finally, the pure EO os stored at −4° C. until further analyzed.


Essential oil analysis—The chemical composition of EO is examined by GC and GC-MS. GC analysis is conducted using gas chromatograph. The proportion of the constituents is determined by the integration of peak areas. In addition, mass spectrometry (MS) can be used to analyze the EO typically under the same conditions as described above for gas chromatography. The identification of the different compounds is defined by comparison of their retention indexes (determined relatively to the retention times of a series of n-alkanes) with those of standards of the Wiley library search routines12, based on fit and purity of mass spectra. Such conditions are used for determining the active ingredients as described below.


Extraction from Satujera thymbra:


Air dried aerial parts from S. thymbra were collected in Lebanon at random during April 2009. For 3 h the plant material was submitted to steam distillation using a clevenger-type apparatus to produce the essential oil with a yield of 0.84% (w/w). Oil is dried using anhydrous magnesium sulfate and stored at 4° C. S. thymbra oil was analyzed by GC/MS.


Extraction from Rhus coriaria (Sumac)


In order to isolate, determine and identify the compounds from the Rhus coriaria fruits, different extracts are taken from the fruit or leaves of the Sumac plant. Some are isolated from aqueous extracts, others from alcoholic extracts and some from lipid extracts. Hydrolysable tannins compose the highest percentage in the Sumac fruits, followed by flavonoids. This emphasizes the antioxidant potential of the fruit. Following hydrolysable tannins, comprising almost 20% of the fruit's mass, are other unidentified compounds. Subsequently there are anthocyanins, isoflavonoids, terpenoids and diterpenes. Analysis of the chemical properties of sumac fruit is conducted on ripe fruits and have found a 2.6% protein content, 7.4% fat content, 14.6% fiber content, 1.8% ash. Also, a calorimetric calculation showed that 100 g of sumac fruit contains 147.8 kcal.


Extraction of Thymoquinone from Nigella sativa


Various methods can be used including microwave-assisted extraction system having temperature controlling function as well as other extraction methods, Soxhlet and conventional solid/liquid extraction.












Nigella Sativa


















24-ETHYL-LOPHENOL
Seed Oil




24-METHYL-LOPHENOL
Seed Oil


24-METHYLENE-CYCLOARTANOL
Seed Oil


5-DEHYDRO-AVENASTEROL
Seed Oil


7-DEHYDRO-AVENASTEROL
Seed Oil


ALANINE
Seed
8,000
10,255


ALPHA-SPINASTEROL
Seed Oil



Seed


ARABIC-ACID
Seed


ARACHIDIC-ACID
Seed Oil

1,900


ARACHIDONIC-ACID
Seed Oil

24,900


ARGININE
Seed
41,500
53,050


ASCORBIC-ACID
Leaf

2,577


ASH
Seed
38,000
53,000


ASPARAGINE
Seed

0


ASPARTIC-ACID
Seed
10,670
13,650


ASTRAGALIN
Seed

200


BETA-AMYRIN
Seed Oil


BETA-SITOSTEROL
Seed Oil



Seed

3,218


BUTYROSPERMOL
Seed Oil


CALCIUM
Seed

10,600


CAMPESTANOL
Seed Oil


CAMPESTEROL
Seed Oil



Seed


CARBOHYDRATES
Seed

339,600


CARVONE
Seed



Essent. Oil



Seed
2,250
9,600


CHOLESTEROL
Seed Oil



Seed


CITROSTADIENOL
Seed Oil


CYCLOARTENOL
Seed Oil


CYCLOEUCALENOL
Seed Oil


CYMENE
Seed


CYSTINE
Seed


D-LIMONENE
Seed


DAMASCENINE
Plant


DEHYDROASCORBIC-ACID
Leaf

295


DITHYMOQUINONE
Seed Oil


EICOSADIENOIC-ACID
Seed Oil

25,000



Seed
8,979
10,525


EO
Seed Oil

4,500



Seed
4,000
16,000


FAT
Seed
354,900
416,000


FIBER
Seed

55,000


FIXED-OIL
Seed

380,000


GLUCOSE
Seed


GLUTAMIC-ACID
Seed
28,085
35,900


GLYCINE
Seed
8,840
20,700


GRAMISTEROL
Seed Oil


HEDERAGENIN
Seed

10,000


INDOLE-3-ACETIC-ACID
Tissue

14



Culture


IRON
Seed

140


ISOLEUCINE
Seed
8,570
10,960


KAEMPFEROL-3-O-BETA-GLUCOPYRANOSYL-(1,2)-O-BETA-
Seed

100


GALACTOPYRANOSYL-(1,2)-BETA-GLUCOPYRANOSIDE


LEUCINE
Seed
23,130
29,595


LINOLEIC-ACID
Seed Oil
487,600
576,300



Seed
128,124
233,459


LINOLENIC-ACID
Seed Oil
5,700
7,000



Seed
2,484
2,912


LIPASE
Seed


LOPHENOL
Seed Oil


LYSINE
Seed
16,200
20,700


MELANTHIGENIN
Seed


MELANTHIN
Plant

15,000



Seed

15,000


METHIONINE
Seed
13,100
16,750


MYRISTIC-ACID
Seed Oil
1,900
18,000



Seed
567
1,082


NIGELLICINE
Seed


NIGELLIDINE
Seed


NIGELLIMINE
Seed

0.13


NIGELLIMINE-N-OXIDE
Seed

0.2


NIGELLIN
Seed


NIGELLINE
Seed



Essent. Oil


NIGELLONE
Seed



Essent. Oil



Plant



Seed


OBTUSIFOLIOL
Seed Oil


OLEIC-ACID
Seed Oil
244,600
262,400



Seed
89,911
184,912


PALMITIC-ACID
Seed Oil
120,000
171,200



Seed
22,464
50,523


PALMITOLEIC-ACID
Seed Oil

2,000


PHENYLALANINE
Seed
16,850
21,560


PHYTOSTEROLS
Seed

5,100


POTASSIUM
Seed

5,820


PROLINE
Seed
11,350
14,520


PROTEIN
Seed
210,000
271,900


QUERCETIN-3′-GLUCOSIDE
Seed

440


QUERCETIN-3-O-(6-FERULOYL-BETA-GLUCOPYRANOSYL)-(1,2)-
Seed

240


BETA-GALACTOPYRANOSYL-(1,2)-BETA-GLUCOPYRANOSIDE


QUERCETIN-3-O-BETA-GLUCOPYRANOSYL-(1,2)-O-BETA-
Seed

1,380


GALACTOPYRANOSYL-(1,2)-BETA-GLUCOPYRANOSIDE


RESIN
Seed


RUTIN
Seed

200


SERINE
Seed
4,210
5,385


SODIUM
Seed

980


STEARIC-ACID
Seed Oil
18,100
60,400



Seed
8,722
10,192


STIGMAST-7-EN-3-BETA-OL
Seed Oil


STIGMASTANOL
Seed Oil


STIGMASTEROL
Seed Oil



Seed


TANNIN
Seed


TARAXEROL
Seed Oil


TELFAIRIC-ACID
Seed Oil


THREONINE
Seed
2,615
3,345


THYMOHYDROQUINONE
Seed


THYMOL
Seed Oil


THYMOQUINONE
Seed



Essent. Oil



Seed Oil



Seed


TIRUCALLOL
Seed Oil


TRYPTOPHAN
Seed


TYROSINE
Seed
12,925
16,530


VALINE
Seed
6,500
8,325









According to a specific embodiment, active ingredients (e.g., which can be obtained by supercritical carbon dioxide extraction method) include but are not limited to:


















Compound
RIexp
RIlit
SFE 1
SFE 2
HD SFE
Identification





















n-Nonanea
905
900
0.12


RI, MS


Tricyclene
926
926
tr


RI, MS


Camphene
953
953


1.64
RI, MS


β-Pinene
958
959


0.40
RI, MS


2,4,(10)-Thujadiene
967
960
4.74
0.19

RI, MS


Sabinene
978
977
1.05


RI, MS


β-Myrcene
990
991
0.31


RI, MS


1,8-Cineole
1013
1010


0.98
RI, MS


α-Terpinene
1025
1026
2.34


RI, MS


Limonene
1034
1034
0.18
0.38
1.03
RI, MS


γ-Terpinene
1054
1056
27.46
13.20
12.87
RI, MS


cis-Sabinene hydrate
1063
1068

0.38
Tr
RI, MS


allo-Ocimenola
1079
1071

0.11

RI, MS


Linalool
1087
1080
0.25
0.19

RI, MS


Terpinolene
1091
1088


Tr
RI, MS


trans-Sabinene hydrate
1099
1097
0.37


RI, MS


Terpinen-1-ola
1124
1120


0.11
RI, MS


1,5,8-p-Menthatrienea
1130
1135
0.43
0.38

RI, MS


Borneol
1152
1152


1.02
RI, MS


Pinocarvone
1167
1165
2.96
3.00

RI, MS


trans-Dihydrocarvone
1208
1202

0.19

RI, MS


Dihydrocarvonea
1215
1214
0.37
2.06

RI, MS


Ocimenone (E)a
1249
1239
1.54
1.50

RI, MS


Thymoquinone
1250
1250
35.05
33.12
38.41
RI, MS, NMR


Thymol
1283
1288
7.43
5.30
16.95
RI, MS, NMR


Carvacrol
1299
1299
1.98
1.73
0.81
RI, MS


2-Undecanone
1312
1315


13.72
RI, MS


n-Octyl isobutyratea
1323
1326


0.12
RI, MS


α-Longipinene
1330
1334
0.26


RI, MS


Citronellyl acetatea
1339
1336


0.50
RI, MS


Thymohydroquinone methyl ethera
1353
1351


Tr
RI, MS


Cyclosativene
1367
1366


1.43
RI, MS


α-Longicyclene
1381
1380
0.43
5.25

RI, MS


α-Copaene
1385
1383
1.54
2.00
0.41
RI, MS


α-Longifolene
1391
1387


0.51
RI, MS


(Z)-Caryophyllenea
1395
1395
0.23


RI, MS


β-Caryophyllene
1420
1417
2.89
5.07
4.80
RI, MS


Thymohydroquinone dimethylethera
1429
1425
0.43


RI, MS


Aromadendrenea
1437
1438


1.04
RI, MS


Thymohydroquinone
1515
1509
1.17
1.12
2.31
RI, MS, NMR


Davanonea
1587
1586
0.31


RI, MS


8-Heptadecenea
1683
1680
1.23
1.13
0.86
RI, MS


Dihydrofarnesyl acetatea
1841
1840
2.28
4.69

RI, MS


Pimaradienea
1934
1935
1.23
2.25

RI, MS


Palmitic acid
1947
1946
0.18


RI, MS


Pimara-8(14),15-diene
1968
1966
0.92


RI, MS


Octadecanoic acid
2145
2157
0.26
12.31

RI, MS


Total identified


99.94
95.55
99.92







Group compounds:













Quinones


44.08
39.54
57.67



Monoterpene hydrocarbons


36.51
14.15
15.94


Oxygenated monoterpenes


7.47
9.16
17.14


Sesquiterpene hydrocarbons


5.35
12.32
8.19


Oxygenated sesquiterpenes


2.59
4.69



Diterpenes


2.15
2.25



Alkane


0.12




Alkenes


1.23
1.13
0.86


Fatty acids


0.44
12.31



Fatty acid esters




0.12









Additional plants that are contemplated herein are of the genus Nigella.



Nigella is a genus of 18 species of annual plants in the family Ranunculaceae, native to Southern Europe, North Africa, South Asia, Southwest Asia and Middle East. Common names applied to members of this genus are Nigella, devil-in-a-bush or love-in-a-mist.

    • Nigella arvensis
    • Nigella carpatha
    • Nigella damascena
    • Nigella degenii
    • Nigella deserti
    • Nigella doerfleri
    • Nigella elata
    • Nigella fumariifola
    • Nigella hispanica
    • Nigella latisecta
    • Nigella nigellastrum
    • Nigella orientalis
    • Nigella oxypetala
    • Nigella papillosa
    • Nigella sativa
    • Nigella segetalis
    • Nigella stricta
    • Nigella unguicularis


According to a specific embodiment the active ingredient is thymoquinone.


Additional plants containing thymoquinone include, but are not limited to:

    • Monarda fistulos (of the genus Monarda);
    • Satureja montana (of the genus Satureja);


Additional families containing thymoquinone include, but are not limited to:

    • Asteraceae—examples include, but are not limited to the subfamilies:
      • Barnadesioideae Bremer & Jansen
      • Carduoideae Sweet
      • Cichorioideae Chevallier
      • Corymbioideae Panero & Funk
      • Famatinanthoideae S. E. Freire, Ariza & Panero
      • Gochnatioideae Panero & Funk
      • Gymnarrhenoideae Panero & Funk
      • Hecastocleidoideae Panero & Funk
      • Mutisioideae Lindley
      • Pertyoideae Panero & Funk
      • Stifftioideae Panero
        • Wunderlichioideae Panero & Funk
    • Cupressaceae
      • Cunninghamioideae
      • Taiwanioideae
      • Athrotaxidoideae
      • Sequoioideae
      • Taxodioideae
      • Callitroideae
      • Cupressoideae
      • Incertae sedis
    • Lamiacea
    • Ranunculacea
      • Hydrastidoideae
      • Glaucidioideae
      • Coptoideae
      • Thalictroideae
      • Ranunculoideae


List of plants that contain Carvacrol include, but are not limited to:

    • Monarda didyma
    • Nigella sativa
    • Origanum compactum
    • Origanum dictamnus
    • Origanum microphyllum
    • Origanum onites
    • Origanum scabrum
    • Origanum syriacum
    • Origanum vulgare
    • Plectranthus amboinicus
    • Thymus glandulosus
    • Lavandula multifida
    • Origanum minutiflorum
    • Satureja thymbra












Active ingredients found in Thymus Capitatus












No
RI
Compound
%
















1
935
α-Thujene
0.54



2
940
α-Pinene
0.38



3
991
Myrcene
0.87



4
1019
α.-αterpinene
1.11



5
1025
p-Cymene
6.25



6
1063
γ-Terpinene
6.75



7
1089
α-terpinolene
0.26



8
1101
Linalool
1.51



9
1179
Terpinen-4-ol
1.40



10
1185
4-Carvomenthenol
0.94



11
1260
Geraniol
0.25



12
1309
Carvacrol
65.38



13
1310
Thymol
1.35



14
1358
Eugenol
0.21



15
1408
Carvacryl Acetate
0.45



16
1427
β-Caryophyllene
4.94



17
1461
α-Humulene
0.10



18
1487
allo-aromadendrene
0.18



19
1685
α-Bisabolol
0.35



20
1774
α-Bisabolol oxide A
0.11



21
1815
Hexadecanal
0.14



22
1870
1-Hexadecanol
0.46



23
1879
1-Hexadecanol
0.13



24
1894
Rimuene
0.28



25
1957
Hexadecanoic acid
0.68





Total identified
95.02





Unknown
4.98










Additional plants contemplated herein are of the genus Thymus.


The genus Thymus (/‘custom-character/TY-custom-character; thymes) contains about 350 species of aromatic perennial herbaceous plants and subshrubs to 40 cm tall in the family Lamiaceae, native to temperate regions in Europe, North Africa and Asia.


Stems tend to be narrow or even wiry; leaves are evergreen in most species, arranged in opposite pairs, oval, entire, and small, 4-20 mm long, and usually aromatic. Thyme flowers are in dense terminal heads with an uneven calyx, with the upper lip three-lobed, and are yellow, white, or purple.


Several members of the genus are cultivated as culinary herbs or ornamentals, when they are also called thyme after its best-known species, Thymus vulgaris or common thyme.


About 350 species, including:

    • Thymus adamovicii
    • Thymus altaicus
    • Thymus amurensis
    • Thymus boissieri
    • Thymus bracteosus
    • Thymus broussonetii
    • Thymus caespititius
    • Thymus camphoratus
    • Thymus capitatus
    • Thymus capitellatus
    • Thymus camphoratus
    • Thymus carnosus
    • Thymus cephalotus
    • Thymus cherlerioides
    • Thymus ciliatus
    • Thymus cilicicus
    • Thymus cimicinus
    • Thymus citriodorus (Thymus x citriodorus) syn. T. fragrantissimus, T. serpyllum citratus, T. serpyllum citriodorum.[7]—citrus thyme
    • Thymus comosus
    • Thymus comptus
    • Thymus curtus
    • Thymus decussatus
    • Thymus disjunctus
    • Thymus doerfleri
    • Thymus glabrescens
    • Thymus herba-barona
    • Thymus hirsutus
    • Thymus hyemalis
    • Thymus inaequalis
    • Thymus integer
    • Thymus lanuginosus, syn. T. serpyllum—woolly thyme
    • Thymus leucospermus
    • Thymus leucotrichus
    • Thymus longicaulis
    • Thymus longiflorus
    • Thymus mandschuricus
    • Thymus marschallianus
    • Thymus mastichina
    • Thymus membranaceus
    • Thymus mongolicus
    • Thymus moroderi
    • Thymus nervulosus
    • Thymus nummularis
    • Thymus odoratissimus
    • Thymus pallasianus
    • Thymus pallidus
    • Thymus pannonicus
    • Thymus praecox—creeping thyme
    • Thymus proximus
    • Thymus pseudolanuginosus, syn. T. serpyllum—woolly thyme
    • Thymus pulegioides—lemon thyme[8]
    • Thymus quinquecostatus
    • Thymus richardii
    • Thymus satureioides
    • Thymus serpyllum
    • Thymus sibthorpii
    • Thymus striatus
    • Thymus thracicus—lavender thyme
    • Thymus villosus
    • Thymus vulgaris—common thyme
    • Thymus zygis


List of plants that contain thymol include, but are not limited to:

    • Euphrasia rostkoviana
    • Lagoecia cuminoides
    • Monarda didyma
    • Monarda fistulosa
    • Mosla chinensis, Xiang Ru
    • Origanum compactum
    • Origanum dictamnus
    • Origanum onites
    • Origanum vulgare
    • Satureja thymbra
    • Thymus glandulosus
    • Thymus hyemalis
    • Thymus vulgaris
    • Thymus zygis
    • Trachyspermum ammi


Active ingredients in Thymus vulgaris:
















Plant
Low
High


Chemical
part
ppm
ppm

















1-OCTEN-3-OL
Shoot
150



Shoot
65



Shoot
80


2,5-DIETHYL-TETRAHYDROFURAN
Shoot
0



Shoot
6



Shoot
6


3-OCTANOL
Shoot
110



Shoot
12



Shoot
30


ALPHA-GUAIENE
Shoot
0.1



Shoot
6



Shoot
0


ALPHA-HUMULENE
Shoot
45



Shoot
20



Shoot
55


ALPHA-PHELLANDRENE
Shoot
0



Shoot
40



Shoot
12


ALPHA-PINENE
Shoot
0



Shoot
265



Shoot
325


ALPHA-TERPINENE
Shoot
840



Shoot
990



Shoot
990


ALPHA-TERPINEOL
Shoot
55



Shoot
55



Shoot
25


ALPHA-THUJENE
Shoot
320



Shoot
0



Shoot
0


BETA-CARYOPHYLLENE
Shoot
175



Shoot
185



Shoot
200


BETA-GUAIENE
Shoot
0.1



Shoot
0



Shoot
3


BETA-PHELLANDRENE
Shoot
80



Shoot
60


BETA-PHELLLANDRENE
Shoot
70


BETA-PINENE
Shoot
30



Shoot
30



Shoot
560


BORNEOL
Shoot
55



Shoot
30



Shoot
15


CAMPHENE
Shoot
30



Shoot
25



Shoot
40


CAMPHOR
Shoot
0



Shoot
0.1



Shoot
0


CARVACROL
Shoot
1,285



Shoot
24,850



Shoot
23,765


CARVONE
Shoot
15



Shoot
20



Shoot
0.1


CARYOPHYLLENE-OXIDE
Shoot
75



Shoot
55



Shoot
45


CIS-CARVEOL
Shoot
0



Shoot
0



Shoot
3


CIS-SABINENE-HYDRATE
Shoot
20



Shoot
0



Shoot
55


CITRONELLOL
Shoot
12



Shoot
0.1



Shoot
0


CITRONELLOL-BUTYRATE
Shoot
0


CITRONELLYL-BUTYRATE
Shoot
0



Shoot
15


DIHYDROCARVONE
Shoot
0



Shoot
0



Shoot
12


EHTYL-CINNAMATE
Shoot
0


EO
Shoot
31,000



Shoot
31,000



Shoot
31,000


ETHYL-CINNAMATE
Shoot
0



Shoot
30


GAMMA-TERPINENE
Shoot
2,700



Shoot
1,015



Shoot
240


GERANIOL
Shoot
0



Shoot
0



Shoot
65


GERANYL-ACETATE
Shoot
0



Shoot
0



Shoot
15


GERANYL-BUTYRATE
Shoot
0



Shoot
20



Shoot
0


GERANYL-HEXANOATE
Shoot
0



Shoot
0



Shoot
6


GERANYL-PROPIONATE
Shoot
0



Shoot
0



Shoot
70


GERMACRENE-D
Shoot
0



Shoot
0



Shoot
50


LIMONENE
Shoot
110



Shoot
55



Shoot
90


LINALOL
Shoot
35



Shoot
55



Shoot
25


METHYL-2-METHYL-BUTYRATE
Shoot
6



Shoot
12



Shoot
9


MYRCENE
Shoot
750



Shoot
565



Shoot
0.1


P-CYMENE
Shoot
4,445



Shoot
1,880



Shoot
3,135


TERPINEN-1-OL
Shoot
6



Shoot
15



Shoot
0


TERPINEN-4-OL
Shoot
435



Shoot
315



Shoot
335


TERPINOLENE
Shoot
0



Shoot
0.1



Shoot
45


THYMOL
Shoot
18,560



Shoot
385



Shoot
280


TRANS-BERGAMOTENE
Shoot
9



Shoot
9


TRANS-BERGAMOTTENE
Shoot
9


TRANS-SABINENE-HYDRATE
Shoot
25



Shoot
120



Shoot
0


TRICYCLENE
Shoot
0



Shoot
0



Shoot
3









Active ingredients on the EO of Thymus vulgaris according to some embodiments of the invention, include, but are not limited to:















No.
RT (min)
Area % of total
Constituents*


















1
5.39
1.06
alpha-Thujene


2
5.63
1.07
alpha-Pinene


3
6.89
0.37
beta-Pinene


4
6.97
1.53
beta-Myrcene


5
7.53
0.33
alpha-Phellandrene


6
7.77
3.76
Carene<δ-2->


7
8.04
0.29
D-Limonene


8
8.26
0.21
beta-Phellandrene


9
8.46
8.41
para-Cymene


10
8.96
30.90
gamma-Terpinene


11
9.48
0.47
Terpineol


12
12.55
0.46
Terpinen-4-ol


13
16.17
47.59
Thymol


14
17.32
2.68
Caryophyllene


15
19.03
0.78
Cyclohexene, 1-methyl-4-(5-





methyl-1-methylene-4-hexeny])


Total

99.91%









Active Ingredients of Satujera thymbra:


Air dried aerial parts from S. thymbra were collected in Lebanon at random during April 2009. For 3 h the plant material was submitted to steam distillation using a clevenger-type apparatus to produce the essential oil with a yield of 0.84% (w/w). Oil was dried using anhydrous magnesium sulfate and stored at 4° C. S. thymbra oil are analyzed by GC/MS. Nineteen compounds representing 98.8% of the oil sample are identified. The major components of Satureja thymbra L. oil are γ-terpinene (34.06%), carvacrol (23.07%) and thymol (18.82%). Also abundant are ρ-cymene (7.58%), caryophyllene (3.96%), α-terpinene (3.53%) and myrcene (1.70%).


Also contemplated herein are plants of the genus Satujera.



Satureja is a genus of aromatic plants of the family Lamiaceae, related to rosemary and thyme. It is native to North Africa, southern and southeastern Europe, the Middle East, and Central Asia. A few New World species were formerly included in Satureja, but they have all been moved to other genera. Several species are cultivated as culinary herbs called savory, and they have become established in the wild in a few places.


Examples include, but are not limited to:

    • Satureja adamovicii Šilic—Balkans
    • Satureja aintabensis P. H. Davis—Turkey
    • Satureja amani P. H. Davis—Turkey
    • Satureja atropatana Bunge—Iran
    • Satureja avromanica Maroofi—Iran
    • Satureja bachtiarica Bunge—Iran
    • Satureja boissieri Hausskn. ex Boiss.—Turkey, Iran
    • Satureja bzybica Woronow—Caucasus
    • Satureja x caroli-paui G. López—Spain (S. innota x S. montana)
    • Satureja cilicica P. H. Davis—Turkey
    • Satureja coerulea Janka—Bulgaria, Romania, Turkey
    • Satureja cuneifolia Ten—Spain, Italy, Greece, Albania, Yugoslavia, Iraq
    • Satureja x delpozoi Sánchez-Gómez, J. F. Jiménez & R. Morales—Spain (S. cuneifolia x S. intricata var. gracilis)
    • Satureja edmondii Briq.—Iran
    • Satureja x exspectata G. López—Spain (S. intricata var. gracilis x S. montana)
    • Satureja fukarekii Šilic—Yugoslavia
    • Satureja hellenica Halácsy—Greece
    • Satureja hortensis L.
    • Satureja horvatii Šilic—Greece, Yugoslavia
    • Satureja icarica P. H. Davis—Greek Islands
    • Satureja innota (Pau) Font Quer—Spain
    • Satureja intermedia C. A. Mey.—Iran, Caucasus
    • Satureja intricata Lange—Spain
    • Satureja isophylla Rech. f.—Iran
    • Satureja kallarica Jamzad—Iran
    • Satureja kermanshahensis Jamzad—Iran
    • Satureja khuzistanica Jamzad—Iran
    • Satureja kitaibelii Wierzb. ex Heuff.—Bulgaria, Romania, Yugoslavia
    • Satureja laxiflora K. Koch—Iran, Iraq, Turkey, Caucasus
    • Satureja linearifolia (Brullo & Furnari) Greuter—Cyrenaica region of Libya
    • Satureja macrantha C. A. Mey.—Iran, Iraq, Turkey, Caucasus
    • Satureja metastasiantha Rech. f.—Iraq
    • Satureja montana L.—winter savory—southern Europe, Turkey, Syria
    • Satureja mutica Fisch. & C. A. Mey.—Caucasus, Iran, Turkmenistan
    • Satureja nabateorum Danin & Hedge—Jordan
    • Satureja x orjenii Šilic—Yugoslavia (S. horvatii x S. montana)
    • Satureja pallaryi J. Thiébaut—Syria
    • Satureja parnassica Heldr. & Sart. ex Boiss.—Greece, Turkey
    • Satureja pilosa Velen.—Italy, Greece, Bulgaria
    • Satureja rumelica” Velen.—Bulgaria
    • Satureja sahendica Bornm.—Iran
    • Satureja salzmannii (Kuntze) P. W. Ball—Morocco, Spain
    • Satureja spicigera (K. Koch) Boiss.—Turkey, Iran, Caucasus
    • Satureja spinosa L.—Turkey, Greek Islands including Crete
    • Satureja subspicata Bartl. ex Vis.—Austria, Yugoslavia, Albania, Bulgaria, Italy
    • Satureja taurica Velen.—Crimea
    • Satureja thymbra L.—Libya, southeastern Europe from Sardinia to Turkey; Cyprus, Lebanon, Palestine
    • Satureja thymbrifolia Hedge & Feinbrun—Israel, Saudi Arabia
    • Satureja visianii Šilic.—Yugoslavia
    • Satureja wiedemanniana (Avé-Lall.) Velen.—Turkey












Active ingredients of Thymbra spicata:












Compounds
%
RI1
RT2







α-pinene
0.56
1028
3.64



α-phellandrene
0.64
1033
3.71



camphene
0.06
1073
4.36



β-pinene
0.10
1113
5.16



δ-3-carene
0.05
1155
6.10



β-myrcene
1.04
1170
6.51



α-terpinene
1.48
1184
6.90



dl-limonene
0.17
1202
7.43



β-phellandrene
0.12
1212
7.69



γ-terpinene
10.73
1252
8.86



p-cymene
12.18
1276
9.69



α-terpinolene
0.05
1286
10.04



oct-1-en-3-ol
0.11
1454
16.17



trans sabinene hydrate
0.05
1465
16.59



cis sabinene hydrate
0.03
1547
19.73



linalool
0.03
1551
19.91



trans caryophyllene
1.28
1589
21.39



4-terpineol
0.53
1598
21.79



isoborneol
0.21
1694
25.36



d-carvone
0.02
1728
26.55



anethole
0.04
1826
30.05



caryophyllene oxide
0.65
1968
34.87



spathulenol
0.15
2125
39.56



thymol
2.77
2218
41.80



carvacro
66.86
2239
42.61



naphthalene3
0.08
2281
44.26








1RT—retention time;





2RI—retention index;





3naphthalene, 1,2,3,4,4a,5,6,7-octahydro-4a-methyl







Also contemplated herein are plants of the genus Thymbra.



Thymbra, common name Mediterranean thyme, is a genus of plants in the family Lamiaceae. As currently categorized, the genus has seven species and one subspecies. It is native to the Mediterranean region of southern Europe, North Africa, and the Middle East.


Examples include, but are not limited to:

    • Thymbra calostachya (Rech. f.) Rech. f.—Crete
    • Thymbra capitata (L.) Cav.—widespread from Morocco+Portugal to Turkey+Palestine
    • Thymbra sintenisii Bornm. & Azn.—Iraq, Turkey
    • Thymbra spicata L.—Greece, Turkey, Syria, Lebanon, Palestine, Israel, Iraq, Iran
    • Thymbra thymbrifolia (Hedge & Feinbrun) Bräuchler, comb. nov.—Israel, Palestine, Judean Desert, Khirbet el Mird
    • Thymbra nabateorum (Danin & Hedge) Bräuchler, comb. nov.—W of Jordan and the adjacent N of Saudi Arabia
    • Thymbra linearifolia (Brullo & Furnari) Bräuchler, comb. nov.—Libya


Chemical Composition of Rhus coriaria (Sumac)


Characterization and identification of chemical compounds of Sumac using HPLC-MS method identified 191 compounds in Rhus coriaria and classified them as generally being:

    • 78 hydrolysable tannins (e.g., gallotannins, e.g., penta, hexa, hepta, octa, nona and decagalloyl-glucoside)
    • 59 flavonoids (e.g., Quercetin, Myrecetin 3-rhamnoside and Quercetin 3-glucoside)
    • 9 anthocyanins (e.g., Delphidin-3-glucoside, Cyanidin 3-(2″-galloyl)galactoside, Cyanidin-3-glucoside, 7-methyl-cyanidin-3-(2″galloyl)galactoside, 7-methyl-cyanidin-3-galactoside)
    • 2 isoflavonoids
    • 2 terpenoids
    • 1 diterpene
    • 38 other unidentified compounds.


According to specific embodiments, the phenolic compounds in Sumac are the compounds that constitute its phytochemical activity along with anthocyanins. The most abundant phenolic compound in sumac fruits was found to be Gallic acid.


Hydrolysable tannins compose the highest percentage in the Sumac fruits, followed by flavonoids. This emphasizes the antioxidant potential of the fruit, a plant part contemplated herein as a specific embodiment. Following hydrolysable tannins, comprising almost 20% of the fruit's mass, are other unidentified compounds. Subsequently there are anthocyanins, isoflavonoids, terpenoids and diterpenes. The chemical properties of sumac fruit is conducted on ripe fruits and have found a 2.6% protein content, 7.4% fat content, 14.6% fiber content, 1.8% ash. Also, a calorimetric calculation showed that 100 g of sumac fruit contains 147.8 kcal.


Hydrolysable tannins compose the highest percentage in the Sumac fruits, followed by flavonoids. This emphasizes the antioxidant potential of the fruit. Following hydrolysable tannins, comprising almost 20% of the fruit's mass, are other unidentified compounds. Subsequently there are anthocyanins, isoflavonoids, terpenoids and diterpenes. The chemical properties of sumac fruit is conducted on ripe fruits and have found a 2.6% protein content, 7.4% fat content, 14.6% fiber content, 1.8% ash. Also, a calorimetric calculation showed that 100 g of sumac fruit contains 147.8 kcal.


Other active ingredients or any combinations thereof include, but are not limited to, methyla gallate, gathisflavone, sumaflavone, hinfikflavone, photocatechuic acid, penta-galloylglucose, hinokiflavone, β-caryophyllene, Delphidin-3-glucoside, Cyanidin 3-(2″-galloyl)galactoside, Cyanidin-3-glucoside, 7-methyl-cyanidin-3-(2″galloyl)galactoside, 7-methyl-cyanidin-3-galactoside, quercetin-3-glucoside, kampferol, myricetin, butein, D-limonine.


According to a specific embodiment, the active ingredient or combination thereof includes a volatile compound, e.g., terpene hydrocarbons, monoterpene and sesquiterpene hydrocarbons, specifically β-caryophyllene and α-pinene, Coririanaphthyl ether, Coriarioic acid and Coriariacthracenyl ester.


According to a specific embodiment, the active ingredient or combination thereof includes a fatty acid, e.g., oleic acid, linoleic acid, palmitic acid, β-caryophillene, cembrene stearic acid, Myristic acid, α-linolenic acid.


According to a specific embodiment, the active ingredient or combination thereof includes a mineral, e.g., potassium, calcium, magnesium, phosphorus, aluminum, iron, sodium, boron, zinc, cadmium, selenium.


According to a specific embodiment, the active ingredient or combination thereof includes a vitamin, e.g., thiamin B1, riboflavin B2, pyridoxine B6, cyanocobalamin B12, nicotinamide, biotin and ascorbic acid.


According to a specific embodiment, a methanol or ethanol extract is performed, e.g., ethanol concentration is 80%; extraction time is 1 h; extraction temperature is 40° C.; particle size 1.0 mm; and solvent to sumac ratios 15:1 ml/g. Other extraction procedures include, but are not limited to, those described in Sakhr and Khatib Heliyon. 2020 January; 6(1): e03207, which is hereby incorporated by reference in its entirety.


According to another embodiment, the plant part is leaf.


Also contemplated herein are plants of the genus Rhus.


Examples include, but are not limited to:


Asia and Southern Europe






    • Rhus chinensis Mill.—Chinese sumac


    • Rhus coriaria—Tanner's sumac


    • Rhus delavayi Franchet





Australia, Pacific






    • Rhus taitensis Guill. (Northeast Australia, Malesia, Micronesia, French Polynesia)


    • Rhus sandwicensis A. Gray—neneleau (Hawaii)





North America






    • Rhus aromatica—fragrant sumac


    • Rhus copallinum—winged or shining sumac


    • Rhus glabra—smooth sumac


    • Rhus integrifolia—lemonade sumac


    • Rhus kearneyi—Kearney sumac


    • Rhus lanceolata—prairie sumac

    • Rhus malloryi Wolfe & Wehr—Ypresian, Washington


    • Rhus michauxii—Michaux's sumac


    • Rhus microphylla—desert sumac, littleleaf sumac


    • Rhus ovata—sugar sumac

    • Rhus republicensis Flynn, DeVore, & Pigg-Ypresian, Washington

    • Rhus rooseae Manchester—Middle Eocene, Oregon


    • Rhus trilobata Nutt.—skunkbush sumac


    • Rhus typhina—staghom sumac


    • Rhus virens Lindh. ex A. Gray—evergreen sumac





Chemical Composition of Panax ginseng (ginseng)


Characterization and identification of chemical compounds of ginseng using a variety of methods identified a large variety of compounds in Panax ginseng and classified them as generally being:

    • Saponin Glycosides (e.g., ginsenosides)
    • Phytosterols (e.g. stigmasterol, beta-sterol)
    • Sesquiterpenes (e.g. beta-alamene and beta-selinine)
    • Flavenoids (e.g. Kaempferol)
    • Polyacetylenes (e.g. panaxynol, ginsenoyne A)
    • Alkaloids (e.g. fumarine, girinimbin)
    • Polysaccharides
    • Phenolic compounds (e.g. elemicin, dauricin, maltol).


According to specific embodiments, the saponin compounds in ginseng and the polysaccharide compounds are the compounds that constitute its phytochemical activity. The most abundant saponin compound in ginseng root was found to be ginsenoside. Polysaccharides from ginseng have been identified as NGP, WGP, 1-KGP, 4-KGP, WGPE and EGP, with WGP and WGPE being the most abundant, depending on the species of ginseng plant material used for extraction.


Most ginseng saponins belong to a family of steroids with a four trans-ring rigid steroid skeleton. They are also referred to as ginsenosides, triterpenoid saponins or dammarane derivatives. More than 200 saponins have been isolated from ginseng plants. In addition to ginseng root, saponins have been identified in ginseng leaves and stems, flower buds, fruits, berries, and seeds. Because steaming or heating changes the saponin profile of ginseng products, ginseng saponins have also been identified in the processed root, leaf, flower-bud and berry.



Ginseng saponins are divided into several groups. Two major groups are the protopanaxadiol (PPD)-type saponins with sugar moieties attached to the C-3 and/or C-20 and the protopanaxatriol (PPT) group with sugar moieties at C-6 and/or at C-20. Other groups include the ocotillol-type with a five-membered epoxy ring at C-20, the oleanane-type with a nonsteroidal structure, and the dammarane type with a modified C-20 side chain. As techniques are developed for chemical purification and structural identification, novel ginseng saponins continue to be discovered.


The table below shows ginsenoside compounds recovered from ginseng extracts prepared by different extraction procedures:












GINSENOSIDES












Solvent systema

Obtained



Material
(volume ratio)
Detectionb
compound
Isolation efficiencyc






P.

Hex-n-BuOH-H2O
TLC
Ginsenosides Rb1,
157, 13, 56, and 17 mg of



notoginseng,

(3:4:7)

Re, Rg1 and
Rb1, Re, Rg1 and R1


root


notoginsenoside R1
from 283 mg MeOH






extract of five tablets



P. ginseng,

CH2Cl2—MeOH—NH4OAc—iPrOH
ELSD
Ginsenosides Rf, Rd,
10.7, 11.0, 13.4 and 13.9


root


Re, and Rb1
mg of Rf, Rd, Re and



(6:2:4:3)


Rb1 from 480 mg






enriched fraction by






macroporous resin



P.

CHCl3—MeOH-2-BuOH—H2O
ELSD
Ginsenosides Rg1,
Not provided



notoginseng,

(5:6:1:4 )

Rd, Re, Rb1 and


root
EtOAc-n-BuOH-H2O

notoginsenoside R1



(1:1:2)


Red P.
CH2Cl2—MeOH—H2O—iPrOH
ELSD
Ginsenosides Rg3,
32.2, 26.6, 28.6 and 8.1



ginseng,

(6:6:4:1)

Rk1, Rg5 and F4
mg of Rg3, Rk1, Rg5 and


steamed root



F4 from 350 mg enriched






fraction by RP-






C18 column



P. ginseng,

EtOAc—iPrOH—
UV
Ginsenoside Ro
61 mg Ro from 100 mg


root
0.1% formic acid


enriched sample by



H2O (3:1:5)


normal-phase MPLC






aAbbreviations: Hex: n-hexane; BuOH: butanol; CH2Cl2: methylene chloride; MeOH: methanol; NH4OAc: ammonium acetate; iPrOH: isopropanol; CHCl3: chloroform; EtOAc: ethyl acetate.




bAbbreviations: TLC: thin layer chromatography; ELSD: evaporative light scattering detection; UV: ultraviolet.




cAbbreviations: RP: reversed-phase; MPLC: medium-pressure liquid chromatography.







The table below shows the chemical formulae of 123 dammarane-type saponins isolated from various parts of Panax plants. They are placed in the order of the structure type.












Dammarane - type saponin ginsenosides










No.
Name
Formula
Plant Material













1
Floralginsenoside M
C53H90O22
Flower buds of P. ginseng


2
Floralginsenoside N
C53H90O22
Flower buds of P. ginseng


3
Floralquinquenoside E
C53H90O22
Flower buds of P. quinquefolius


4
Ginsenoside Rh5
C37H64O9
Roots and rhizomes of P.






vietnamensis



5
Notoginsenoside FP 1
C47H80O18
Fruit pedicels of P. notoginseng


6
Notoginsenoside M
C48H82O19
Roots of P. notoginseng


7
Notoginsenoside N
C48H82O19
Roots of P. notoginseng


8
Notoginsenoside Rw1
C46H78O17
Rhizomes of P. notoginseng


9
Notoginsenoside T3
C38H66O9
Acid hydrolysate roots of P.






notoginseng



10
Notoginsenoside U
C42H72O14
Roots of P. notoginseng


11
Quinquenoside L17
C47H80O18
Leaves and stems of P. quinquefolius


12
Yesanchinoside D
C44H74O15
Underground part of P. japonicus


13
Yesanchinoside E
C54H92O23
Underground part of P. japonicus


14
Yesanchinoside F
C56H94O24
Underground part of P. japonicus


15
20(S)-acetylated Rg2
C44H74O14
Roots of P. quinquefolius


16
20(R)-acetylated Rg2
C44H74O14
Roots of P. quinquefolius


17
Malonylginsenoside Ra3
C62H102O30
Fresh roots of P. ginseng


18
Malonylnotoginsenoside R4
C62H102O30
Roots of P. ginseng


19
Notoginsenoside FP2
C58H98O26
Fruit pedicels of P. notoginseng


20
Notoginsenoside FT1
C47H80O17
Acid hydrolysate roots of P.






notoginseng



21
Notoginsenoside L
C53H90O22
Roots of P. notoginseng


22
Notoginsenoside O
C52H88O21
Flower buds of P. notoginseng


23
Notoginsenoside P
C52H88O21
Flower buds of P. notoginseng


24
Notoginsenoside Q
C63H106O30
Flower buds of P. notoginseng


25
Notoginsenoside S
C63H106O30
Flower buds of P. notoginseng


26
Notoginsenoside T
C64H108O31
Flower buds of P. notoginseng


27
Quinquenoside L10
C47H80O17
Leaves and stems of P. quinquefolius


28
Quinquenoside L14
C47H80O17
Leaves and stems of P. quinquefolius


29
Yesanchinoside J
C61H102O28
Underground part of P. japonicus


30
Floralginsenoside A
C42H72O16
Flower buds of P. ginseng


31
Floralginsenoside C
C41H70O15
Flower buds of P. ginseng


32
Floralginsenoside H
C50H84O21
Flower buds of P. ginseng


33
Floralginsenoside J
C48H82O20
Flower buds of P. ginseng


34
Floralginsenoside Ka
C36H62O11
Flower buds of P. ginseng


35
Floralginsenoside Tc
C53H90O24
Flower buds of P. ginseng


36
Floralquinquenoside B
C42H72O15
Flower buds of P. quinquefolius


37
Floralquinquenoside D
C42H72O15
Flower buds of P. quinquefolius


38
Floranotoginsenoside B
C53H90O24
Flowers of P. notoginseng


39
Floranotoginsenoside C
C53H90O24
Flowers of P. notoginseng


40
Ginsenoside I
C48H82O20
Flower buds of P. ginseng


41
Ginsenoside II
C48H82O20
Flower buds of P. ginseng


42
Ginsenoside SL1
C36H62O11
Steamed leaves of P. ginseng


43
Floralginsenoside B
C42H72O16
Flower buds of P. ginseng


44
Floralginsenoside D
C41H70O15
Flower buds of P. ginseng


45
Floralginsenoside E
C42H72O15
Flower buds of P. ginseng


46
Floralginsenoside F
C42H72O15
Flower buds of P. ginseng


47
Floralginsenoside G
C50H84O21
Flower buds of P. ginseng


48
Floralginsenoside I
C48H82O20
Flower buds of P. ginseng


49
Floralginsenoside K
C48H82O21
Flower buds of P. ginseng


50
Floralginsenoside O
C53H90O24
Flower buds of P. ginseng


51
Floralginsenoside P
C53H90O23
Flower buds of P. ginseng


52
Floralquinquenoside A
C36H62O11
Flower buds of P. quinquefolius


53
Floralquinquenoside C
C42H72O15
Flower buds of P. quinquefolius


54
Ginsenoside Rh6
C36H62O11
Leaves of P. ginseng


55
Floralginsenoside La
C48H82O19
Flower buds of P. ginseng


56
Floralginsenoside Lb
C48H82O19
Flower buds of P. ginseng


57
Floranotoginsenoside D
C53H90O23
Flowers of P. notoginseng


58
Ginsenoside Rg7
C36H60O9
Leaves of P. ginseng


59
Notopanaxoside A
C36H62O10
Roots of P. notoginseng


60
Notoginsenoside FT3
C47H80O18
Acid hydrolysate roots of P.






notoginseng



61
Floranotoginsenoside A
C53H90O23
Flowers of P. notoginseng


62
Ginsenoside ST2
C36H62O10
Steamed leaves of P. ginseng


63
Notoginsenoside Rw2
C41H70O14
Rhizomes of P. notoginseng


64
Notoginsenoside ST5
C47H80O18
Steamed roots of P. notoginseng


65
Yesanchinoside H
C53H90O23
Underground part of P. japonicus


66
Ginsenoside Ki
C36H62O10
Leaves of P. ginseng


67
Ginsenoside Km
C36H62O10
Leaves of P. ginseng


68
Quinquenoside L2
C48H82O19
Leaves and stems of P. quinquefolius


69
Dammar-25(26)-ene-3,6,12,20,22,24-
C30H52O6
Leaves of P. ginseng



hexanol


70
Floralginsenoside Kb
C45H76O19
Flower buds of P. ginseng


71
Floralginsenoside Kc
C45H76O20
Flower buds of P. ginseng


72
Floralginsenoside Ta
C36H60O10
Flower buds of P. ginseng


73
Vina-ginsenoside R25
C42H70O15
Roots and rhizomes of P.






vietnamensis



74
Floralginsenoside Tb
C35H62O11
Flower buds of P. ginseng


75
Quinquenoside L9
C42H74O15
Leaves and stems of P. quinquefolius


76
Quinquenoside L16
C54H94O25
Leaves and stems of P. quinquefolius


77
25-OCH3-PPD
C31H56O4
Leaves of P. notoginseng


78
25-OH-PPD
C30H54O4
Fruits of P. ginseng


79
25-OH-PPT
C30H54O5
Fruits of P. ginseng


80
Notoginsenoside FT2
C47H82O18
Acid hydrolysate roots of P.






notoginseng



81
Notoginsenoside T4
C36H62O11
Acid hydrolysate roots of P.






notoginseng



82
Quinquenoside L1
C48H80O18
Leaves and stems of P. quinquefolius


83
Quinquefoloside La
C54H92O23
Leaves of P. quinquefolius


84
Quinquefoloside Lc
C54H92O23
Leaves of P. quinquefolius


85
Dammar-(E)-20(22)-ene-3,12,25-triol
C30H52O3
Acid hydrolysate roots of P. ginseng


86
Notoginsenoside ST1
C36H62O10
Steamed roots of P. notoginseng


87
Ginse noside Rg6
C42H70O12
Stem-leaves of P. ginseng


88
Ginsenoside Rs4
C42H70O12
Steamed roots of P. notoginseng


89
Ginsenoside Rs6
C42H70O12
Steamed roots of P. notoginseng


90
Isoginsenoside Rh3
C36H60O7
Fruits of P. ginseng


91
Ginsenoside Rh5
C36H60O9
Leaves of P. ginseng


92
Ginsenoside SL2
C42H70O14
Steamed leaves of P. ginseng


93
Ginsenoside ST1
C36H60O10
Steamed leaves of P. ginseng


94
Notoginsenoside ST2
C43H74O15
Steamed roots of P. notoginseng


95
Notoginsenoside ST3
C43H74O15
Steamed roots of P. notoginseng


96
Ginsenoside Rg8
C42H70O12
Roots of P. quinquefolius


97
Notoginsenoside T1
C36H60O10
Acid hydrolysate roots of P.






notoginseng



98
Notoginsenoside T2
C36H62O10
Acid hydrolysate roots of P.






notoginseng



99
Ginsenoside Rg1-12,23-epoxy
C42H70O14
Leaves of P. ginseng


100
Ginsenoside Rh9
C36H60O9
Leaves of P. ginseng


101
Quinquefoloside-Lb
C53H88O22
Leaves of P. quinquefolius


102
Ginsenoside Rkl
C42H70O12
Processed roots of P. ginseng


103
Ginsenoside Rk2
C36H60O7
Processed roots of P. ginseng


104
Ginsenoside Rk3
C36H60O8
Processed roots of P. ginseng


105
Ginsenoside Rs5
C38H62O9
Steamed roots of P. notoginseng


106
Ginsenoside Rs7
C38H62O9
Steamed roots of P. notoginseng


107
Notoginsenoside T5
C41H68O12
Acid hydrolysate roots of P.






notoginseng



108
Ginsenoside Rzl
C42H70O12
Steamed roots of P. notoginseng


109
Ginsenoside SL3
C42H70O14
Steamed leaves of P. ginseng


110
Ginsenoside Rh8
C36H60O9
Leaves of P. ginseng


111
Ginsenoside Rh7
C36H60O9
Leaves of P. ginseng


112
Yesanchinoside G
C53H88O23
Underground part of P. japonicus


113
Yesanchinoside I
C59H100O26
Underground part of P. japonicus


114
Hexanordammaran
C24H40O4
Leaves of P. ginseng


115
Notoginsenoside R10
C30H50O9
Steamed leaves of P. ginseng


116
Yesanchinoside A
C44H74O16
Underground part of P. japonicus


117
Yesanchinoside B
C48H82O20
Underground part of P. japonicus


118
Yesanchinoside C
C47H80O19
Underground part of P. japonicus


119
Panaxadione
C30H48O5
Seeds of P. ginseng


120
Polyacetyleneginsenoside Ro
C65H100O21
Roots of P. ginseng


121
Isodehydroprotopanaxatriol
C30H50O3
Acid hydrolysate roots of P. ginseng


122
20,25-epoxy-dammaran-2-en-6,12-diol
C30H50O3
Acid hydrolysate roots of P. ginseng


123
3-methyl-28-nordammaran-2-en-6,12-diol
C30H50O3
Acid hydrolysate roots of P. ginseng









Analysis of ginseng root (Japanese ginseng) has indicated (per 100 grams root) 0.17 g (0.17%) total fat, 50 mg sodium, 8.82 g (8.82%) total carbohydrates comprising 2.3 g dietary fiber and 3.85 g sugars and 0.71 g (0.71%) protein content. Calorimetric calculation showed that 100 g of ginseng root contains 37 kcal.


According to a specific embodiment, the active ingredient or combination thereof includes a ginsenoside, e.g. a protopanaxadiol (PPD)-type saponin with sugar moieties attached to the C-3 and/or C-20, a protopanaxatriol (PPT) saponin with sugar moieties at C-6 and/or at C-20, an ocotillol-type saponin with a five-membered epoxy ring at C-20, an oleanane-type saponin with a nonsteroidal structure, and a dammarane type saponin. Some specific ginsenosides include, but are not limited to notoginsenosides, yesanchinosides, panaxodione, floralginsenosides and ginsenosides Rg1, Rd, Re, Rb1, R1, Rg3, Rk1, Rf, Rg5, F4, Ro.


According to a specific embodiment, the active ingredient or combination thereof includes a volatile compound, e.g., terpene hydrocarbons, monoterpene and sesquiterpene hydrocarbons, specifically β-alamene and β-selenine.


According to a specific embodiment, the active ingredient or combination thereof includes a phytosterol, e.g., stigmasterol, beta-sterol.


According to a specific embodiment, the active ingredient or combination thereof includes a polyacetylene, e.g., panaxynol, ginsenoyne A.


According to a specific embodiment, the active ingredient or combination thereof includes a flavenoid, e.g., Kaempferol.


According to a specific embodiment, the active ingredient or combination thereof includes an alkaloid, e.g., fumarine, girinimbin.


According to a specific embodiment, the active ingredient or combination thereof includes a polysaccharide, e.g., WGP, KGP-1, KGP-4, WGPE, NGP, EGP.


According to a specific embodiment, the active ingredient or combination thereof includes a phenolic compound, e.g., elemicin, dauricin, maltol.


According to a specific embodiment, the active ingredient or combination thereof includes a mineral, e.g., potassium, calcium, magnesium, phosphorus, aluminum, iron, sodium, boron, zinc, cadmium, selenium.


According to a specific embodiment, the active ingredient or combination thereof includes a vitamin, e.g., vitamin D, vitamin A and vitamin C.


According to a specific embodiment, a methanol or ethanol extract is performed, e.g., ethanol concentration is 80%; extraction time is 24 h; extraction temperature is 80-90° C.; particle size 1.0 mm; and solvent to ginseng ratio of 20:1 ml/g. Other extraction procedures include, but are not limited to, those described in Dong et al. 2017 Phytother Res Aug; 19(8): 684-688, which is hereby incorporated by reference in its entirety.


According to another embodiment, the plant part is leaf.


Also contemplated herein are plants of the genus Panax.


Examples include, but are not limited to:

















Common name and




geographical



Ginseng Species
designation










P.
gensing

Korean ginseng




P.
quinquefolius

American ginseng




P.
notoginseng

Chinese ginseng




P.
japonicas

Japanese ginseng




P.
omiensis

Omei gensing




P.
pseudoginseng

Himalayan ginseng




P.
assamicus

N/A




P.
shangianus

N/A




P.
sinensis

N/A




P.
stipuleanatus

Pingpien ginseng




P.
trifolius

Dwarf ginseng




P.
variabilis

N/A




P.
vietnamensis

Vietnamese ginseng




P.
wangianus

Narrow-leaved




P.
bipinnatifidus

Feather-leaf bamboo




ginseng




P.
sokpayensis

N/A




P.
zingiberensis

Ginger ginseng










Korean ginseng cultivars suitable for use with the present invention include, but are not limited to: Chunpoong, Yunpoong, Gopoong, Sunpoong, Gumpoong, Cheongsun, Sunhyang, Sunun, Sunone, K-1, G-1 and Kowon. Chinese ginseng cultivars suitable for use with the present invention include, but are not limited to Jilin Huangguo Reshen, Jishen 01, Fuxing 01, Fuxing 02, Kangmei 01, Xinkaihe 01, Xinkaihe 02, Zhongnong Huangfengshen and Zhongda Linxiashen.


Chemical Composition of Boswellia Species (Frankincense, Olibanum)


Olibanum, also known as frankincense, is a natural oleo-gum-resin that exudes from tappings in the bark of Boswellia trees. There are approximately 23 species of trees in the genus Boswellia, which grow mainly in Arabia, on the eastern coast of Africa and in India. Characterization and identification of chemical compounds of Olibanum using a variety of methods identified a large variety of compounds in the gum resin of Boswellia tree species and classified them as generally being:

    • Alcohol-soluble resins (e.g. diterpenes, triterpenes)
    • Highly aromatic essential oils (e.g. mono- and sesquiterpenes)
    • Water soluble gums


According to specific embodiments, Olibanum comprises 65-85% alcohol-soluble resins, about 5-9% highly aromatic essential oils and the remainder water soluble gums.


In India, the main commercial sources of Boswellia serrata are Andhra Pradesh, Gujarat, Madhya Pradesh, Jharkhand and Chhattisgarh. Regionally, it is also known by different names. The botanical origin and vernacular names of Boswellia serrata are given in below Table 1. Salai, an oleo gum-resin, is a plant exudate of genus Boswellia (Family: Burseraceae). It is tapped from the incision made on the trunk of the tree, which is then stored in specially made bamboo basket. The semi-solid gum-resin is allowed to remain in the basket for about a month during which its fluid content locally known as ‘ras’ keeps flowing out. The residue, semi-solid to solid part, is the gum-resin which hardens slowly into amorphous, tear-shaped products with an aromatic scent. Then, it is broken into small pieces by wooden mallet or chopper and during this process all impurities including bark pieces etc. are removed manually. The gum-resin is then graded according to its flavour, colour, shape and size. Generally four grades i.e. Superfine, Grade I, Grade II and Grade III are available in the market. The fresh gum obtained from the tree is hot with pleasant flavour and slightly bitter in taste. It had been the ‘frankincense’ of ancient Egyptians, Greeks and Romans who used it as prized incense, fumigant as well as a multipurpose aromatic. It is generally used in making incense powder and sticks.









TABLE 1







BOTANICAL ORIGIN AND VERNACULAR NAMES


OF BOSWELLIASERRATA








Botanical origin
Vernacular names





Division: Spermatophyta
English: Indian Ollbanum text missing or illegible when filed   or



Indian frankinsense


Sub-division: Anglospermae
Hindi: Kundur, Salai


Tribe: Rosopsida
Bengall: Kundur, Salai


Sub-tribe: Rosidae S. Lat.
Gujarati text missing or illegible when filed  : Dhup, Gugall


Over class: Rutanae
Kannada: Chitta, Guguladhuph


Class: Anacardiales
Malayalam text missing or illegible when filed  : Parangl text missing or illegible when filed  , Saambraani


Family: Burseraceae
Tamil: Parangi, Saambraani


Genus: Boswallia
Telugu: Phirangi, Saambraani


Species: serrata
Sanskrit: Ashvamutri, Kundara, Skallaki.






text missing or illegible when filed indicates data missing or illegible when filed







The oleo gum-resins contain 30-60% resin, 5-10% essential oils, which are soluble in the organic solvents, and the rest is made up of polysaccharides (˜65% arabinose, galactose, xylose) which are soluble in water. The resins have a fragrant aroma because of the presence of essential oils and this accounts for their commercial importance.


According to specific embodiments, the common components of Olibanum belonging to the terpene and sesquiterpene families, or their terpenoid derivatives include, but are not limited to α- and β-pinene, α-limonene, myrcene, linalool, α-cubebene, γ-cadinene, β-bourbonene, and α-phellandrene dimer compounds in Olibanum are the compounds that constitute its phytochemical activity. Several oxygenated isoprenoid derivatives have also been identified, such as carbonyl derivatives (e.g., carvone, fenchone) and alcohol-containing terpene and sesquiterpene derivatives (e.g., transpinocarveol, cis-verbenol, and cembrenol), as well as ester-containing compounds (e.g., α-terpinyl acetate and bornyl acetate).


Diverse investigators have reported that limonene is the most abundant volatile in Olibanum, while others have identified octanol acetate, α-pinene and α-thujene as most abundant depending on the species of Boswellia resin used for extraction.


More than 300 essential oils have been isolated from Boswellia ssp.


The table below shows the essential oils recovered from Olibanum extracts prepared by different extraction procedures, from diverse Boswellia ssp.:













Number
Compound
















1
5,5-Dimethyl-1-vinylbicyclo-[2.1.1]-hexane


2
Anethol


3
Benzyl tiglate


4
trans-α-Bergamotene


5
Bornyl acetate


6
β-Bourbonene


7
Cadinene


8
γ-Cadinene


9
Camphene


10
Camphor


11
m-Camphorene


12
p-Camphorene


13
Carene-3


14
(E)-β-Caryophyllene


15
Cembrene A


16
Cembrenol


17
1,8 Cineol


18
Citronellol


19
α-Copaene


20
β-Copaene


21
p-Cymene


22
m-Cymene


23
Elemol


24
Elemicine


25
epi-Cubenol


26
Estragol


27
Eudesmol


28
10-epi-γ-Eudesmol


29
Fenchone


30
Geraniol


31
Germacrene D


32
Humulene epoxide


33
Isoincensole


34
Isomenthone


35
Kessane


36
Limonene


37
Linalool


38
Linalyl acetate


39
Menthone


40
Methylchavicol


41
Methylisoeugenol


42
Methyleugenol


43
γ-Muurolene


44
Myrcene


45
Neocembrene A


46
Nerolidol


47
cis-β-ocimene


48
(Z)-Ocimene


49
(E)-β-Ocimene


50
Perillene


51
α-Phellandrene


52
β-Phellandrene


53
α-Pinene


54
β-Pinene


55
trans-Pinocarveol


56
Sabinene


57
cis-Sabinol


58
Terpinin-4-ol


59
Terpinen-4-ol


60
Terpinolene


61
α-Terpineol


62
α-Terpinene


63
α-Terpinene


64
γ-Terpinene


65
Terpinyl acetate


66
Terpinyl isobutyrate


67
Tetrahydrolinalool


68
α-Thujene


69
α-Thujone


70
β-Thujone


71
Tricyclene


72
Undecenol


73
trans-Verbenol


74
β-Ylangene


75
Zingiberene


76
Abieta-8,12-diene


77
α-Amorphene


78
alloaromadendrene


79
Benzyl benzoate


80
Beyerene


81
Bisabolene


82
Isopentyl-2-methylbutanoate


83
cis-Calamenene


84
α-Cadinene


85
τ-Cadinol


86
2-Carene


87
Campholenealdehyde


88
Caryophyllene oxide


89
cis-Carveol


90
( + ) trans-Carveol


91
Carvone


92
α-Cedrene


93
Cedrol


94
Cembra-1,3,7,11-tetraene


95
Cembra-3,7,11,15-tetraene


96
Cembrene


97
Cembrene C


98
Citronellyl acetate


99
α-Cubebene


100
β-Cubebene


101
o-Cymene


102
Chrysanthenone


103
1,4-Cyclohexadiene


104
p-Cymen-8-ol


105
Decanol


106
Decyl acetate


107
2,6-Dimethoxytoluene


108
3,5-Dimethoxytoluene


109
Duva-3,9,13-trien-1,5α-diol


110
Duva-4,8,13-trien-1a,3α-diol


111
Duva-3,9,13-trien-1,5α-diol-1-acetate


112
Duva-3,9,13-triene-1α-ol-5,8-oxide-1-acetate


113
β-Elemene


114
Farnesyl acetate


115
Geranyl acetate


116
α-Gurjunene


117
Hedycariol


118
1,3,6-Trimethylencycloheptane


119
1-Hexanol


120
Hexyl acetate


121
Hexyl hexanoate


122
α-Humulene


123
Incensole


124
Incensole acetate


125
Isodurene


126
Isocembrene


127
Isophyllocladene (kaur-15-ene)


128
Kaurene


129
Ledol


130
Maaliane


131
p-Mentha-1,5-dien-8-ol


132
o-Methyl anisole


133
α-Muurolene


134
α-Muurolol


135
Myrtenal


136
Naphthalene


137
Naphthalene 1,2,3,4,4a,7-hexahydro-1,6-dimethyl-4-



(1-methylethyl


138
Neryl acetate


139
cis-Nerolidol


140
(S)-trans-Nerolidol


141
(E)-Nerolidol


142
1-Octanol


143
n-Octanol


144
Octanol acetate


145
Octyl acetate


146
Octyl formate


147
allo-Ocimene


148
Phenanthrene-7-ethenyl-9,10,10a-dodeca-hydro-1-1-4a-



7-tetramethyl


149
α-Phellandrene epoxide


150
Phyllocladene


151
α-Pinene-epoxide


152
1-β-Pinene


153
2-β-Pinene


154
Isopinocampheol


155
Piperitone


156
Pyrimidine


157
Sabinyl acetate


158
Sandaracopimara-8(14)-15-diene


159
Sclarene


160
α-Selinene


161
β-Selinene


162
δ-Selinene


163
trans-Terpine


164
4-Terpineol


165
Terpinolene


166
Isoterpinolene


167
2,4(10)-Thujadiene


168
Thujopsene


169
Thunbergol


170
Isomyl-valerate


171
Verticilla-4(20),7,11-triene


172
Verbenone


173
cis-Verbenol


174
Verticiol


175
Viridiflorol


176
Benzene, 1methoxy-2-methyl


177
endo-Borneol


178
γ-Campholene aldehyde


179
α-Campholene aldehyde


180
Cara-2,4-diene


181
Carvacrol


182
Carvotanacetone


183
trans-Dihydrocarvone


184
Cumin alcohol


185
m-Cymene-8-ol


186
p-Cymene-9-ol


187
p-Cymenene


188
Dodecanol


189
Eucalyptol


190
Eucarvone


191
Isopropyl benzaldehyde


192
Isopropyl benzalcohol


193
cis-1,2-Limonene epoxide


194
8,9-Limonene epoxide II


195
8,9-Limonene-epoxide I


196
trans-1,2-Limonene epoxide


197
cis-Linalool oxide


198
trans-Linalool oxide


199
p-Mentha-1,5-diene-7-ol


200
p-Mentha-1,8-diene-4-ol


201
cis-p-Menth-2-en-1-ol


202
cis-p-Mentha-1(7),8-diene-2-ol


203
cis-p-Mentha-2,8-diene-1-ol


204
trans-p-Menth-2-en-1-ol


205
trans-p-Mentha-1(7),8-diene-2-ol


206
trans-p-Mentha-2,8-diene-1-ol


207
2,4(8)-p-Menthadiene


208
p-Mentha-6,8-dien-2-one


209
p-Methylanisole


210
Myrtenol


211
Nerol


212
trans-Ocimene


213
(E)-β-Ocimene epoxide


214
α-Phellandrene-dimer


215
α-Phellandrene-8-ol


216
α-Pinene oxide


217
Pinocamphone


218
Pinocarvone


219
Piperitenone


220
Isopiperitenone


221
trans-Piperitol


222
α-Terpineol


223
Sabina ketone


224
cis-Sabinene hydrate


225
trans-Sabinene hydrate


226
trans-Sabinol


227
2,5-Dimethylstyrene


228
cis-1,2-Epoxy-terpin-4-ol


229
Thuj-3-en-10-al


230
Thujanol


231
Thunbergene


232
Thymol


233
Umbellulone


234
Verticellol


235
5,5-Dimethyl-1-vinylbicyclo-[2.1.1]-hexane


236
p-Anisaldehyde


237
Aromadendrene


238
Benzyl tigilate


239
p-Camphorene


240
Isocaryophyllene


241
Cumaldehyde


242
Cyclosativene


243
γ-Eudesmol


244
Guaioxide


245
5-Guaiene-11-ol


246
Isogermacrene D


247
4-Methylene-1-(1-methylethyl)-bicyclo[3.1.0]hex-2-ene


248
2-Methyl-5-(1-methylethyl)-1,3-cyclohexadiene



monoepoxide


249
n-Pentadecan


250
Perilla alcohol


251
Perillol


252
Thujol


253
m-Thymol


254
α-Ylangene


255
γ-Campholene aldehyde


256
n-Decanoic acid


257
β-Eudesmene


258
β-Cyclogeranylacetate


259
n-Hexanoic acid


260
Hexylcaprylate


261
Incensyl acetate


262
Incensole oxide


263
Incensole oxide acetate


264
Lauric acid


265
p-Methylacetophenone


266
p-Methyleugenol


267
β-Myrcene


268
n-Nonanoic acid


269
n-Octanoic acid


270
3,4-Dimethoxystyrene


271
α-Cadinol


272
1,Hydroxy-1,7-dimethyl-4-isopropyl-2,7-cyclodecadiene


273
1,5,5,8-Tetramethyl-12-oxabicyclo-[9.1.0]-dodeca-3,7-diene


274
1-Methyl-4-(1-methylethenyl)-1,2-cyclohexanediol


275
trans-p-Mentha-2,8-dienol


276
1,2,3,4,6,8a-hexahydro-1-isopropyl-4,7-dimethyl-



naphthalene


277
2-Isopropenyl-4a,8-dimethyl-1,2,3,4,4a,5,6,8a-



ctahydronaphthalene


278
3,5-Dimethoxytoluene


279
(Z)-α-Hydroxymanool


280
Hydroxy-manool


281
Methyl linoleate


282
1-Acetyl-4-isopropenylcyclopentene


283
2,4-Dimethylacetophenone


284
α-Amyrenone


285
β-Amyrenone


286
10-Hydroxy-4-cadinen-3-one


287
2-Hydroxy-1,4-cineole


288
Cryptone


289
Eucarvone


290
Isopropylidencyclohexane


291
1,2,4-Trihydroxy-p-menthane


292
Δ4-p-Menthen-2-one


293
5-Hydroxy-p-menth-6-en-2-one


294
Myrtenoic acid


295
Nopinone


296
3,6,6,-Trimethyl-norpinan-2-one


297
o-Methylacetophenone


298
Perillaaldehyde


299
Phellandra


300
Pinocamphone/isopinocamphone


301
Thujone


302
24-Noroleana-3,12-diene


303
24-Noroleana-3,9(11),12-triene


304
24-Norursa-3,12-diene


305
24-Norursa-3,9(11),12-triene


306
24-Norursa-3.12-dien-11-one


307
α-Amyrine


308
epi-α-Amyrine


309
β-Amyrine


310
Lupeol


311
Terpinenyl acetate


312
1,5-Isopropyl-2-methylbicyclo[3.1.0]hex-3-en-2-ol


313
α-Campholenal


314
(3E,5E)-2,6-Dimethyl-1,3,5,7-octatetraene


315
(E)-2,3-Epoxycarene


316
3,4-Dimethylstyrene


317
1-(2,4-Dimethylphenyl)ethanol


318
4-Methylbenzoic acid


319
p-Menth-1(7)-en-2-one


320
Caryophyllene


321
Methylcycloundecanecarboxylate


322
Nonanoic acid


323
Hexadecanoic acid


324
1,4-Cineol


325
Sabinene hydrate


326
Methyl-trans-2-cis-4-decadienoate


327
2-Hydroxy-5-methoxy-acetophenone


328
(E)-β-Farnesene


329
2-Dodecenoic acid methyl ester


330
Calacorene


331
n-Dodecanoic acid


332
α-Guaiol


333
Caryophylla-3(15),7(14)-dien-6-ol


334
Cadalene


335
Eudesma-4(15),7-dien-1β-ol


336
n-Heptadecane


337
n-Tetradecanoic acid


338
n-Octadecane


339
Galaxolide


340
Manool









Although many Boswellia species produce Olibanum, the major sources of commercial Olibanum are B. serrata (India), B. sacra (Oman), and B carteri (Somalia). The table below shows the major components of Olibanum derived from diverse Boswellia species, according to their percentage representation:















Boswellia

Predominant
Percentage


specie
Source of resin
compound(s)
(%)



















B.
serrata

Commercial (Hamburg, Germany)
Myrcene
38



B.
serrata

NA
α-Thujene
22.7-47.4



B.
serrata

NA
α-Thujene
29.3



B.
serrata

NA
α-Thujene
61.36



B.
carteri

Purchased from the local market of
Duva-3,9,13-triene-
21.4



herbs and spices in Egypt
1a-ol-5,8-oxide-1-





acetate




B.
sacra

Botanically certified oleogum resin
E-β-Ocimene
32.3



B.
carteri/sacra

NM
Octanol acetate
45.2



B.
carteri

Authentic sample from Ethiopia
Octyl acetate
39.3



certified for its authenticity from the





Agricultural Department of the





Ethiopian government





B.
rivae

NA
Limonene
28.0



B.
rivae

Authentic sample from Ethiopia
α-Pinene
16.7



B.
rivae

NA
α-Pinene
13.3



B.
rivae

NA
Octanol
17.8



B.
neglecta

NA
α-Pinene
16.7



B.
neglecta

Authentic sample from Ethiopia
α-Pinene
21.3



B.
papyrifera

NA
Octyl acetate
63.5



B.
papyrifera

NA
Octyl acetate
56.0



B.
pirottae

NA
Trans-Verbenol
15.5



B.
pirottae

NA
Terpinen-4-ol
14.6



B.
frereana

Commercial (Hamburg, Germany)
α-Pinene
38.0









One exemplary analysis of Olibanum has indicated the following components

    • Acid resin (6%), soluble in alcohol and having the formula C20H32O4
    • gum (similar to gum arabic) 30-36%
    • 3-acetyl-beta-boswellic acid (Boswellia sacra)
    • alpha-boswellic acid (Boswellia sacra)
    • incensole acetate, C21H34O3
    • phellandrene


Another analysis of B. serrata resin revealed that the resinous part of Boswellia serrata contains monoterpenes (α-thujene); diterpenes (macrocyclic diterpenoids such as incensole, incensole oxide, iso-incensole oxide, a diterpene alcohol [serratol]); triterpenes (such as α- and β-amyrins); pentacyclic triterpenic acids (boswellic acids); tetracyclic triterpenic acids (tirucall-8,24-dien-21-oic acids). The structures of four major pentacyclic triterpenic acids (boswellic acids) as also some of their characteristic features of four pentacyclic triterpene acids (Boswellic acid) are given in the following table:


















Acetyl-
11-keto-
Acetyl-11-keto-


Properties
B-Boswellic acid
B-Boswellic acid
B-Boswellic acid
B-Boswellic acid







Molecular formula
C text missing or illegible when filed  H text missing or illegible when filed  O text missing or illegible when filed
C text missing or illegible when filed  H text missing or illegible when filed  O text missing or illegible when filed
C text missing or illegible when filed  H text missing or illegible when filed  O text missing or illegible when filed
C text missing or illegible when filed  H text missing or illegible when filed  O text missing or illegible when filed


Molecular weight
456.7
498.74
470.09
512.73


Chemical name
3α-Hydroxy-urs-
3α-Acetoxy-urs-
3α-Hydroxy-urs-12-
3α-Acetoxy-urs-12-



12-en-23-olc acid
12-en-23-olc acid
en-11-keto-23-olc acid
en-11-keto-23-olc acid


Melting point
226-228°
252-255°
195-197°
271-274°


Specific roatation
+106.8°
+138°
+78.5°
+88.5°


UV-MeOH
Maxima at 208 nm
Maxima at 208 nm
Maxima at 250 nm
Maxima at 250 nm


NMR
5.15, CH═C, 4.08,
5.31, CH═C, 5.2,
5.55, CH═C, 4.08,
5.55, CH═C, 5.2,


(In CDCl3, δ ppm)
CH—OH;
CH—OAc;
CH—OH;
CH—OAc;



2.3-1.1, Methylenes and
2.1, COCH3, 1.9-1.25,
2.6-1.4, Methylenes and
2.6-1.4, Methylenes and



methines, 23 protons;
Methylenes and methines,
methines, 21 protons;
methines, 21 protons;



1.1-0.7 Methyls,
23 protons, 1.2-0.7,
1.25-0.75, Methyls 21
1.25-0.75 Methyls,



21 protons
Methyls, 21 protons
protons
21 protons


FTIR
3500 (OH),
1732 (OAc),
3460 (OH),
1740 (Ac), 1701 (COOH),


(In KBr, cm−1)
1699.5 (COOH)
1701 (COOH)
1693 (COOH)
647(α,β-unsaturated






carbonyl)


GC MS
394 (M-68[44 due to
394 (M-104[44 due
408 (M-68[44 due
408 (M-68[44 due



—CO2 and 18
to —CO2 and 60
to —CO2 and
to —CO2 and 18



due to —H2O]);
due to HOAc]);
18 due to —H2O]);
due to HOAc]);



Other fragments;
218 (base peak)
232 (base peak);
232 (base peak); Other



203, 189, 175, 161

Other fragments;
fragments: 217, 175,





217, 175, 161, 135
161, 135






text missing or illegible when filed indicates data missing or illegible when filed







The Olibanum gum component contains polysaccharides and polymeric components. The proteoglycans in Olibanum comprise mainly D-galactose units in the main chain and glucuronic acid, uronic acids, 4-O-methyl-glucuronic acid and arabinose in the side chains.


According to a specific embodiment, the active ingredient or combination thereof includes an alcohol soluble acid resin, a water soluble gum, an alpha-boswellic acid, an incensole acetate and a phellandrene.


According to a specific embodiment, the active ingredient or combination thereof includes a volatile compound, e.g. α-Thujene, Duva-3,9,13-triene-1a-ol-5,8-oxide-1-acetate, E-β-Ocimene, Octanol acetate, Octyl acetate, Limonene, α-Pinene, Octanol, Trans-Verbenol and Terpinen-4-ol.


According to a specific embodiment, the active ingredient or combination thereof includes a mineral, e.g., potassium, calcium, magnesium, phosphorus, aluminum, iron, sodium, boron, zinc, cadmium, selenium.


According to a specific embodiment, a water or alcohol extract is performed.


In some embodiments, the Olibanum is prepared by water extract. An exemplary water extract is described herein:


Preparation of olibanum extract by water. At first, Olibanum is carefully powdered. The powder (25 g) is mixed with 200 ml of deionized water and stirred with 800 rpm overnight at room temperature. This mixture is centrifuged at 1,500 rpm for 10 min and the supernatant collected. Thereafter, the supernatant is again centrifuged at 2,500 rpm for 10 min and successively at 10,000 rpm for 20 min, and then filtered. The filtrates can be stored at −20 C and then freeze-dried −58 C and 0.5 Torr for 24 h to yield 4.02 gr of water soluble extract. At the next step, the resulted powder is dissolved in 100 ml methanol and stirred for 12 hr. at room temperature, then allowed to settle. The precipitate phase is collected and dried in an oven. Again the powder is dissolved in deionized water, centrifuged repeatedly and refiltered. The filtrates can be stored and then freeze-dried.


In some embodiments, the Olibanum is prepared by alcohol extract. An exemplary alcohol extract is described herein:


Preparation of olibanum extract by alcohol: In this method, 100 gr of Olibanum powder with 400 ml of methanol is mixed. This mixture is then stirred at 650 rpm for 24 hours. The resulting mixture is made up of two phases, the upper phase is alcoholic and yellow, and contains substances that are soluble in alcohol. The material is then dried in an oven at 50 C. The bottom phase has a sedimentary and white state, which is set to in the oven until dry. The resulting powder in the water is well dissolved and the obtained solution is centrifuged at 1,500 rpm for 10 min and the supernatant collected. Thereafter, the supernatant is again centrifuged at 2,500 rpm for 10 min and successively at 10,000 rpm for 20 min, and then filtered. The filtrates can be stored at −20 C and then freeze-dried.


Other extraction procedures include, but are not limited to, those described in Mertens et al, et al. 2009, Flavor and Fragrance, 24:279-300 and Hamm et al, Phytochemistry 2005, 66:1499-1514, which are hereby incorporated by reference in their entirety.


Also contemplated herein are Olibarum and other compositions from trees of the genus Boswellia.


Examples include, but are not limited to:












Some Boswellia


Species








B.
socotrana




B.
elongata




B.
ameero




B.
carteri




B.
neglecta




B.
sacra




B.
thurifera




B.
frereana




B.
dioscorides




B.
rivae




B.
papyrifera




B.
serrata










Chemical Composition of Gynostemma pentaphyllum (Jiaogulan)



Gynostemma pentaphyllum is a perennial herb from the Cucurbitaceae family, with 5-lobed leaves and a gourd-like, inedible fruit which grows in forests, thickets or roadsise on mountain slopes in many areas of Northeast and Southeast Asia, including China, Taiwan, S Korea, Japan, Thailand, Vietnam and Laos. G. pentphyllum also grows in Bangladesh, Bhutan, India, Indonesia, Malaysia, Myanmar, Nepal, New Guinea and Sri Lanka. Jiaogulan is prized for its reputation as a “longevity plant”. Characterization and identification of chemical compounds of Gynostemma pentaphyllum using a variety of methods identified a large variety of compounds in Gynostemma pentaphyllum (Thun.) Makino and classified them as generally being:

    • Saponin Glycosides (e.g., gypenosides)
    • Phenolic compounds
    • Flavenoids (e.g. Kaempferol, quercetin, rutin, ombuin, isorahmnetin)
    • Polysaccharides
    • Sterols (e.g. ergostane, cholestane, stigmastane)
    • Trace elements (e.g. Cu, Fe, Zn, Mn, Co, Ni, Se, Mo and Sr)
    • Carotenoids
    • Volatiles (e.g. malonic acid, benzyl-O-beta-D-glucopyranoside, lutein, vomifoliol, palmitic acid, linoleic acid)


According to specific embodiments, the saponin compounds in Jiaogulan and the polysaccharide compounds are the compounds that constitute its phytochemical activity. The most abundant saponin compound in Jiaogulan was found to be gypenoside.


Most Jiaogulan saponins belong to a family of triterpenoid saponins. They are also referred to as gypenosides, and dammarane derivatives. More than 150 saponins have been isolated from G. pentaphyllum plants. Saponins have been identified in Jiaogulan leaves and stems, flower buds, fruits, berries, and seeds.


The table below shows the phytochemical properties of 5 different Gynostemma pentaphyllum samples from different sources:





















TPC
TSC
TFC
RUTIN
QUERCITIN
R + Q




(mg
(mg
(mg
CONTENT
CONTENT
(umol


SAMPLE
SOLVENT
GAE/g)
GE/g)
RE/g)
(ug/g)
(ug/g)
QE/g)






















GP1
50% acetone
44.3
38.02
21.44
3049.5
4906.5
21.2



50% ethanol
37.5
41.39
26.40
7948.2
7431.8
37.6



100% ethanol
33.6
87.28
26.87
11235.4
7279.1
42.5


GP2
50% acetone
14.9
90.17
10.6
2527.3
117.5
4.5



50% ethanol
12.9
114.48
14.27
3588.1
136.2
6.3



100% ethanol
6.9
132.57
13.84
2131.9
166.2
4.0


GP3
50% acetone
12.3
47.62
10.52
8614.9
358.9
15.3



50% ethanol
10.6
59.13
9.51
9954.0
411.0
17.7



100% ethanol
6.7
64.57
8.05
7193.0
549.4
13.6


GP4
50% acetone
43.2
77.64
63.48
1409.2
241.3
3.1



50% ethanol
30.4
82.12
54.04
680.2
150.8
1.6



100% ethanol
17.7
104.1
36.47
579.4
151.3
1.4


GP5
50% acetone
13.1
23.61
14.55
nd
nd



50% ethanol
10.2
60.7
16.53
nd
nd



100% ethanol
8.9
123.97
22.11
nd
nd





GP1-5 represent G. pentaphyllum samples from different sources. Data are per gram of dry botanical basis and are expressed as mean (SD. Different letters represent significant differences (P < 0.05). nd stands for not detectable. TPC, TSC, and TFC stand for total phenolic content, total saponin content, and total flavonoid content by spectrometric methods, respectively. GAE, GE, RE, and QE stand for gallic acid equivalents, gypenoside equivalents, rutin equivalents, and quercetin equivalents. Rutin and quercetin contents were flavonoid profile obtained by HPLC. R + Q stands for total amount of rutin and quercetin.


Ethanol extraction: 12 g sample in 250 ml 100% ethanol, 5 hours in Soxhlet apparatus.


50% acetone extraction and 75% ethanol extraction: 2 g sample in 20 ml solvent at ambient temperature and filtration through 45 micron filter.






Water content of the Jiaogulan samples ranged from 3.79 to 7.57 g/100 g sample. Dietary fiber content ranged from 0.6 g/g to 0.24 g/g sample. Selenium content ranged from 1.7 mg/kg to 0.94 mg/kg.


According to a specific embodiment, the active ingredient or combination thereof includes a gypenoside. Some specific ginsenosides include, but are not limited to CP-1-6.


According to a specific embodiment, the active ingredient or combination thereof includes a volatile compound, e.g., malonic acid, benzyl-O-beta-D-glucopyranoside, lutein, vomifoliol, palmitic acid, linoleic acid.


According to a specific embodiment, the active ingredient or combination thereof includes a phytosterol, e.g., stigmasterol, ergostane.


According to a specific embodiment, the active ingredient or combination thereof includes a flavenoid, e.g., Kaempferol, quercetin, rutin.


According to a specific embodiment, the active ingredient or combination thereof includes a phenolic compound.


According to a specific embodiment, the active ingredient or combination thereof includes a mineral, e.g., potassium, calcium, magnesium, phosphorus, aluminum, iron, sodium, boron, zinc, cadmium, selenium.


According to a specific embodiment, the active ingredient or combination thereof includes a vitamin, e.g., vitamin D, vitamin A and vitamin C.


According to a specific embodiment, a methanol or ethanol extract is performed, e.g., ethanol concentration is 100 or 75%; 5 hours in Soxhlet apparatus, or 50% acetone extraction and 75% ethanol extraction: 2 g sample in 20 ml solvent at ambient temperature and filtration through 45 micron filter. Other extraction procedures include, but are not limited to, those described in Yantao et al. 2016 Chi Med 11:43, which is hereby incorporated by reference in its entirety.


According to another embodiment, the plant part is leaf.


Also contemplated herein are plants of the genus Gynostemma.



Origanum syriacum


According to a specific embodiment, the plants of this species include flavones, monoterpenoids and monoterpenes. Over 60 different compounds have been identified, with the primary ones being carvacrol and thymol ranging to over 80%, while lesser abundant compounds include p-cymene, γ-terpinene, caryophyllene, spathulenol, germacrene-D, β-fenchyl alcohol and δ-terpineol.


The table below shows a profile of the organic compounds identified in Origanum extract through fractional distillation:


Profile of the organic compounds found in the fractions analyzed.
















% de Relative Area
















Compound
Boiling Point ° C.
Code
F1
F2
F3
F4
Unoil
Ooil


















α-thujene
150-152
MH1
5.03
0.389
ND
ND

1.74


α-pinene
156
MH2
3.01
ND
ND
ND
ND
1.07


β-myrcene
166-168
MH3
11.62
6.93
1.08
ND
ND
5.50


Phellandrene
172
MH4
1.32
1.00
ND
ND
ND
0.72


α-terpinene
174
MH5
8.91
8.32
2.90
ND
ND
5.57


o-cymene
174
MH6
47.96
53.97
38.14
1.31
0.973
39.13


Limonene
175
MH7
2.29
2.71
1.25
ND
ND
1.58


1,8-cineole
177
MO1
1.51
1.77
2.74
ND
ND
1.53


γ-terpinene
181-183
MH8
15.59
24.43
40.57
1.40
0.94
22.34


Thymol
232
MO2
ND
ND
ND
5.08
3.77
1.71


Carvacrol
237-238
MO3
ND
ND
4.58
60.03
64.31
12.60


Trans-caryophyllene
268
SeH1
ND
ND
2.97
18.96
13.78
3.47


α-humulene
276
SeH2
ND
ND
0.34
6.16
8.36
1.56


Monoterpene hydrocarbons

(MH)
95.73
97.75
83.94
2.71
1.91
77.65


Monoterpene oxygenated

(MO)
1.51
1.77
7.32
65.11
68.08
15.84


Sesquiterpene hydrocarbons

(SeH)
ND
ND
3.31
25.12
22.14
5.03


Total identified components


97.24
99.52
94.57
92.94
92.13
98.52





Oregano essential oil (Ooil) was obtained through the steam entrainment method and the oil fractions through a fractional distillation system. The first fraction started to distill at a temperature of 82° C. and the last fraction distilling at 140° C., finally undistilled oil (Unoil) was obtained. At the end of the process, five fractions named Fraction 1 (F1), Fraction 2 (F2), Fraction 3 (F3), Fraction 4 (F4), and undistilled oil (Unoil) were obtained.






When Origanum extract was analyzed on HPLC, a variety of phenolic compounds were identified:


Phenolic compounds determined by the HPLC method in O. vulgare ssp. vulgare extract.


















[M—H],
Retention Time
UV
MS
Concentration


Compounds
m/z
(tR), min
Detection
Detection
(mg/g)







Gentisic acid
153
 3.69 ± 0.04
NO
YES
<0.02


Chlorogenic acid
353
 6.43 ± 0.05
YES
YES
 2.10 ± 0.14


p-Coumaric acid
163
 9.48 ± 0.08
NO
YES
<0.02


Hyperoside
463
18.60 ± 0.12
YES
YES
 1.05 ± 0.03


Isoquercitrin
463
20.29 ± 0.10
YES
YES
 0.71 ± 0.19


Rutin
609
20.76 ± 0.15
YES
YES
 0.64 ± 0.15


Rosmarinic acid
360
21.80 ± 0.10
YES
YES
12.83 ± 2.19


Quercitrin
447
23.64 ± 0.13
YES
YES
 0.50 ± 0.08


Quercetin
301
27.55 ± 0.15
NO
YES
<0.02


Luteolin
285
29.64 ± 0.19
YES
YES
 0.10 ± 0.04





Values are the mean ± SD (n = 3).






Total polyphenol content and antioxidant activity of O. vulgare ssp. vulgare extract.



















TPC (mg
Flavonoid
Caffeic Acids
CUPRAC
FRAP
SO Scavenging


Sample
GAE/g)
(mg RE/g)
(mg CAE/g)
(μM TE/g)
(μM TE/g)
(μM TE/g)








O. vulgare

94.69 ± 4.03
38.46 ± 3.54
29.92 ± 1.08
1284 ± 66
794.40 ± 25.80
44.00 ± 0.56





Each value is the mean ± SD of three independent measurements. TPC, total polyphenols content; SO, superoxide; GAE, gallic acid equivalents; RE, rutin equivalents; CAE, caffeic acid equivalents; TE, Trolox equivalents.






Also contemplated herein are plants of the genus Origanum.



Origanum is a genus of herbaceous perennials and subshrubs in the family Lamiaceae, native to Europe, North Africa, and much of temperate Asia, where they are found in open or mountainous habitats. A few species also naturalized in scattered locations in North America and other regions.


The plants have strongly aromatic leaves and abundant tubular flowers with long-lasting coloured bracts. The genus includes the important group of culinary herbs: marjoram (Origanum majorana) and oregano (Origanum vulgare).


Examples include, but are not limited to:

    • Origanum acutidens (Hand.-Mazz.) Ietsw.—Turkey, Iraq
    • Origanum x adanense Baser & H. Duman—Turkey (O. bargyli x O. laevigatum)
    • Origanum x adonidis Mouterde—Lebanon (O. libanoticum x O. syriacum subsp. bevanii)
    • Origanum akhdarense Ietsw. & Boulos—Cyrenaica region of eastern Libya
    • Origanum amanum Post—Hatay region of Turkey
    • Origanum x barbarae Bornm.—Lebanon (O. ehrenbergii x O. syriacum subsp. bevanii)
    • Origanum bargyli Mouterde—Turkey, Syria
    • Origanum bilgeri P. H. Davis—Antalya region of Turkey
    • Origanum boissieri Ietsw.—Turkey
    • Origanum calcaratum Juss.—Greece
    • Origanum compactum Benth.—Spain, Morocco
    • Origanum cordifolium (Montbret & Aucher ex Benth.) Vogel—Cyprus
    • Origanum cyrenaicum Bég. & Vacc.—Cyrenaica region of eastern Libya
    • Origanum dayi Post—Israel
    • Origanum dictamnus L.—hop marjoram, Cretan dittany, dittany of Crete—endemic to Crete
    • Origanum x dolichosiphon P. H. Davis—Seyhan region of Turkey (O. amanum x O. laevigatum)
    • Origanum ehrenbergii Boiss.—Lebanon
    • Origanum elongatum (Bonnet) Emb. & Maire—Morocco
    • Origanum floribundum Munby—Algeria
    • Origanum x haradjanii Rech. f—Turkey (O. laevigatum x O. syriacum subsp. bevanii)
    • Origanum haussknechtii Boiss.—Turkey
    • Origanum husnucan-baseri H. Duman, Aytac & A. Duran—Turkey
    • Origanum hypericifolium O. Schwarz & P. H. Davis—Turkey
    • Origanum x intercedens Rech. f.—Greece, Turkey (O. onites x O. vulgare subsp. hirtum)
    • Origanum x intermedium P. H. Davis—Denizli region of Turkey (O. onites x O. sipyleum)
    • Origanum isthmicum Danin—Sinai
    • Origanum jordanicum Danin & Kunne—Jordan
    • Origanum laevigatum Boiss.—Turkey, Syria, Cyprus
    • Origanum leptocladum Boiss.—Turkey
    • Origanum libanoticum Boiss.—Lebanon
    • Origanum majorana L.—(sweet) marjoram—Turkey, Cyprus; naturalized in scattered locations in Europe, North Africa, North+South America
    • Origanum x lirium Heldr. ex Halácsy—Greece (O. scabrum x O. vulgare subsp. hirtum)
    • Origanum x majoricum Cambess.—hardy sweet marjoram—Spain including Balearic Islands (O. majorana x O. vulgare subsp. virens)
    • Origanum microphyllum (Benth.) Vogel—Crete
    • Origanum x minoanum P. H. Davis—Crete (O. microphyllum x O. vulgare subsp. hirtum)
    • Origanum minutiflorum O. Schwarz & P. H. Davis—Turkey
    • Origanum munzurense Kit Tan & Sorger—Turkey
    • Origanum x nebrodense Tineo ex Lojac—Sicily (O. majorana x O. vulgare subsp. viridulum)
    • Origanum onites L.—Greece, Turkey, Sicily
    • Origanum x pabotii Mouterde—Syria (O. bargyli x O. syriacum subsp. bevanii)
    • Origanum pampaninii (Brullo & Furnari) Ietsw—Cyrenaica region of eastern Libya
    • Origanum petraeum Danin—Jordan
    • Origanum punonense Danin—Jordan
    • Origanum ramonense Danin—Israel
    • Origanum rotundifolium Boiss.—Turkey, Caucasus
    • Origanum saccatum P. H. Davis—Turkey
    • Origanum scabrum Boiss. & Heldr. in P. E. Boissier—Greece
    • Origanum sipyleum L.—Turkey, Greek Islands
    • Origanum solymicum P. H. Davis—Antalya region of Turkey
    • Origanum symes Carlström—Islands of the Aegean Sea
    • Origanum syriacum L.—Turkey, Cyprus, Syria, Lebanon, Jordan, Palestine, Israel, Sinai, Saudi Arabia
    • Origanum vetteri Briq. & Barbey—Crete
    • Origanum vogelii Greuter & Burdet—Turkey
    • Origanum vulgare L.—oregano—Europe, North Africa, temperate Asia (Iran, Siberia, Central Asia, China, etc.); naturalized in parts of North America, New Zealand, Venezuela.


According to a specific embodiment, the active ingredient or combination thereof includes an organic compound component of Origanum extract.


According to a specific embodiment, the active ingredient or combination thereof is selected from the group consisting of α-thujene α-pinene, β-myrcene, Phellandrene, α-terpinene, o-cymene, Limonene, 1,8-cineole, γ-terpinene, Thymol, Carvacrol, Trans-caryophyllene and α-humulene.


According to a specific embodiment, the active ingredient or combination thereof includes a monoterpene hydrocarbon, an oxygenated monoterpene and a sesquiterpene hydrocarbon.


According to a specific embodiment, the active ingredient or combination thereof includes a phenolic compound, e.g., gentisic acid, chlorogenic acid, p-coumaric acid, hyperoside, isoquercitrin, rutin, rosmarinic acid, quercirtin, quercetin and luteolin.


According to a specific embodiment, the active ingredient or combination thereof includes a mineral, e.g., potassium, calcium, magnesium, phosphorus, aluminum, iron, sodium, boron, zinc, cadmium, selenium.


Sesame


Sesame seeds contain thelignans, sesamolin, sesamin, pinoresinol andlariciresinol. Insoluble 11S globulin and soluble 2S albumin, conventionally termed α-globulin and β-globulin, are the two major storage proteins and constitute 80-90% of total seed proteins in sesame. Comparison of amino acid composition indicated that they are substantially less hydrophobic than the known oleosins, and thus should not be aggregated multimers of oleosins. The results of immuno-recognition to sesame proteins reveals that these three polypeptides are unique proteins gathered in oil bodies, accompanying oleosins and triacylglycerols, during the active assembly of the organelles in maturing seeds. The phospholipid, oleic and linoleic acids, chlorophyll and sesamolin, sesamol and γ-tocopherol are found. 10 compounds [2-furfurylthiol, 2-phenylethylthiol, 2-methoxyphenol, 4-hydroxy2, 5-dimethyl-3[2H]-furanone, 2-pentylpyridine, 2-ethyl-3,5-dimethylpyrazine, acetylpyrazine, [E,E]-2,4-decadienal, 2-acetyl-1-pyrroline and 4-vinyl-2-methoxy-phenol] are quantified. On the basis of high OAVs in oil, especially 2-acetyl-1-pyrroline [roasty], 2-furfurylthiol [coffee-like], 2-phenylethylthiol [rubbery] and 4-hydroxy-2,5-dimethyl3[2H]-furanone [caramel-like] are elucidated as important contributors to the overall roasty, sulphury odour of the crushed sesame material. The structures of novel sesaminol glucosides isolated from sesame seed are determined to be sesaminol 2′-O-β-d-glucopyranoside, sesaminol 2′-O-β-d-glucopyranosyl [1→2]-O-β-dglucopyranoside and sesaminol 2′-O-β-d-glucopyranosyl [1»2]-O-[β-d-glucopyransyl [1»6]]-[β-dglucopyranoside. Also minor sesame lignans such as -(7S,8′R,8R)-acuminatolide piperitol and pinoresinol (as mentioned).


Also contemplated herein are plants of the genus Sesamum.


Examples include, but are not limited to:

    • Sesamum abbreviatum Merxm.
    • Sesamum alatum Thonn.
    • Sesamum angolense Welw.
    • Sesamum biapiculatum De Wild.
    • Sesamum calycinum Welw.
    • Sesamum capense Burm. f.
    • Sesamum digitaloides Welw. ex Schinz
    • Sesamum gracile Endl.
    • Sesamum hopkinsii Suess.
    • Sesamum indicum L.
    • Sesamum lamiifolium Engl.
    • Sesamum latifolium J. B. Gillett
    • Sesamum lepidotum Schinz
    • Sesamum macranthum Oliv.
    • Sesamum marlothii Engl.
    • Sesamum mombazense De Wild. & T. Durand
    • Sesamum parviflorum Seidenst.
    • Sesamum pedalioides Welw. ex Hiern
    • Sesamum radiatum Schumach. & Thonn.
    • Sesamum rigidum Peyr.
    • Sesamum rostratum Hochst.
    • Sesamum sabulosum A. Chev.
    • Sesamum schinzianum Asch.
    • Sesamum somalense Chiov.
    • Sesamum thonneri De Wild. & T. Durand
    • Sesamum triphyllum Welw. ex Asch.


Plants that contain Lignan according to some embodiments of the invention include a wide variety of plant foods, including seeds (flax, pumpkin, sunflower, poppy, sesame), whole grains (rye, oats, barley), bran (wheat, oat, rye), beans, fruit (particularly berries), and vegetables (Broccoli and curly kale are rich sources of lignans. Other vegetables such as white and red cabbage, Brussels sprouts, cauliflower, carrots, green and red sweet peppers are also good sources).


Additional plants that contain Sesamin include but are limited to Eleutherococcus senticosus.


Thus, any combination of the above plants is contemplated including 2, 3, 4, 5, 6, 7 of the plants. According to another embodiment, a combination of extracts or fractions including 2, 3, 4, 5, 6, 7 of the different plants.


Examples include, but are not limited to, Nigella sativa, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Thymbra spicata, Satujera thymbra, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Satujera thymbra, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum.



Nigella sativa, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Satujera thymbra, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Sesamum indicum and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, and Rhus coriaria.



Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra.



Nigella sativa, Thymus capitatus.



Nigella sativa, Thymus vulgaris.



Nigella sativa, Origanum syriacum.



Nigella sativa, Thymbra spicata.



Nigella sativa, Satujera thymbra.



Nigella sativa, Sesamum indicum.



Nigella sativa, Rhus coriaria.


Also contemplated are various combinations without Nigella sativa.


According to another embodiment, a combination of active ingredients e.g., thymoquinone, carvacrol, thymol; thymoquinone, carvacrol; thymoquinone, thymol; carvacrol, thymol.



Nigella sativa, Thymus capitatus, Thymus vulgaris.



Nigella sativa, Thymus vulgaris, Origanum syriacum.



Nigella sativa, Origanum syriacum, Thymbra spicata.



Nigella sativa, Thymbra spicata, Satujera thymbra.



Nigella sativa, Satujera thymbra, Sesamum indicum Rhus coriaria.


According to some embodiments the plants and active ingredients thereof are listed in the Table below.
















Origanum
Carvacrol



Syricaum
thymol









Thymus Capitatus
Carvacrol




p-cymene




y-terpinene




b-caryophyllene



Thymus Vulgaris
Thymol



Thymbra Spicata
Carvacrol




y-terpinene




p-cymene



Satureja Thymbra
y-terpinene




p-cymene




carvacrol




thymol



Sumac ’
Tannin



Seasame
Lignans




Seasamolin




Seasamin




Pinoresinol




Lariciresinol



Nigella sativa
Thymoquinone










Other embodiments, which comprise any of the Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum and Rhus coriaria plants or genera thereof in combinations of 2, 3, 4, 5, 6, 7 and 8 plants are contemplated herein.


Other embodiments of the present invention include in the composition Bromelain or products thereof.


Other embodiments of the present invention include in the composition Pineapple extracts comprising Bromelain.


According to some embodiments of the invention, the component comprises tryptophan or an analog thereof.


According to an aspect of the invention there is provided a food supplement, composition or extracts further including “Beduin Tea” comprising


Rose Leaves Micromeria fruticose, Salvia, cymbopgon (Citral) Aloysia, Verbena officinalis, Origanum majorana, menthe

According to an aspect of the invention there is provided a food supplement, composition or extracts further including “Beduin Tea” comprising


Thyme, sage, cardamom, cinnamon, black tea, habuk, Marmaya.


The plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof can be used in the treatment of NMRD.


As used herein, “non-malignant respiratory disease” (NMRD) refers to a group of acute and chronic conditions affecting the respiratory tract and interfering with gas exchange, exclusive of cancerous and malignant proliferative pulmonary disease.


Examples of NMRD which are contemplated herein include, but are not limited to, pulmonary hypertension, interstitial lung disease, acute lung injury (ALI), acute respiratory distress syndrome (ARDS), sarcoidosis of the lung, bronchiectasis, asbestosis, berylliosis, silicosis, anthracosis, Histiocytosis X, pneumotitis, smoker's lung, bronchiolitis obliterans, lung scarring due to tuberculosis and pulmonary fibrosis, chronic obstructive pulmonary disease, pneumoconiosis, traumatic pulmonary injury, pulmonary infections and pulmonary manifestations of connective tissue diseases, including systemic lupus erythematosus, rheumatoid arthritis, scleroderma, polymyositis and dermatomyositis.


In some embodiments, the NMRD is the result of a pulmonary infection. Exemplary pulmonary infections which can cause NMRD include bronchitis, pneumonia, bronchiolitis, influenza, tuberculosis and malaria. Pulmonary infections which can cause NMRD include, but are not limited to viral, bacterial, fungal or parasitic infection. Pathogenic agents of pulmonary infections include, but are not limited to influenza viruses A, B and C, Streptococcus pneumonia, Haemophilus influenza, Mycoplasma pneumoniae, Respiratory Syncytial Virus, Pneumocystis jirovecii, Aspergillus, Histoplasma capsulatum, Plasmodium falciparum and parasitic helminth species.


In particular embodiments, types of NMRD contemplated herein include Chronic Obstructive Pulmonary Disease (COPD), Idiopathic Pulmonary Fibrosis (IPF) and Asthma.


In some embodiments, NMRD is the result of a primarily pulmonary disease, as in diseases caused by chronic inhalation of silica, anthracite, toxic fumes, asbestos, etc. In other embodiments, NMRD can be a secondary manifestation of pulmonary injury or dysfunction caused by scarring, fibrosis and other effects of non-pulmonary diseases or conditions, such as connective tissue disease (e.g. RA, scleroderma), infectious disease (e.g. polio) or autoimmune disease (SLA).


Individuals suffering from NMRD typically experience coughing, shortness of breath, wheezing, difficulty in breathing, pulmonary or nasal congestion, copious mucus, inflammation of the airways, chest tightness and exercise intolerance. Moderate to severe cases of NMRD, and particularly in older individuals, those with underlying medical problems and individuals frequently exposed to common triggers of respiratory disease such as particulate pollution (e.g. coal dust, asbestos, silica, etc), dust mites and allergens are more likely to develop into serious, chronic illness, in many cases requiring intensive care with ventilation and life-threatening disease.


According to specific embodiments, the NMRD is COPD.


The plant-derived component or components of the present invention can be co-administered with other medications to increase therapeutic bioavailability, boost therapeutic efficacy, and minimize side effects. The plant-derived component or components of the present invention may be administered in a linear or cyclical form, or in any conformation deemed physiologically appropriate as a means of conveying treatment.


In addition to targeted vasodilation, targeted anti-coagulation can be accomplished. For example, in a disease like acute lung injury, which is often marked by pulmonary intra-alveolar coagulation, targeted anti-coagulation can be delivered to the affected pulmonary area by co-administering an effective dose of the plant-derived component or components of the present invention with an anti-coagulant such as tissue factor pathway inhibitor (TFPI) or site-inactivated factor VIIa in a minimal dose to achieve targeted pulmonary anticoagulation with minimal changes in clotting ability over the areas of the body not undergoing thrombosis. Selective pulmonary anti-coagulation can also be utilized to treat other pulmonary diseases marked by pulmonary thrombosis such as pulmonary hypertension, lung transplant rejection and others.


In a disease like chronic obstructive pulmonary disease, which is often marked by shortness of breath, the plant-derived component or components of the present invention can be co-administered to boost the effective concentration and potency of drugs to relax airway smooth muscles such as long lasting beta-2 agonists such as salmeterol or formoterol.


Many pulmonary diseases are often marked by a decrease in glutathione (GSH), a powerful antioxidant. The plant-derived component or components of the present invention can be co-administered with N-Acetylcysteine (NAC), a glutathione precursor, in diseases like pulmonary fibrosis, PAH, ALI, and other pulmonary disorders to boost GSH production and scavenge reactive oxidants often found in pulmonary diseases. GSH may also serve to dampen the inflammatory immune response by binding to triggering receptor expressed on myeloid cells 1 (TREM1) and diminishing monocyte/macrophage- and neutrophil-mediated inflammatory responses. Co-administration of CAR with NAC can serve to lessen the severe inflammatory immune response that often characterizes severe pulmonary and fibrotic diseases like ALI, pulmonary hypertension, autoimmune diseases and many other conditions.


Treatments for pulmonary diseases like pulmonary fibrosis, PAH and ALI can also be improved by co-administering the plant-derived component or components of the present invention with TGF-beta inhibitors like decorin.


In pulmonary hypertension, pulmonary fibrosis and other pulmonary diseases, the benefits of endothelin (ET-1) receptor antagonists, prostacyclin derivatives, phosphodiesterase type 5 inhibitors can be increased for patients through the co-administration with the plant-derived component or components of the present invention.


Other pulmonary and fibrotic disease treatments such as Ketoconazole which inhibits thromboxane and leukotriene synthesis can be improved in its efficacy while minimizing side effects through co-administration with the plant-derived component or components of the present invention.


The term “treating” refers to inhibiting, preventing or arresting the development of a pathology (disease, disorder or condition) and/or causing the reduction, remission, or regression of a pathology. Those of skill in the art will understand that various methodologies and assays can be used to assess the development of a pathology, and similarly, various methodologies and assays may be used to assess the reduction, remission or regression of a pathology.


As used herein, the term “preventing” refers to keeping a disease, disorder or condition from occurring in a subject who may be at risk for the disease, but has not yet been diagnosed as having the disease.


As used herein, the term “subject” includes mammals, preferably human beings, male or female, at any age or gender, who suffer from the pathology. Preferably, this term encompasses individuals who are at risk to develop the pathology (e.g., above 65 of age, exposed to airborne pollutants, particulates, toxic fumes, allergens).


The composition of matter comprising the component(s) (a plant species or genus thereof-derived component selected from the group consisting of a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein said component is capable of ameliorating symptoms of NMRD) of the present invention can be administered to the subject per se, or in a pharmaceutical composition where it is mixed with suitable carriers or excipients.


As used herein a “pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.


Herein the term “active ingredient” refers to the composition of matter comprising the components accountable for the biological effect.


Hereinafter, the phrases “physiologically acceptable carrier” and “pharmaceutically acceptable carrier” which may be interchangeably used refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound. An adjuvant is included under these phrases.


Herein the term “excipient” refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols.


Techniques for formulation and administration of drugs may be found in “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, PA, latest edition, which is incorporated herein by reference.


Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal or parenteral delivery, including intramuscular, subcutaneous and intramedullary injections as well as intrathecal, direct intraventricular, intracardiac, e.g., into the right or left ventricular cavity, into the common coronary artery, intravenous, intraperitoneal, intranasal, or intrapulmonary or intraocular injections.


In various exemplary embodiments of the invention, the composition is provided as a pharmaceutical or dietary supplement dosage form suitable for oral administration. Dosage forms suitable for oral administration include tablets, soft capsules, hard capsules, pills, granules, powders, emulsions, suspensions, sprays, syrups and pellets. In various other embodiments of the invention, the composition is provided as a pharmaceutical dosage form suitable for parenteral administration such as liquid formulations for administration as drops or by injection, or as solid or semisolid dosage forms for suppositories.


Conventional approaches for drug delivery to the central nervous system (CNS) include: neurosurgical strategies (e.g., intracerebral injection or intracerebroventricular infusion); molecular manipulation of the agent (e.g., production of a chimeric fusion protein that comprises a transport polypeptide that has an affinity for an endothelial cell surface molecule in combination with an agent that is itself incapable of crossing the BBB) in an attempt to exploit one of the endogenous transport pathways of the BBB; pharmacological strategies designed to increase the lipid solubility of an agent (e.g., conjugation of water-soluble agents to lipid or cholesterol carriers); and the transitory disruption of the integrity of the BBB by hyperosmotic disruption (resulting from the infusion of a mannitol solution into the carotid artery or the use of a biologically active agent such as an angiotensin polypeptide). However, each of these strategies has limitations, such as the inherent risks associated with an invasive surgical procedure, a size limitation imposed by a limitation inherent in the endogenous transport systems, potentially undesirable biological side effects associated with the systemic administration of a chimeric molecule comprised of a carrier motif that could be active outside of the CNS, and the possible risk of brain damage within regions of the brain where the BBB is disrupted, which renders it a suboptimal delivery method.


Alternately, one may administer the pharmaceutical composition in a local rather than systemic manner, for example, via injection of the pharmaceutical composition directly into a tissue region of a patient.


Pharmaceutical compositions of some embodiments of the invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes.


Pharmaceutical compositions for use in accordance with some embodiments of the invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations which, can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.


For injection, the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.


For oral administration, the pharmaceutical composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.


Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.


Pharmaceutical compositions which can be used orally, include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.


For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.


In specific embodiments, the components and/or compositions of the invention are provided in form suitable for administration by inhalation or nasal administration.


For administration by nasal inhalation, the active ingredients for use according to some embodiments of the invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane or carbon dioxide. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, e.g., gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.


The pharmaceutical composition described herein may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative. The compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.


Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acids esters such as ethyl oleate, triglycerides or liposomes. Aqueous injection suspensions may contain substances, which increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the active ingredients to allow for the preparation of highly concentrated solutions.


Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water based solution, before use.


The pharmaceutical composition of some embodiments of the invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, e.g., conventional suppository bases such as cocoa butter or other glycerides.


Pharmaceutical compositions suitable for use in context of some embodiments of the invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a therapeutically effective amount means an amount of active ingredients (composition of matter comprising the components accountable for the biological effect) effective to prevent, alleviate or ameliorate symptoms of a disorder (e.g., NMRD) or prolong the survival of the subject being treated.


Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.


For example, any in vivo or in vitro method of evaluating the severity of NMRD symptoms may be employed.


For any preparation used in the methods of the invention, the therapeutically effective amount or dose can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.


Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration and dosage can be chosen by the individual physician in view of the patient's condition. (See e.g., Fingl, et al., 1975, in “The Pharmacological Basis of Therapeutics”, Ch. 1 p. 1).


Dosage amount and interval may be adjusted individually to provide the active ingredient at a sufficient amount to induce or suppress the biological effect (minimal effective concentration, MEC). The MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.


Depending on the severity and responsiveness of the condition to be treated, dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks or until cure is effected or diminution of the disease state is achieved.


The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.


Compositions of some embodiments of the invention may, if desired, be presented in a pack or dispenser device, such as an FDA approved kit, which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser may also be accommodated by a notice associated with the container in a form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions or human or veterinary administration. Such notice, for example, may be of labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert. Compositions comprising a preparation of the invention formulated in a compatible pharmaceutical carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as is further detailed above.


In another embodiment, the invention provides a nutritional or dietary compositions in the form of foods or beverages, which comprise the component(s) described herein. These foods or beverages comprise various exemplary embodiments of the inventive compositions. These foods or beverages can be prepared or provided as cereals, baby foods, healthy foods, or food for specified health uses such as solid food like chocolate or nutritional bars, semisolid food like cream or jam, or gel; and also as beverages. Specific and non-limiting examples of such food or beverage items include refreshing beverages, lactic acid bacteria beverages, drops, candies, chewing gum, chocolate, gummy candy, yoghurts, ice creams, puddings, soft adzuki bean jellies, jellies, cookies and the like.


The present teachings further envisage treating with other anti-viral drugs or anti-inflammatory drugs or anti-coagulants as separate treatments or in a co-formulation.


Without being limited to NMRD but for the sake of example, according to a specific embodiment, the antiviral drug is selected from the group consisting of remdesivir, an interferon, ribavirin, adefovir, tenofovir, acyclovir, brivudin, cidofovir, fomivirsen, foscarnet, ganciclovir, penciclovir, amantadine, rimantadine and zanamivir.


Also contemplated are plasma treatments from infected persons who survived and/or anti-HIV drugs such as lopinavir and ritonavir, as well as chloroquine.


Specific examples of drugs that are routinely used for the treatment of NMRD include, but are not limited to, anti-inflammatories, bronchodilators such as beta-2-agonists and anticholinergics, corticosteroids, phosphodieaterase-4-inhibitors, theophylline, antibiotics and antivirals could be the drug treatment options for NMRD.


As used herein the term “about” refers to ±10%


The terms “comprises”, “comprising”, “includes”, “including”, “having” and their conjugates mean “including but not limited to”.


The term “consisting of” means “including and limited to”.


The term “consisting essentially of” means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.


As used herein, the singular form “a”, “an” and “the” include plural references unless the context clearly dictates otherwise. For example, the term “a compound” or “at least one compound” may include a plurality of compounds, including mixtures thereof.


Throughout this application, various embodiments of this invention may be presented in a range format. It should be understood that the description in range format is merely for convenience and brevity and should not be construed as an inflexible limitation on the scope of the invention. Accordingly, the description of a range should be considered to have specifically disclosed all the possible subranges as well as individual numerical values within that range. For example, description of a range such as from 1 to 6 should be considered to have specifically disclosed subranges such as from 1 to 3, from 1 to 4, from 1 to 5, from 2 to 4, from 2 to 6, from 3 to 6 etc., as well as individual numbers within that range, for example, 1, 2, 3, 4, 5, and 6. This applies regardless of the breadth of the range.


Whenever a numerical range is indicated herein, it is meant to include any cited numeral (fractional or integral) within the indicated range. The phrases “ranging/ranges between” a first indicate number and a second indicate number and “ranging/ranges from” a first indicate number “to” a second indicate number are used herein interchangeably and are meant to include the first and second indicated numbers and all the fractional and integral numerals therebetween.


As used herein the term “method” refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.


As used herein, the term “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.

    • As used herein the term “viral entry mechanism” refers to viral proteins that mediate entry into cells. The viral entry mechanism proteins include attachment proteins and other proteins that are required for entry of non-enveloped and enveloped viruses into cells. Different viruses use different entry proteins, however, both non-enveloped and enveloped viruses share the same two main steps and routes of virus entry; (1) attachment to cell-surface receptors (2) conformational changes of the viral entry proteins or the host-cell receptors, the viral entry can occur either by penetration of the cell membrane (for non-enveloped viruses) or fusion (for enveloped viruses) to the cell membrane (see “Virus entry: molecular mechanisms and biomedical applications”, Dimitrov, 2004)


It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.


Various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below find experimental support in the following examples.


EXAMPLES

Reference is now made to the following examples, which together with the above descriptions illustrate some embodiments of the invention in a non-limiting fashion.


Generally, the nomenclature used herein and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, “Molecular Cloning: A laboratory Manual” Sambrook et al., (1989); “Current Protocols in Molecular Biology” Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., “Current Protocols in Molecular Biology”, John Wiley and Sons, Baltimore, Maryland (1989); Perbal, “A Practical Guide to Molecular Cloning”, John Wiley & Sons, New York (1988); Watson et al., “Recombinant DNA”, Scientific American Books, New York; Birren et al. (eds) “Genome Analysis: A Laboratory Manual Series”, Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; “Cell Biology: A Laboratory Handbook”, Volumes I-III Cellis, J. E., ed. (1994); “Culture of Animal Cells—A Manual of Basic Technique” by Freshney, Wiley-Liss, N. Y. (1994), Third Edition; “Current Protocols in Immunology” Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), “Basic and Clinical Immunology” (8th Edition), Appleton & Lange, Norwalk, CT (1994); Mishell and Shiigi (eds), “Selected Methods in Cellular Immunology”, W. H. Freeman and Co., New York (1980); available immunoassays are extensively described in the patent and scientific literature, see, for example, U.S. Pat. Nos. 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521; “Oligonucleotide Synthesis” Gait, M. J., ed. (1984); “Nucleic Acid Hybridization” Hames, B. D., and Higgins S. J., eds. (1985); “Transcription and Translation” Hames, B. D., and Higgins S. J., eds. (1984); “Animal Cell Culture” Freshney, R. I., ed. (1986); “Immobilized Cells and Enzymes” IRL Press, (1986); “A Practical Guide to Molecular Cloning” Perbal, B., (1984) and “Methods in Enzymology” Vol. 1-317, Academic Press; “PCR Protocols: A Guide To Methods And Applications”, Academic Press, San Diego, CA (1990); Marshak et al., “Strategies for Protein Purification and Characterization—A Laboratory Course Manual” CSHL Press (1996); all of which are incorporated by reference as if fully set forth herein. Other general references are provided throughout this document. The procedures therein are believed to be well known in the art and are provided for the convenience of the reader. All the information contained therein is incorporated herein by reference.


Example 1
Assays for Amelioration of NMRD

Many cell-based, in-vitro systems for evaluation of conditions comprising NMRD are available, in addition to traditional in-vivo animal models. To model NMRD, primary as well as cell lines of airway-associated cells such as pleural mesothelial cells (PMC), small airway epithelial cells (SAEC), bronchial epithelial cells (BEC), nasal epithelial cells, and bronchial fibroblasts are cultured and exposed to chemical/pathogenic insults. Cell responses, and in particular inflammatory processes, senescence and cell activation are determined in the presence or absence of added compositions and/or components of the invention, in order to evaluate the ability of the compositions and/or components of the invention to reduce or otherwise modify the effect of the insults on the cells. Cells can be propagated in 2-D or 3-D cultures.


For example, cells for in-vitro modeling of asthma include human pulmonary smooth muscle cells and airway epithelial cells. Cells for modeling of pulmonary fibrosis include pleural mesothelial cells. COPD can be modeled in vitro with bronchial fibroblasts, and asbestos/silica-induced damage can be evaluated using pleural mesothelial cells.


In an exemplary cell-based assay of fibrotic respiratory disease (e.g. COPD, asthma, IPF), fibroblast-to-myofibroblast transformation and epithelial-to-mesenchymal transition are assayed in normal lung fibroblasts and small airway epithelial cells.


Primary human lung fibroblasts or SAEC cells are induced into transformation with the pro-fibrotic growth factor TGFbeta-1, and markers of the different phenotypes (aSMA, collagen-I, fibronectin, E-cadherin) are evaluated. Anti-fibrotic effects of the compositions and/or components of the invention on the extent and character of transformation are evaluated.


Animal Models of Non-Malignant Respiratory Disease


Animal models for NMRD include induced animal models and naturally occurring animal models of the respiratory diseases and conditions.


Induced animal models suitable for assessing efficacy of the compositions and components of the invention include, but are not limited to:

    • COPD: Rodents, exposure to cigarette smoke, assessment of vascular wall and airway remodeling;
    • EMPHYSEMA: Rodents, exposure to elastin-degrading elastase, transgenic overexpression of collagenase;
    • LOWER RESPIRATORY INFECTION: Rodents, cohabitation, aerosolization, nebulizer, direct airway installation;
    • PULMONARY FIBROSIS: Rodents, environmental exposure (silicosis, asbestosis, allergens, Bleomycin induction, hypochlorous acid HOCl, hapteninduced lung fibrosis;
    • ASTHMA: Rodents, repeated challenge with allergens (leading to goblet-cell hyperplasia, airway inflammation and hyperresponsiveness (remodeling), exposure to dust mites.


A few naturally occurring animal models for NMRD are known and may be suitable for assessment of the effects of the compositions and/or components of the invention: IPF susceptibility is documented in certain breeds cats, dogs, horses and donkeys, and the asthma-like Hyperreactive Airway Disease is well known in horses and cats.


Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims.


Example 2
Assays for Amelioration of SARS-CoV-2 by Plant Components

COVID-19 is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The cell membrane ACE-2 receptor is an attachment and entry site for SARS-CoV-2. The ACE-2 receptor is a type I transmembrane metallocarboxypeptidase with homology to ACE, an enzyme long-known to be a key player in the Renin-Angiotensin system (RAS) and a target for the treatment of hypertension. There is evidence that SARS-CoV-2 utilizes ACE-2 as a cellular entry receptor. Zhou et al. showed that SARS-CoV-2 could use ACE-2 from humans, Chinese horseshoe bats, civet cats, and pigs to gain entry into ACE-2-expressing HeLa cells (See Zhou, P., Yang, X L., Wang, X G. et al. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579, 270-273 (2020)). The spike (S) protein of SARS-CoV-2, which plays a key role in the receptor recognition and cell membrane fusion process, is composed of two subunits, S1 (120 kDa) and S2 (80 kDa). The S1 subunit contains a receptor-binding domain that recognizes and binds to the host receptor angiotensin-converting enzyme 2 (ACE-2). The S2 subunit mediates viral cell membrane fusion by forming a six-helical bundle via the two-heptad repeat domain (see Huang, Y., Yang, C., Xu, Xf. et al. Structural and functional properties of SARS-CoV-2 spike protein: potential antivirus drug development for COVID-19. Acta Pharmacol Sin 41, 1141-1149 (2020)). Interfering, attenuating, impairing the function of the S1 and S2 subunits will eventually lead to an attenuated, impaired and less infective virus.


Viral Protein Digestion Assays

The materials used in all the following viral protein digestion assays are disclosed in table 1.











TABLE 1






Company
cat #







Recombinant SARS-Cov-2 S1 Subunit
KavBiotech
230-30161


Recombinant SARS-Cov-2 S2 Subunit
BaySiotack
230-30163


Recombinant SARS-Cov-2 Nucleocapsid
RayBiotech
230-30154


Proteinase K
Sigma
P4850


Instanst Blue
Expedeon
ISB1L


DMSO
SIGMA
D2650


Criterion ™ TGX Stain-Free Precast Midi
BIDRAD
5671084


Gels 4-15%, 18 wells, 30 μl




10X Tris/Glycine/SDS
BIORAD
161-0772


4X Laemmli Sample Buffer
BIORAD
1610747


B-Mercaptoethanol 14.2M
SIGMA
M3148


Precision protein standard
BIORAD
161-0373










Each of the tested plant based treatment was numbered as disclosed in table 2 and the tested combinations complexes were classified as disclosed in table 3










TABLE 2





Treatment



No.
Name







1
Oregano Oil


2
Thyme Oil


3
Nigela Sativa


4
Sumac Oil


5
Sesame Oil


6
Olibanum Oil



















TABLE 3









Complex A
Oils 1 + 2 + 3



Complex B
Oils 1 + 2 + 3 + 4



Complex C
Oils 1 + 2 + 3 + 4 + 5



Complex D
Oils 1 + 2 + 3 + 4 + 5 + 6











Oils mixtures were prepared by mixing equal amounts of each oil. The mix was then diluted 1:2 with DMSO, to acquire a solution of 50% DMSO, 50% Oil mix and the final reaction concentration was 5% oil mix, 5% DMSO.


For each assay reaction, 1 μg protein per reaction was incubated with 3 μl of the oil mixture at final reaction volume of 30 μl. The reaction was incubated for 6 hours at 37° C.


Following incubation, the reaction was stopped by adding 10 μl/reaction of sample buffer 4× and incubation 10 minutes at 72° C. Samples were then run in 4-15% TGX Criterion Gel (BIORAD) for 50 minutes at 200 Volt. Following run, the gel was incubated for 1 hour with Instant Blue reagent (Expedeon) and further washed with water until distinct bands were observed.


Densitometry was preformed, pictures were analyzed with ImageJ software.


The protein digestion assay was conducted as disclosed in table 4. SARS-CoV-2 S1 subunit, SARS-CoV-2 S2 subunit, SARS-CoV-2 Nucleocapsid protein and a negative control with no protein, were incubated with different plant oils or combinations for 6 h at 37° c. and subsequently run on SDS-page, stained with “instant blue” for the presence of proteins in the gel. The untreated control appeared at the expected molecular weight and the effect of different treatments were compared to this control (see FIGS. 2-4). Significant disappearance pf the protein was observed following treatment with the protease proteinase K.














TABLE 4





No #
Protein
Treatment
Incubation
μg/lane
Expected MW (KDa)




















1
SARS-CoV-2.51 Subunit
Non treated—control
W/O Incubation
1 μg
120 kDa


2

Positive control—Proteinase K
6 hours—37° C.




3

Complex B





4

Complex C





5

Complex D





6

Negative control—DMSO





7

Non treated—control





8
SARS-CoV-2.52 Subunit
Non treated—control
W/O Incubation
1 μg
 80 kDa


9

Positive control—Proteinase K
6 hours—37° C.




10

Complex B





11

Complex C





12

Complex D





13

Negative control—DMSO





14

Non treated—control





15
SARS-CoV-2 Nucleocapsid
Non treated—control
W/O Incubation
1 μg
 55 kDa


16

Positive control—Proteinase K
6 hours—37° C.




17

Complex B





18

Complex C





19

Complex D





20

Negative control—DMSO





21

Non treated—control





22
No protein
Complex B
6 hours—37° C.




23

Complex C





24

Complex D










Densitometry of the SARS-CoV-2 S1 subunit, SARS-CoV-2 S2 subunit, SARS-CoV-2 and the Nucleocapsid protein assays disclose that although the Nucleocapsid protein underwent little to no digestion with either of the tested treatments as compared to the protein K treatment the two SARS-CoV-2 subunits S1 and S2 underwent a substantial digestion (see FIGS. 5-7 respectively).


To conclude, the viral protein digestion assay demonstrates that there is a significant digestion of the S1 and S2 subunits without destroying the Nucleocapsid protein.


Recombinant SARS-Cov-2 S1 Subunit

Following incubation of the protein with a mix prepared from equal volumes from items 1+2+3+4 (complex B), a 26% reduction in the protein signal was observed.


Recombinant SARS-Cov-2 S2 Subunit Protein

Following incubation of the protein with a mix prepared from equal volumes from items 1+2+3+4 (complex B), a 19% reduction in the protein signal was observed.


Following incubation of the protein with a mix prepared from equal volumes from items 1+2+3+4+5 (complex C), a 27% reduction in the protein signal was observed


Following incubation of the protein with a mix prepared from equal volumes from items 1+2+3+4+5+6 (complex D), a 47% reduction in the protein signal was observed.


These significant digestion rates of both S1 and S2 subunits of the spike protein are likely to result in the subsequent attenuation of the Coronavirus cell attachment and internalization mechanism. It is clear that attenuating the virus cell attachment and internalization mechanism, disease levels and viral load can be reduced in an infected subject, or prevent an uninfected subject from getting infected.


It is the intent of the applicant(s) that all publications, patents and patent applications referred to in this specification are to be incorporated in their entirety by reference into the specification, as if each individual publication, patent or patent application was specifically and individually noted when referenced that it is to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. To the extent that section headings are used, they should not be construed as necessarily limiting. In addition, any priority document(s) of this application is/are hereby incorporated herein by reference in its/their entirety.

Claims
  • 1. A method of reducing the infectivity of a non-malignant respiratory disease (NMRD) virus by modifying the viral entry mechanism proteins in a subject, the method comprising administering to the subject an effective amount of a composition of claim 5.
  • 2. A method of reducing the infectivity of a virus by modifying the viral entry mechanism proteins in a subject, the method comprising administering to the subject an effective amount of a composition of claim 5.
  • 3. A method of treating non-malignant respiratory disease (NMRD) in a subject in need thereof, the method comprising administering to the subject an effective amount of a plant species or genus thereof-derived component selected from a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein said component is capable of ameliorating symptoms of NMRD and wherein said plant species is selected from Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.
  • 4. A vaccine against NMRD comprising an effective amount of a plant species or genus thereof-derived component selected from a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein said component is capable of ameliorating symptoms of NMRD and wherein said plant species is selected from Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.
  • 5. A pharmaceutical composition comprising an effective amount of a plant species or genus thereof-derived component selected from a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein said component is capable of ameliorating symptoms of NMRD and wherein said plant species is selected from Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum, Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng for use in preventing or treating NMRD.
  • 6. The composition of claim 5 comprising at least 2 of said plant species or genus thereof-derived components.
  • 7. A food supplement comprising a combination of at least 2 of a plant species or genus thereof-derived component selected from a plant part, extract thereof, fraction thereof, active ingredient thereof, synthetic analog thereof, mimetic thereof or combination thereof, wherein said component is capable of ameliorating symptoms of NMRD and wherein said plant species is selected from Nigella sativa, Thymus capitatus, Thymus vulgaris, Origanum syriacum, Thymbra spicata, Satujera thymbra, Sesamum indicum Rhus coriaria, Gynostemma pentaphyllum, Boswellia sacra and Panax ginseng.
  • 8. The method of claim 1, wherein said NMRD is selected from pulmonary hypertension, interstitial lung disease, acute lung injury (ALI), acute respiratory distress syndrome (ARDS), sarcoidosis of the lung, bronchiectasis, asbestosis, berylliosis, silicosis, anthracosis, Histiocytosis X, pneumotitis, smoker's lung, bronchiolitis obliterans, lung scarring due to tuberculosis and pulmonary fibrosis, chronic obstructive pulmonary disease, pneumoconiosis, traumatic pulmonary injury, pulmonary infections and pulmonary manifestations of connective tissue diseases, including systemic lupus erythematosus, rheumatoid arthritis, scleroderma, polymyositis and dermatomyositis.
  • 9. The method of claim 1, wherein said NMRD is Chronic Obstructive Pulmonary Disease (COPD), Idiopathic Pulmonary Fibrosis (IPF) or Asthma.
  • 10. The method of claim 1, wherein said symptoms are selected from coughing, shortness of breath, wheezing, difficulty in breathing, inflammation of the airways, chest tightness and exercise intolerance.
  • 11. The method of claim 1, wherein said component comprises at least 2 components.
  • 12. The method of claim 1, wherein said component comprises at least 3 components.
  • 13. The method of claim 1, wherein said component comprises at least 4 components.
  • 14. The method of claim 1, wherein said component comprises at least 5 components.
  • 15. The method of claim 1, wherein said component comprises 5-10 components.
  • 16. The method of claim 1, wherein said component comprises thymoquinone or an analog thereof.
  • 17. The method of claim 1, wherein said component comprises thymol or an analog thereof.
  • 18. The method of claim 1, wherein said component comprises carvacrol or an analog thereof.
  • 19. The method of claim 1, wherein said component comprises bromelain or an analog thereof.
  • 20. The method of claim 1, wherein said component comprises pineapple extract comprising bromelain or an analog thereof.
  • 21. The method of claim 1, wherein said component comprises tryptophan or an analog thereof, or extract of a plant containing tryptophan.
  • 22. The method of claim 1 wherein said plant species or genus thereof-derived component further comprises “Beduin Tea” comprising Rose Leaves Micromeria fruticose, Salvia, cymbopgon (Citral) Aloysia, Verbena officinalis, Origanum majorana, and menthe.
  • 23. The method of claim 1 wherein said plant species or genus thereof-derived component further comprises “Beduin Tea” comprising Thyme, sage, cardamom, cinnamon, black tea, habuk, Marmaya.
PCT Information
Filing Document Filing Date Country Kind
PCT/IL2021/050308 3/19/2021 WO
Provisional Applications (1)
Number Date Country
62992276 Mar 2020 US