WO2023161921A2 - Compositions destinées à être utilisées en tant que thérapie d'appoint dans le traitement du cancer du sein - Google Patents

Compositions destinées à être utilisées en tant que thérapie d'appoint dans le traitement du cancer du sein Download PDF

Info

Publication number
WO2023161921A2
WO2023161921A2 PCT/IL2023/050159 IL2023050159W WO2023161921A2 WO 2023161921 A2 WO2023161921 A2 WO 2023161921A2 IL 2023050159 W IL2023050159 W IL 2023050159W WO 2023161921 A2 WO2023161921 A2 WO 2023161921A2
Authority
WO
WIPO (PCT)
Prior art keywords
weight
amount
extract
composition
total
Prior art date
Application number
PCT/IL2023/050159
Other languages
English (en)
Other versions
WO2023161921A3 (fr
Inventor
Efrat ORON
Basem FARES
Dobroslav Melamed
Itzhak ANGEL
Tamar Peretz
Original Assignee
Cannabotech Ltd.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cannabotech Ltd. filed Critical Cannabotech Ltd.
Publication of WO2023161921A2 publication Critical patent/WO2023161921A2/fr
Publication of WO2023161921A3 publication Critical patent/WO2023161921A3/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/01Hydrocarbons
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/01Hydrocarbons
    • A61K31/015Hydrocarbons carbocyclic
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • A61K31/138Aryloxyalkylamines, e.g. propranolol, tamoxifen, phenoxybenzamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41961,2,4-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/658Medicinal preparations containing organic active ingredients o-phenolic cannabinoids, e.g. cannabidiol, cannabigerolic acid, cannabichromene or tetrahydrocannabinol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/07Basidiomycota, e.g. Cryptococcus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K36/00Medicinal preparations of undetermined constitution containing material from algae, lichens, fungi or plants, or derivatives thereof, e.g. traditional herbal medicines
    • A61K36/06Fungi, e.g. yeasts
    • A61K36/07Basidiomycota, e.g. Cryptococcus
    • A61K36/074Ganoderma

Definitions

  • the present invention relates to cannabinoid- and mushroom-based compositions for use as an adjunctive therapy to a conventional therapy for treating a breast cancer.
  • Cannabis-derived substances in cancer therapy. These substances, collectively referred to as cannabinoids, have varying degrees of biochemical activity, and specifically anticancer activity which makes them effective in a wide spectrum of tumor cells both in vivo and in vitro.
  • cannabinoids such as A 8 - tetrahydrocannabinol (A 8 -THC), A 9 -tetrahydrocannabinol (A 9 -THC), and cannabidiol (CBD) inhibited the DNA synthesis and growth of lung adenocarcinoma in cultured cells as well as in mouse tumor models. Similar effects were seen in both in vitro and in vivo models of other cancers including glioma, breast, pancreas, prostate, colorectal carcinoma, and lymphoma. There are various proposed mechanisms of action behind these findings, including cell cycle arrest and induction of apoptosis, as well as inhibition of neovascularization, migration, adhesion, invasion, and metastasis.
  • Cannabinoids have also been tested in combination with other chemotherapeutic drugs or radiotherapy to establish whether they can enhance the efficacy of conventional treatments.
  • the administration of CBD together with some anticancer drugs has been shown to increase the susceptibility of glioblastoma cells to the cytotoxic effects of the drugs (Nabissi et al. 2013).
  • CBD and bortezomib synergistically reduced the viability of transient receptor potential cation channel subfamily V member 2 (TRPV2)-transfected and un-transfected multiple myeloma cell lines.
  • TRPV2 transient receptor potential cation channel subfamily V member 2
  • Terpenes are naturally occurring compounds found especially in essential oils, resins, and balsam, which have a broad range of biological properties.
  • terpenes are known as skin penetration enhancers, and were found to have cancer chemo - preventive effects as well as anti-inflammatory activities.
  • Medicinal mushrooms have a proven history of use worldwide.
  • medicinal mushrooms are used as dietary food or food supplement products; dietary pet and veterinary food supplements; natural bio-control agents in plant; cosmeceuticals and nutricosmetics; as well as drugs (referred to as “mushroom pharmaceuticals”).
  • medicinal mushrooms have shown various medicinal functions including, e.g., antitumor, immunomodulating, antioxidant, radical scavenging, cardiovascular, cholesterol-lowering, antiviral, antibacterial, anti-parasitic, antifungal, detoxification, hepatoprotective, anti-diabetic, anti-obesity, neuroprotective, and neuro-regenerative activities.
  • substances derived from medicinal mushrooms have been shown as active painkillers or analgesics.
  • the best implementation of medicinal mushroom-based drugs and dietary supplements has been in preventing immune disorders and maintaining a good quality of life, especially in immunodeficient and immuno-depressed patients, patients under chemotherapy or radiotherapy, patients with different types of cancers, chronic blood-borne viral infections of Hepatitis B, C and D, different types of anemia, the human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS), Herpes simplex virus (HSV), Epstein Bar virus, Influenza viruses A and B, H5N1, COVID-19 (Murphy et al., 2020), West Nile virus, chronic fatigue syndrome, patients with chronic gastritis and gastric ulcers caused by Helicobacter pylori, and people suffering from dementia (especially Alzheimer’s disease) (Wasser, 2010, Chang and Wasser, 2018).
  • HIV/AIDS human immunodeficiency virus/acquired immunodeficiency syndrome
  • HSV Herpes simplex virus
  • Epstein Bar virus Epstein Bar virus
  • Influenza viruses A and B H
  • mushrooms were used as adjuvant treatment with conventional chemo- or radiotherapy in different kinds of cancer (Wasser, 2017, Vetvicka et al., 2019).
  • a composition comprising CBD, A 9 -THC, CBDV, and CBDA (herein referred to as “a cannabinoid composition ) and a composition comprising a mixture of extracts obtained from Agaricus bisporus, Ganoderma lucidum, Ganoderma lucidum spore, Grifola frondosa, Lentinus edodes, and Trametes versicolor (herein referred to as “a mushroom composition ), each had cytotoxic activity on various breast cancer cell lines, more specifically MCF7, BT474, SKBR3, and MDA-MB231.
  • each one of those compositions when applied to breast cancer cell lines together with a conventional chemotherapeutic agent, more specifically taxol or carboplatin, had a synergistic effect in reducing cell viability compared to application of either the composition or chemotherapeutic agent alone. Based on these data, it is expected that a combination of each one of these compositions, when administered as an adjunctive therapy to a conventional therapy, e.g., a chemotherapy, aimed at treating a breast cancer, will have even a greater synergistic effect increasing or maximizing the efficacy of said conventional therapy.
  • a conventional therapy e.g., a chemotherapy
  • the present invention thus relates to a method for treatment of a breast cancer in a subject in need thereof, said subject undergoing a conventional therapy for treating said breast cancer, and said method comprising administering to said subject, as an adjunctive therapy, a cannabinoid composition, a mushroom composition, or a combination of said cannabinoid composition and said mushroom composition, wherein: (i) said cannabinoid composition comprises cannabidiol (CBD) or an enantiomer, diastereomer, or a mixture thereof; A 9 -tetrahydrocannabinol (A 9 -THC) or an enantiomer, diastereomer, or a mixture thereof; cannabidivarin (CBDV) or an enantiomer, diastereomer, or a mixture thereof; cannabidiolic acid (CBDA) or an enantiomer, diastereomer, or a mixture thereof, and optionally one or more terpene
  • CBD cannab
  • said cannabinoid composition comprises one or more terpenes, e.g., at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene, constituting up to about 3%, e.g., from about 1% to about 2%, by weight of said composition.
  • terpenes e.g., at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene
  • the present invention relates to a cannabinoid composition, a mushroom composition, or a combination of said cannabinoid composition and said mushroom composition, for use as an adjunctive therapy to a conventional therapy in the treatment of a breast cancer
  • said cannabinoid composition comprises CBD or an enantiomer, diastereomer, or a mixture thereof; A 9 -THC or an enantiomer, diastereomer, or a mixture thereof; CBDV or an enantiomer, diastereomer, or a mixture thereof; CBDA or an enantiomer, diastereomer, or a mixture thereof; and optionally one or more terpenes
  • said mushroom composition comprises a mixture of Agaricus bisporus extract, Ganoderma lucidum extract, Ganoderma lucidum spore extract, Grifola frondosa extract, Lentinus edodes extract, and Trametes versicolor extract
  • said cannabinoid composition comprises one or more terpenes, e.g., at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene, constituting up to about 3%, e.g., from about 1% to about 2%, by weight of said composition.
  • terpenes e.g., at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene
  • the present invention provides a kit comprising a cannabinoid composition, a mushroom composition, and optionally instructions for administration of said compositions, as a combined treatment, for providing an adjunctive therapy to asubject having a breast cancer and undergoing a conventional therapy for treating said breast cancer, wherein: (i) said cannabinoid composition comprises CBD or an enantiomer, diastereomer, or a mixture thereof; A 9 -THC or an enantiomer, diastereomer, or a mixture thereof; CBDV or an enantiomer, diastereomer, or a mixture thereof; CBDA or an enantiomer, diastereomer, or a mixture thereof; and optionally one or more terpenes; and (ii) said mushroom composition comprises a mixture of Agaricus bisporus extract, Ganoderma lucidum extract, Ganoderma lucidum spore extract, Grifola frondosa extract, Lentinus e
  • said cannabinoid composition comprises one or more terpenes, e.g., at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene, constituting up to about 3%, e.g., from about 1% to about 2%, by weight of said composition.
  • terpenes e.g., at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene
  • Figs. 1A-1C show the effect of a combination of CBD, A 9 -THC, CBDV, and CBDA on cell viability (%) as compared to each one of the cannabinoids when administered alone, in various breast cancer cell lines, more specifically MCF7 (1A), MDA-MB231 (IB), and SKBR3 (1C).
  • the concentration of all cannabinoids is in the low pM range ( ⁇ 10 pM).
  • Fig. 2 shows the effect of a combined treatment of a cannabinoid composition and taxol-based chemotherapy on cell viability (%) as compared to each one of the cannabinoid composition and taxol-based chemotherapy, when administered alone, in MCF7 (left panel) and SKBR3 (right panel) breast cancer cell lines.
  • Fig. 3 shows the effect of a combined treatment of a mushroom composition and taxol-based chemotherapy on cell viability (%) as compared to each one of the mushroom composition and taxol-based chemotherapy, when administered alone, in MCF7 (left panel) and SKBR3 (right panel) breast cancer cell lines.
  • Fig. 4 shows the effect of a combined treatment of a mushroom composition and carboplatin-based chemotherapy on cell viability (%) as compared to each one of the mushroom composition and carboplatin-based chemotherapy, when administered alone, in MDA-MB231 breast cancer cell line, at carboplatin concentration of 300 pM (left panel) and 1000 pM (right panel).
  • cannabinoid refers to a chemical compound acting on a cannabinoid receptor, i.e., cannabinoid type 1 (CB1) and/or cannabinoid type 2 (CB2) receptor.
  • CB1 cannabinoid type 1
  • CB2 cannabinoid type 2
  • Ligands for these receptor proteins include the endocannabinoids produced naturally in the body; the phytocannabinoids found in Cannabis saliva. Cannabis indica. Cannabis ruderalis. and some other plants; and synthetic cannabinoids.
  • cannabinoid composition denotes a composition, e.g., a nutraceutical or pharmaceutical composition, comprising the four specific cannabinoids listed above, i.e., CBD (2-[(17?,67?)-3-methyl-6-prop-l-en-2-ylcyclohex-2-en-l-yl]-5-pentylbenzene-l,3- diol) or an enantiomer, diastereomer, or a mixture (e.g., racemate) thereof; A 9 -THC (6,6,9- trimethyl-3-pentyl-6a,7,8,10a-tetrahydrobenzo[c] chromen-l-ol) or an enantiomer, diastereomer, or a mixture (e.g., racemate) thereof; CBDV (2-[( l /?,6/?)-3-mcthyl-6-prop-l - en-2-ylcyclohex-2
  • Preferred cannabinoid compositions comprise CBD, A 9 -THC, CBDV and CBDA. Yet, for the sake of simplicity and if not otherwise explicitly specified, all references made throughout this specification to any one of CBD, A 9 -THC, CBDV and CBDA independently refers to an enantiomer, diastereomer, or a mixture, e.g., racemate, of said cannabinoid.
  • the overall amount by weight of the cannabinoids composing the cannabinoid composition i.e., the sum of weight amounts of CBD, A 9 -THC, CBDV, CBDA, is referred to herein as “total cannabinoids”.
  • a 9 -THC which is the primary psychotropic constituent of cannabis, is associated with undesirable effects such as tachycardia, anxiety, altered cognitive perception, and other behavioral issues, and has a maximum tolerated daily dose of only 15-90 mg in adults.
  • a 9 -THC reduces cell viability, cell proliferation, invasion and tumor growth both in vitro and in vivo (Takeda et al., 2013; Ligresti et al., 2006; Murase et al., 2014).
  • a 9 -THC inhibits the growth and proliferation of cancerous cells through the modulation of cannabinoid receptors (CB1-R and CB2-R), and is capable of reducing human breast cancer cell proliferation by blocking the progression of the cell cycle and inducing apoptosis through activation of CB2-R (Caffarel et al., 2006).
  • a 9 -THC reduced tumor growth and the amount/severity of lung metastases in a genetically engineered animal model of ErbB2-driven metastatic breast cancer (MMTV-neu mice) (Caffarel et al., 2010); and disrupted human epidermal growth factor receptor 2 (HER2)- CB2-R complexes by selectively binding to CB2-R, which leads to the inactivation of HER2 through disruption of HER2-HER2 homodimers, and the subsequent degradation of HER2.
  • HER2 human epidermal growth factor receptor 2
  • Such activity triggers antitumor responses in vitro and in vivo (Blasco-Benito et al., 2019).
  • a 9 -THC may alter the immune system function and increase the susceptibility of a patient to microbial infections; and in spite of its demonstrated anti-cancer properties, under certain circumstances it may promote tumor growth, invasion and metastasis in some cancer cell types.
  • a 9 -THC-mediated increased tumor growth and metastasis was associated with inhibition of the anti-tumor specific immune responses in vivo (McKallip et al., 2005).
  • a retrospective analysis further demonstrated that cannabis use is associated with the markedly reduced effectiveness of immune checkpoint inhibitors (Taha et al., 2019).
  • CBD which is the second most abundant cannabinoid in cannabis
  • CBD has demonstrated robust anti-proliferative and pro-apoptotic effects on a wide variety of cancer types, including breast cancer, both in cultured cancer cell lines and in mouse tumor models, while having milder effects on normal cells from the same tissue/organ (McAllister et al., 2007; Shrivastava et al., 2011; Sultan et al., 2018).
  • CBD induced significant cell death in human breast adenocarcinoma cell line MDA-MB-231 after 24 h at low doses of 1 to 5 pM, inhibited the survival of both estrogen receptor-positive and estrogen receptor-negative breast cancer cell lines, and induced apoptosis in a concentration-dependent manner. Moreover, at said concentrations, CBD had little effect on the nontumorigenic mammary (MCF-10A) cells (Kosgodage et al., 2018).
  • CBD can also inhibit tumor migration, invasion and neovascularization, suggesting that it not only acts on tumor cells but can also affect the tumor microenvironment, e.g., by modulating infiltrating mesenchymal cells and immune cells.
  • CBDA anticancer activity of CBDA has been preliminarily tested on CEM (acute lymphocytic leukemia) and HL60 (promyelocytic leukemia) cells, and the effect of CBDA was evaluated on cell viability, cell proliferation, and cell-cycle dynamics. Data from those experiments, as well as from MTT assay on human prostate carcinoma androgen receptorpositive (LNCaP) cells (De Petrocellis et al., 2013), evidenced that CBDA was less active than CBD.
  • CEM acute lymphocytic leukemia
  • HL60 promyelocytic leukemia
  • CBDA inhibits the enzymatic activity of cyclooxygenase-2 (COX-2), an enzyme involved in inflammatory processes and in the metastasis of cancer cells, with an IC50 value of 2.2 M, and diminishes the expression of COX-2 in human breast cancer MDA-MB-231 cells, a highly aggressive triple-negative breast cancer cell line (Formato et al., 2020; Takeda et al., 2017).
  • COX-2 cyclooxygenase-2
  • CBDA inhibited the MDA-MB-231 breast cancer cell migration, and the mechanism responsible for the inhibitory effects likely involves activation of small GTPase RhoA via inhibition of a c AMP-dependent protein kinase (PKA) (Takeda et al., 2012).
  • PKA c AMP-dependent protein kinase
  • CBDV is a minor cannabinoid and is a structural analogue of CBD, more specifically a propyl analogue of CBD. Due to the absence of psychotropic activity and its promising utility as an anticonvulsive agent, CBDV has a great therapeutic potential.
  • CBDV is being tested in pre-clinical studies for autism spectrum disorders (ASD) and childhood epilepsy (Alves et al., 2020).
  • CBDV was found as a potent inhibitor of 1-a- lysophosphatidylinositol (LPI)-induced G protein-coupled receptor 55 (GPR55) signaling.
  • LPI 1-a- lysophosphatidylinositol
  • GPR55 G protein-coupled receptor 55
  • each one of the cannabinoids comprised within the cannabinoid composition may independently derive from a Cannabis extract, e.g., Cannabis Sativa extract, using any suitable extraction and purification procedures known in the art, or may alternatively be synthesized following any one of the procedures disclosed in the literature.
  • a Cannabis extract e.g., Cannabis Sativa extract
  • the cannabinoid composition comprises CBD in an amount of from about 12% to about 32%, preferably from about 17% to about 27%, more preferably from about 20% to about 24%, e.g., from about 21% to about 23%, by weight of total cannabinoids;
  • a 9 -THC in an amount of from about 3% to about 23%, preferably from about 8% to about 18%, more preferably from about 11% to about 15%, e.g., from about 12% to about 14%, by weight of total cannabinoids;
  • CBDV in an amount of from about 33% to about 53%, preferably from about 38% to about 48%, more preferably from about 41% to about 45%, e.g., from about 42% to about 44%, by weight of total cannabinoids;
  • CBDA in an amount of from about 12% to about 32%, preferably from about 17% to about 27%, more preferably from about 20% to about 24%, e.g., from about 21%
  • said cannabinoid composition comprises CBD in an amount of about 22% by weight of total cannabinoids; A 9 -THC in an amount of about 13% by weight of total cannabinoids; CBDV in an amount of about 43% by weight of total cannabinoids; and CBDA in an amount of about 22% by weight of total cannabinoids.
  • the cannabinoid composition according to any one of the embodiments above comprises one or more, e.g., two, three, four, or five, terpenes constituting up to about 3%, e.g., from about 0.25% to about 2.5%, from about 0.5% to about 2%, from about 0.75% to about 1.5%, or about 1%, by weight of said cannabinoid composition.
  • total terpenes refers to the overall amount by weight of the terpenes, i.e., the sum of weight amounts of all terpenes comprised within said cannabinoid composition.
  • terpene refers to a hydrocarbon produced by a variety of plants and by some insects. Terpenes often have a strong odor and may protect the plants producing them by deterring herbivores and by attracting predators and parasites of herbivores. Terpenes are also major biosynthetic building blocks. Steroids, for example, are derivatives of the triterpene squalene. Modified terpenes, i.e., terpenes containing a functional group, usually an oxygen-containing group, are called terpenoids or isoprenoids. The terpenes optionally comprised within the cannabinoid composition as defined herein may independently be either naturally produced and optionally purified, or synthetic.
  • Terpenes and terpenoids are the primary constituents of the essential oils of many types of plants and flowers.
  • Essential oils are used widely as fragrances in perfumery and traditional medicine, such as aromatherapy.
  • Synthetic variations and derivatives of natural terpenes and terpenoids also greatly expand the variety of aromas used in perfumery and flavors used in food additives.
  • Monoterpenes are a class of terpenes consisting of two isoprene units and having the molecular formula C10H16. Monoterpenes may be linear (acyclic) or contain rings (cyclic). Modified terpenes, such as those containing oxygen functionality or missing a methyl group, are called monoterpenoids. Monoterpenes and monoterpenoids are used in the pharmaceutical, cosmetic, agricultural, and food industries.
  • Non-limiting examples of monoterpenes or monoterpenoids comprised within the terpene combination of the invention include a pinene selected from a-pinene, P-pinene, and y-pinene, limonene, linalool, myrcene, camphene, nerol, geraniol, a terpineol selected from a-terpineol, P- terpineol, y-terpineol, and terpinen-4-ol, or an enantiomer or diastereomer thereof.
  • Sesquiterpenes are a class of terpenes consisting of three isoprene units and often have the molecular formula C15H24. Like monoterpenes, sesquiterpenes may be acyclic or contain rings, including many unique combinations. Biochemical modifications such as oxidation or rearrangement produce the related sesquiterpenoids.
  • Non-limiting examples of sesquiterpene or sesquiterpenoid include longifolene, copaene, patchoulol, farnesol, humulene, famesene, P-caryophyllene, or an enantiomer or diastereomer thereof.
  • said one or more terpenes comprises at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene.
  • said terpenes comprise three or four of the specific terpenes listed above, i.e., linalool, limonene and a-pinene; linalool, limonene and P-caryophyllene; linalool, limonene and P-myrcene; limonene, a-pinene and P-caryophyllene; limonene, a-pinene and P-myrcene; a-pinene, P-caryophyllene, and P-myrcene; linalool, limonene, a-pinene, and P-caryophyllene; linalool, limonene, a-pinen
  • Preferred embodiments are those wherein said terpenes comprise all five specific terpenes listed above, i.e., linalool, limonene, a-pinene, P- caryophyllene, and P-myrcene, preferably at equal amounts, i.e., wherein each of said terpenes constitutes about 20% by weight of total terpenes.
  • Medicinal mushrooms have become an attractive source of functional food and therapeutic products mainly because of their chemical composition. It is established that, undoubtedly, mushrooms have the greatest potential for medical and pharmaceutical use due to their ability to synthesize many highly beneficial bioactive compounds and could be a great source of bioactive substances such as essential amino acids, fatty acids, dietary fiber, polysaccharides (mainly P-glucans), vitamins, micro- and macro -elements, lectins, terpenoids, steroids, statins, phenols, alkaloids, and antibiotics (Cohen et al., 2014, Wasser 2018, Xue et al., 2020).
  • bioactive substances such as essential amino acids, fatty acids, dietary fiber, polysaccharides (mainly P-glucans), vitamins, micro- and macro -elements, lectins, terpenoids, steroids, statins, phenols, alkaloids, and antibiotics (Cohen et al., 2014, Wasser 2018, Xue et al., 2020).
  • Medicinal mushrooms are comparable to “medicinal plants” and can be defined as macroscopic fungi, mostly higher Basidiomycetes and some Ascomycetes, which are used in the form of extracts or powder for prevention, alleviation, or healing multiple diseases, and/or in balancing a healthy diet.
  • herebal drugs dried fruit bodies, my celia, or spores are considered “mushroom drugs” or “fungal drugs”.
  • the resulting mushrooms preparations should be considered as “mushroom pharmaceuticals”, “mushroom drugs” or “mushroom preparations”.
  • New class of drugs were developed from MMs called “mushroom pharmaceuticals or mushroom drugs”, or biological response modifiers (BRMs) like krestin (PSK) and PSP (polysaccharide peptide) from Trametes versicolor, lentinan, isolated from Lentinus edodes', schizophyllan (sonifilan, sizofiran, or SPG) from Schizophyllum commune', befungin from Inonotus obliquus', D-fraction from Grifola frondosa, GLPS polysaccharide fraction from Ganoderma lucidum', active hexose correlated compound (AHCC), and some others (Wasser, 2010, Chang and Wasser, 2018).
  • BRMs biological response modifiers
  • PSK krestin
  • PSP polysaccharide peptide
  • lentinan isolated from Lentinus edodes'
  • schizophyllan sonifilan, sizofiran, or
  • muscle composition denotes a composition, e.g., a nutraceutical or pharmaceutical composition, comprising extracts obtained from the five medicinal mushrooms listed above, i.e., Agaricus bisporus, Ganoderma lucidum, Grifola frondosa, Lentinus edodes, and Trametes versicolor.
  • the overall amount by weight of the extracts composing the mushroom composition i.e., the sum of weight amounts of Agaricus bisporus extract, Ganoderma lucidum extract, Ganoderma lucidum spore extract, Grifola frondosa extract, Lentinus edodes extract, and Trametes versicolor extract, is referred to herein as “total extracts”.
  • Agaricus bisporus extract refers to an extract of Agaricus bisporus fruiting bodies, which is rich in polysaccharides, i.e., comprises at least 27%, preferably at least 30%, polysaccharides originated from said mushroom.
  • said extract is obtained by extracting fruiting bodies of said mushroom with water, e.g., at a temperature in a range of 20-90°C, preferably 30-70°C.
  • said extract is the commercial product manufactured by Zhejiang Suichang Limin Pharmaceutical Co. Ltd., Yunfeng Food Branch, China, which includes at least 30% polysaccharides, about 25% proteins, and about 0.5% fats.
  • Ganoderma lucidum extract refers to an extract of Ganoderma lucidum fruiting bodies and/or mycelium, which is rich in polysaccharides and triterpenes (also referred to as “ganoderic acids”), i.e., comprises at least 24%, preferably at least 27%, polysaccharides, and at least 4%, preferably at least 4.5%, triterpenes, originated from said mushroom.
  • said extract is obtained by extracting fruiting bodies and/or mycelium of said mushroom with an alcohol such as ethanol, at a temperature in a range of 20-90°C, preferably 30-70°C.
  • said extract is the commercial product (GLAE) manufactured by Zhejiang Suichang Limin Pharmaceutical Co. Ltd., Yunfeng Food Branch, China, which includes at least 30% polysaccharides, about 25% proteins, at least 5% triterpenes, and about 0.5% fats.
  • GLAE commercial product manufactured by Zhejiang Suichang Limin Pharmaceutical Co. Ltd., Yunfeng Food Branch, China, which includes at least 30% polysaccharides, about 25% proteins, at least 5% triterpenes, and about 0.5% fats.
  • Ganoderma lucidum spore extract refers to an extract of Ganoderma lucidum spores/fruiting bodies, which is rich in adenosine/triterpene, i.e., comprises at least 16%, preferably at least 20%, adenosine/triterpenes originated from said mushroom (such extract is also referred to herein as “ Ganoderma lucidum spore oil”).
  • said extract is obtained by extracting spores/fruiting bodies of said mushroom with CO2.
  • said extract is the commercial product manufactured by Hangzhou Johncan Mushroom Bio-Technology Co.
  • Grifola frondosa extract refers to an extract of Grifola frondosa fruiting bodies, which is rich in beta-glucans and polysaccharides, i.e., comprises at least 40%, preferably at least 45%, beta-glucans; and at least 28%, preferably at least 32%, polysaccharides, originated from said mushroom.
  • said extract is obtained by extracting fruiting bodies of said mushroom with water, e.g., at a temperature in a range of 20-90°C, preferably 30-70°C (such extract is also referred to herein as “Grifola frondosa D-fraction extract”, indicating protein-bound p-glucans).
  • said extract is the commercial product manufactured by Hangzhou Johncan Mushroom Bio-Technology Co. Ltd., China, which includes about 51% beta-glucans and about 36% polysaccharides.
  • Lentinus edodes extract refers to an extract of Lentinus edodes fruiting bodies, which is rich in polysaccharides, i.e., comprises at least 28%, preferably at least 32%, polysaccharides originated from said mushroom.
  • said extract is obtained by extracting fruiting bodies of said mushroom with water, e.g., at a temperature in a range of 20-90°C, preferably 30-70°C.
  • said extract is the commercial product manufactured by Zhejiang Suichang Limin Pharmaceutical Co. Ltd., Yunfeng Food Branch, China, which includes at least 30% polysaccharides, about 24.5% proteins, and about 0.5% fats.
  • Trametes versicolor extract refers to an extract of Trametes versicolor mycelium, which is rich in polysaccharides, i.e., comprises at least 24%, and preferably at least 27%, polysaccharides originated from said mushroom.
  • said extract is obtained by extracting mycelium of said mushroom with water, e.g., at a temperature in a range of 20-90°C, preferably 30-70°C, and filtering the crude product thus obtained to obtain a product having at least 50% of polysaccharides having a molecular weight greater than 40000 Dalton (such extract is also referred to herein as “Trametes versicolor pure krestin extract”).
  • said extract is the commercial product manufactured by Zhejiang Suichang Limin Pharmaceutical Co. Ltd., Yunfeng Food Branch, China, which includes at least 30% polysaccharides, about 25% proteins, and about 0.5% fats.
  • the mushroom composition comprises an Agaricus bisporus extract in an amount of from about 5% to about 25%, preferably from about 10% to about 20%, more preferably from about 13% to about 17%, e.g., from about 14% to about 16%, by weight of total extracts, a Ganoderma lucidum extract in an amount of from about 18% to about 38%, preferably from about 23% to about 33%, more preferably from about 26% to about 30%, e.g., from about 27% to about 29%, by weight of total extracts, a Ganoderma lucidum spore extract in an amount of from about 1% to about 3%, preferably from about 1.5% to about 2.5%, more preferably from about 1.75% to about 2.25%, by weight of total extracts, a Grifola frondosa extract in an amount of from about 4% to about 24%, preferably from about 9% to about 19%, more preferably from about 12% to about 16%, e.g.,
  • said mushroom composition comprises Agaricus bisporus extract in an amount of about 15% by weight of total extracts, Ganoderma lucidum extract in an amount of about 28% by weight of total extracts, Ganoderma lucidum spore extract in an amount of about 2% by weight of total extracts, Grifola frondosa extract in an amount of about 14% by weight of total extracts, Lentinus edodes extract in an amount of about 23% by weight of total extracts, and Trametes versicolor extract in an amount of about 19% by weight of total extracts.
  • the method of the present invention comprises administering to said subject, as an adjunctive therapy, a cannabinoid composition, mushroom composition, or combination thereof, wherein said cannabinoid composition comprises CBD in an amount of from about 12% to about 32%, preferably from about 17& to about 27%, more preferably from about 20% to about 24%, e.g., from about 21% to about 23%, by weight of total cannabinoids;
  • a 9 -THC in an amount of from about 3% to about 23%, preferably from about 8% to about 18%, more preferably from about 11% to about 15%, e.g., from about 12% to about 14%, by weight of total cannabinoids;
  • CBDV in an amount of from about 33% to about 53%, preferably from about 38% to about 48%, more preferably from about 41% to about 45%, e.g., from about 42% to about 44%, by weight of total cannabinoids;
  • CBDA in an amount of from about 12%
  • said cannabinoid composition comprises CBD in an amount of about 22% by weight of total cannabinoids; A 9 -THC in an amount of about 13% by weight of total cannabinoids; CBDV in an amount of about 43% by weight of total cannabinoids; and CBDA in an amount of about 22% by weight of total cannabinoids; and said mushroom composition comprises an Agaricus bisporus extract in an amount of about 15% by weight of total extracts, a Ganoderma lucidum extract in an amount of about 28% by weight of total extracts, a Ganoderma lucidum spore extract in an amount of about 2% by weight of total extracts, a Grifola frondosa extract in an amount of about 14% by weight of total extracts, a Lentinus edodes extract in an amount of about 23% by weight of total extracts, and a Trametes versicolor extract in an amount of about 19% by weight of total extracts.
  • the cannabinoid composition referred to hereinabove comprises one or more terpenes constituting up to about 3%, e.g., from about 0.25% to about 2.5%, from about 0.5% to about 2%, from about 0.75% to about 1.5%, or about 1%, by weight of said cannabinoid composition.
  • said one or more terpenes comprises at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene, more particularly three or four of said terpenes, i.e., linalool, limonene and a- pinene; linalool, limonene and P-caryophyllene; linalool, limonene and P-myrcene; limonene, a-pinene and P-caryophyllene; limonene, a-pinene and P-myrcene; a-pinene, P- caryophyllene, and P-myrcene; linalool, limonene, a-pinene, and P-caryophyllene; linalool, limonene, a-pinene, P-myrcene,
  • said terpenes comprises linalool, limonene, a-pinene, P- caryophyllene, and P-myrcene, preferably at equal amounts, i.e., wherein each of said terpenes constitutes about 20% by weight of total terpenes.
  • compositions or compositions administered according to the method disclosed herein may be prepared by conventional techniques, e.g., as described in Remington: The Science and Practice of Pharmacy, 19 th Ed., 1995. More specifically, such compositions may be prepared, e.g., by uniformly and intimately bringing the active agents, i.e., the cannabinoids or the mixture of mushroom extracts, into association with a liquid carrier, a finely divided solid carrier, or both, and then, if necessary, shaping the product into the desired formulation.
  • the compositions may independently be in liquid, solid or semisolid form and may further include various acceptable fillers, carriers, diluents or adjuvants, and other inert ingredients and excipients.
  • each one of the compositions administered may independently be either a pharmaceutical or nutraceutical composition, optionally further comprising a pharmaceutically or nutraceutically acceptable carrier, respectively.
  • Particular such compositions are formulated for either oral or oral mucosal (e.g., sublingual, supra-lingual, or buccal) administration, or for inhalation. More particular such compositions are formulated as a liquid dosage form, e.g., a solution, emulsion, syrup, or elixir; or as a solid dosage form, e.g., a tablet, capsule, or pill.
  • both the cannabinoid composition and the mushroom compositions are nutraceutical compositions formulated each independently as either a liquid dosage form (e.g., a solution, emulsion, syrup, or elixir); or as a solid dosage form (e.g., a tablet, capsule, or pill).
  • a liquid dosage form e.g., a solution, emulsion, syrup, or elixir
  • solid dosage form e.g., a tablet, capsule, or pill
  • the method of the present invention comprises administering to said subject, as an adjunctive therapy, only one of the compositions referred to above, i.e., either a cannabinoid composition or a mushroom composition, each according to any one of the embodiments above.
  • the method of the present invention comprises administering to said subject, as an adjunctive therapy, a combination of said cannabinoid composition and said mushroom composition, according to any one of the embodiments above.
  • the cannabinoid composition and the mushroom composition administered as a combination may be formulated as two separate compositions for the same or different administration mode, and may thus be administered concomitantly, or sequentially at any order.
  • said compositions may be formulated as a sole composition, e.g., a sole pharmaceutical or nutraceutical composition.
  • the compositions disclosed herein are for use either alone or in combination, as an adjunctive therapy to a conventional therapy aimed at treating a breast cancer.
  • inventions and “conventional treatment”, as used herein interchangeably, refer to any conventional treatment of cancer, more specifically breast cancer, by the use of a chemotherapy, hormonotherapy, immunotherapy, tyrosine kinase inhibitor-based therapy, antiangiogenic based-therapy, antibody-based therapy, or any combination thereof.
  • chemotherapy “conventional chemotherapeutic treatment”, and “chemotherapy”, as used herein interchangeably, refer to a chemotherapy-based conventional therapy for treatment of breast cancer, and mean administration of one or more chemotherapeutic agents/drugs, to a subject in need thereof, following any treatment regimen known in the art.
  • the conventional therapy referred to herein is a chemotherapy comprising administration of a taxane, e.g., paclitaxel (Taxol®), albumin paclitaxel (Abraxane®), and docetaxel (Taxotere®); a nonsteroidal aromatase inhibitor, e.g., anastrozole (Arimidex®), letrozole (Femara®), and tamoxifen (Soltamox®); an anthracycline, e.g., doxorubicin (Adriamycin®), liposomal doxorubicin (Doxil®), and epirubicin (Ellence®); platinum-based agents, e.g., cisplatin and carboplatin; or antibody drug conjugates, e.g., ado-trastuzumab emtansine (Kadcyla®), fam-t
  • a taxane
  • adjunct therapy refers to a therapy given to a subject having a breast cancer and undergoing a conventional therapy for treating said breast cancer, in addition and in parallel to said conventional therapy, so as to increase or maximize the efficacy of said conventional therapy.
  • the breast cancer treated by the method disclosed herein may be any type of breast cancer currently known such as, without being limited to, luminal A breast cancer, luminal B breast cancer, human epidermal growth factor receptor 2 (HER2) -positive breast cancer, and triple-negative breast cancer.
  • HER2 human epidermal growth factor receptor 2
  • subject refers to any mammal, e.g., a human, nonhuman primate, horse, ferret, dog, cat, cow, and goat. In a preferred embodiment, the term “subject” denotes a human, i.e., an individual.
  • the present invention relates to a cannabinoid composition, mushroom composition, or a combination thereof, for use as an adjunctive therapy to a conventional therapy, e.g., a chemotherapy, in the treatment of a breast cancer
  • a conventional therapy e.g., a chemotherapy
  • said cannabinoid composition comprises CBD, A 9 -THC, CBDV, CBDA, and optionally one or more terpenes
  • said mushroom composition comprises a mixture of extracts, more specifically an Agaricus bisporus extract, a Ganoderma lucidum extract, a Ganoderma lucidum spore extract, a Grifola frondosa extract, a Lentinus edodes extract, and a Trametes versicolor extract.
  • the cannabinoid composition and the mushroom composition used, either alone or in combination, as an adjunctive therapy to a conventional therapy, according to this aspect of the present invention, are each as defined in any one of the embodiments above.
  • said cannabinoid composition comprises CBD in an amount of from about 12% to about 32%, preferably from about 17% to about 27%, more preferably from about 20% to about 24%, e.g., from about 21% to about 23%, by weight of total cannabinoids;
  • a 9 -THC in an amount of from about 3% to about 23%, preferably from about 8& to about 18%, more preferably from about 11% to about 15%, e.g., from about 12% to about 14%, by weight of total cannabinoids;
  • CBDV in an amount of from about 33% to about 53%, preferably from about 38% to about 48%, more preferably from about 41% to about 45%, e.g., from about 42% to about 44%, by weight of total cannabinoids;
  • CBDA in an amount of from about 12% to about 32%, preferably from about 17% to about 27%, more preferably from about 20% to about 24%, e.g., from about 21%
  • said cannabinoid composition comprises CBD in an amount of about 22% by weight of total cannabinoids; A 9 -THC in an amount of about 13% by weight of total cannabinoids; CBDV in an amount of about 43% by weight of total cannabinoids; and CBDA in an amount of about 22% by weight of total cannabinoids; and said mushroom composition comprises an Agaricus bisporus extract in an amount of about 15% by weight of total extracts, a Ganoderma lucidum extract in an amount of about 28% by weight of total extracts, a Ganoderma lucidum spore extract in an amount of about 2% by weight of total extracts, a Grifola frondosa extract in an amount of about 14% by weight of total extracts, a Lentinus edodes extract in an amount of about 23% by weight of total extracts, and a Trametes versicolor extract in an amount of about 19% by weight of total extracts.
  • the cannabinoid composition referred to hereinabove comprises one or more terpenes constituting up to about 3%, e.g., from about 0.25% to about 2.5%, from about 0.5% to about 2%, from about 0.75% to about 1.5%, or about 1%, by weight of said cannabinoid composition.
  • said one or more terpenes comprises at least two of linalool, limonene, a-pinene, P-caryophyllene, and P-myrcene, more particularly three or four of said terpenes, i.e., linalool, limonene and a- pinene; linalool, limonene and P-caryophyllene; linalool, limonene and P-myrcene; limonene, a-pinene and P-caryophyllene; limonene, a-pinene and P-myrcene; a-pinene, P- caryophyllene, and P-myrcene; linalool, limonene, a-pinene, and P-caryophyllene; linalool, limonene, a-pinene, P-myrcene,
  • said terpenes comprises linalool, limonene, a-pinene, P- caryophyllene, and P-myrcene, preferably at equal amounts, i.e., wherein each of said terpenes constitutes about 20% by weight of total terpenes.
  • the present invention relates to only one of the compositions referred to above, i.e., either a cannabinoid composition or a mushroom composition, each according to any one of the embodiments above, for use as an adjunctive therapy to a conventional therapy in the treatment of a breast cancer.
  • the present invention relates to a combination of said cannabinoid composition and said mushroom composition, according to any one of the embodiments above, for use as an adjunctive therapy to a conventional therapy in the treatment of a breast cancer.
  • said compositions may be formulated as two separate compositions for the same or different administration mode, which are to be administered concomitantly, or sequentially at any order.
  • said compositions may be formulated as a sole composition, e.g., a sole pharmaceutical or nutraceutical composition.
  • the present invention provides a kit comprising a cannabinoid composition, a mushroom composition, and optionally instructions for administration of said compositions, as a combined treatment, for providing an adjunctive therapy to a subject having a breast cancer and undergoing a conventional therapy for treating said breast cancer, wherein: (i) said cannabinoid composition comprises CBD, A 9 -THC, CBDV, CBDA, and optionally one or more terpenes; and (ii) said mushroom composition comprises a mixture of extracts, more specifically Agaricus bisporus extract, Ganoderma lucidum extract, Ganoderma lucidum spore extract, Grifola frondosa extract, Lentinus edodes extract, and Trametes versicolor extract.
  • said cannabinoid composition comprises CBD, A 9 -THC, CBDV, CBDA, and optionally one or more terpenes
  • said mushroom composition comprises a mixture of extracts, more specifically Agaricus bisporus extract, Ganoderma lucidum
  • Mushroom extracts Trametes versicolor Pure Krestin Extract (lot number S200528001), Lentinus edodes extract (lot number S200408002), Agaricus bisporus extract (lot number S200408002), and Ganoderma lucidum extract (GLAE, lot number S201016002) were supplied by Zhejiang Suichang Limin Pharmaceutical Co. Ltd. Yunfeng Food Branch, China.
  • Grifola frondosa D-fraction extract (Matakei Extract, batch number 0/HSH-FE20200901-S08,) and Ganoderma lucidum spore extract (Reishi spore oil, batch number 0/LZBZY-FT20200701-S13) were supplied by Hangzhou Johncan Mushroom Bio-Technology Co. Ltd., China.
  • Cannabinoids for the studies described in Examples 1-4 were provided by Lumir Lab, Israel.
  • the THC used in study described in Example 5 is dronabinol (a synthetic product); CBD (>98%) was provided by Mile High Labs (International Ltd, Falcon Way, Harbor, Northern Ireland, BT12 6SQ); and CBDA (>97%) and CBDV (99.9%) were provided by Precision Plant Molecules (PPM, 2300 E 76 th Avenue Suite D-100, Welby, CO 80229 United States).
  • the cannabinoid composition was prepared by re-suspending the cannabinoids CBDA, CBD, CBDV, and THC in dimethyl sulfoxide (DMSO), to obtain a stock concentration of 100 mM for each one of the cannabinoids.
  • DMSO dimethyl sulfoxide
  • Cells culture and treatments were cultured according to standard mammalian tissue culture protocols and sterile technique.
  • Human adenocarcinoma breast cancer cell lines MCF-7, MDA-MB-231, SK-BR-3, and BT-747 were cultured in cellspecific media (DMEM for MCF-7 and MDA-MB-231 cells, McCoy's 5A for SK-BR-3 cells, and Hybri-care media for BT-747). All media were supplemented with 10% fetal bovine serum, streptomycin (100 mg/ml), penicillin (100 U/ml) and nystatin (12.5 U/ml). Cells were incubated in 5% CO2 at 37°C.
  • MTT MTT standard protocol
  • the media was removed, and all cells were incubated with serum-free media containing 0.5 mg/ml MTT for 4 hours at the incubator.
  • the MTT purple crystals formed by the viable cells were dissolved using isopropanol containing 0.04 mol/L HC1.
  • the quantification was determined by measuring the optical density at 570 nm in a spectrophotometer reader (Spark, Tecan). Results were presented as proportional viability (%) by comparing between treated and untreated groups.
  • Example 4 The combined effect of each one of the cannabinoid- and mushroomcompositions with a chemotherapeutic agent on selected breast cancer cell lines
  • the combined effect of the cannabinoid composition and taxol on MCF7 and SKBR3 cell lines [0074] Results.
  • the combined treatment had synergistic effect as compared to the administration of the cannabinoid composition and taxol only, and reduced the MCF7 cells viability to about 40%.
  • Example 5 The anti-neoplastic activity of combinations of the cannabinoid- and mushroom compositions, administered with taxol, on breast cancer samples
  • the present study is aimed at testing the anti-neoplastic activity of a cannabinoid composition as disclosed herein, more specifically the composition shown in Table 4 and identified herein as CNTC 10401; a mushroom composition as disclosed herein, more specifically the composition shown in Table 5 and herein identified CNTC20401; when administered separately, in combination, or in combination and in addition to taxol, on breast cancer samples (ER+/Her2-, triple negative (TN)), i.e., individual patient’s tumor tissue obtained by either biopsy or surgery.
  • a cannabinoid composition as disclosed herein, more specifically the composition shown in Table 4 and identified herein as CNTC 10401
  • a mushroom composition as disclosed herein, more specifically the composition shown in Table 5 and herein identified CNTC20401
  • ER+/Her2-, triple negative (TN) i.e., individual patient’s tumor tissue obtained by either biopsy or surgery.
  • cResponseTM platform https://www.curesponse.com
  • TME tumor microenvironment
  • the cResponseTM platform also allows for long-term tumor culture with high viability and tissue preservation. This is important since many drugs require several days in culture to achieve maximum efficacy. Using this platform enables assessing the response of various treatments and treatment combinations on a single specimen - reflecting the complexity of real- world patients.
  • Comparison of treatments/concentrations using the cResponseTM platform is based on a score, which reflects the responsiveness of the tissue to the treatment, and is determined by curesponse proprietary algorithm integrating data from a panel of immunohistochemical markers.
  • the scores range from 0 to 100, and categorized as: no response: 0-30, weak response: 31-55, moderate response: 56-80, and strong response: 81- 100.
  • concentration A demonstrated a strong response
  • concentrations B demonstrated a weak response
  • concentrations C-E showed no response.
  • Working concentration B was thus elected for further studies.
  • the weak response shown by that concentration is suitable for interaction testing by allowing a large enough margin for demonstrating synergy if exists with the other materials tested (i.e., mushroom composition, taxol).
  • formulation 1 demonstrated a moderate response and formulation 2 demonstrated a strong response. Based on the results we suspected that DMSO may interfere with the activity of components in the extract and formulation 2 was thus elected for further studies. Furthermore, in a combination testing, each of the combined substances when tested alone should give a weak response such that a strong response is only observed when all tested items are combined. As such, based on the strong response observed for the tested concentration of formulation 2, a decision to proceed to the main experiment with a 1:2 dilution of the tested concentration was taken.
  • Cancer samples are brought to the Curesponse laboratory within 6 hours of harvest in preservation medium. Upon arrival in the laboratory, background details and images of the sample are documented. A DNA sample is frozen for later analysis.
  • Tissue is sectioned into multiwell 12 well cResponse plates and cultured in Curesponse’ s proprietary culture medium (Day 0). 3. The following day (Day 1), drugs are added according to the following (in duplicates).
  • Taxol is used at a low concentration, which indicates a range of lOpM to 40pM such that very sensitive tissues may demonstrate a response to that concentration of Taxol, but the majority will not.
  • Culture medium are collected on day 1 (pre-treatment) and day 3 (post-treatment) and frozen at -20C for multiplex ELISA analysis of cytokine secretion (TNF-a, Interferon gamma, IL2, IL4, IL6, IL8, IL10, Granzyme B, , TGFb, VEGF).
  • the combined expected to be effect achieved by the combination of the compositions is expected to be further enhanced when applying said combination together with taxol, demonstrating a synergistic effect between said combination and said chemotherapeutic drug, as shown above in vitro.
  • the combination of the two compositions, when applied together with taxol, is expected to show more founded synergistic effect, and display enhanced cell death and modulation of the immune microenvironment.
  • Non-THC cannabinoids inhibit prostate carcinoma growth in vitro and in vivo: Pro-apoptotic effects and underlying mechanisms, Br. J. Pharmacol. 2013, 168, 79-102 Formato M., Crescente G., Scognamiglio M., Fiorentino A., Pecoraro M.T., Piccolella S., Catauro M., Pacifico S. (-)-Cannabidiolic acid, a still overlooked bioactive compound: An introductory review and preliminary research. Molecules, 2020, 25, 2638

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Natural Medicines & Medicinal Plants (AREA)
  • Mycology (AREA)
  • Biotechnology (AREA)
  • Engineering & Computer Science (AREA)
  • Alternative & Traditional Medicine (AREA)
  • Botany (AREA)
  • Medical Informatics (AREA)
  • Microbiology (AREA)
  • Molecular Biology (AREA)
  • Medicines Containing Plant Substances (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

La présente invention concerne en particulier des compositions à base de cannabinoïdes et de champignons destinées à être utilisées en tant que thérapie d'appoint pour une thérapie classique pour le traitement d'un cancer du sein.
PCT/IL2023/050159 2022-02-23 2023-02-16 Compositions destinées à être utilisées en tant que thérapie d'appoint dans le traitement du cancer du sein WO2023161921A2 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263313031P 2022-02-23 2022-02-23
US63/313,031 2022-02-23

Publications (2)

Publication Number Publication Date
WO2023161921A2 true WO2023161921A2 (fr) 2023-08-31
WO2023161921A3 WO2023161921A3 (fr) 2023-11-02

Family

ID=87766793

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IL2023/050159 WO2023161921A2 (fr) 2022-02-23 2023-02-16 Compositions destinées à être utilisées en tant que thérapie d'appoint dans le traitement du cancer du sein

Country Status (1)

Country Link
WO (1) WO2023161921A2 (fr)

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6440420B1 (en) * 2001-03-19 2002-08-27 Xin Liu Method for extracting oleaginous substances from germination-activated Ganoderma lucidum spores
GB2478595B (en) * 2010-03-12 2018-04-04 Gw Pharma Ltd Phytocannabinoids in the treatment of glioma
WO2019131444A1 (fr) * 2017-12-26 2019-07-04 花王株式会社 Agent d'amélioration des fonctions cognitives
GB201806953D0 (en) * 2018-04-27 2018-06-13 Gw Res Ltd Cannabidiol Preparations
EP4185131A1 (fr) * 2020-07-21 2023-05-31 AI Pharmaceuticals Jamaica Limited Compositions et méthodes de traitement de cancers

Also Published As

Publication number Publication date
WO2023161921A3 (fr) 2023-11-02

Similar Documents

Publication Publication Date Title
Chugh et al. Fungal mushrooms: a natural compound with therapeutic applications
Sultana et al. Molecular targets for the management of cancer using Curcuma longa Linn. phytoconstituents: A Review
Karbasforooshan et al. Topical silymarin administration for prevention of acute radiodermatitis in breast cancer patients: A randomized, double‐blind, placebo‐controlled clinical trial
Li et al. Enhancement of cyclophosphamide-induced antitumor effect by a novel polysaccharide from Ganoderma atrum in sarcoma 180-bearing mice
Venugopal et al. Phytochemicals in diets for breast cancer prevention: The importance of resveratrol and ursolic acid
Shokrzadeh et al. Cytotoxicity of hydro-alcoholic extracts of Cucurbita pepo and Solanum nigrum on HepG2 and CT26 cancer cell lines
Rattanachaikunsopon et al. Bacteriostatic effect of flavonoids isolated from leaves of Psidium guajava on fish pathogens
Ramakrishna et al. Anticancer activities of plant secondary metabolites: Rice callus suspension culture as a new paradigm
US7553501B2 (en) Immune phyto-neutraceutical composition
CA3186850A1 (fr) Compositions et methodes de traitement de cancers
Lee et al. Induction apoptosis of erinacine A in human colorectal cancer cells involving the expression of TNFR, Fas, and Fas ligand via the JNK/p300/p50 signaling pathway with histone acetylation
El-Garawani et al. Candelariella vitellina extract triggers in vitro and in vivo cell death through induction of apoptosis: A novel anticancer agent
Bao et al. Astragalus polysaccharide RAP selectively attenuates paclitaxel-induced cytotoxicity toward RAW 264.7 cells by reversing cell cycle arrest and apoptosis
Kumar et al. Moringa oleifera L. leaf extract induces cell cycle arrest and mitochondrial apoptosis in Dalton's Lymphoma: An in vitro and in vivo study
Park et al. Anti-inflammatory effects of Cordyceps mycelium (Paecilomyces hepiali, CBG-CS-2) in Raw264. 7 murine macrophages
Zhang et al. Apoptosis induction activity of polysaccharide from Lentinus edodes in H22-bearing mice through ROS-mediated mitochondrial pathway and inhibition of tubulin polymerization
Zhao et al. Antidepressant-like effect of Ganoderma lucidum spore polysaccharide-peptide mediated by upregulation of prefrontal cortex brain-derived neurotrophic factor
Schanknecht et al. Phytochemical constituents and derivatives of Cannabis sativa; bridging the gap in melanoma treatment
Farid et al. Aloe vera gel as a stimulant for mesenchymal stem cells differentiation and a natural therapy for radiation induced liver damage
Lowenthal et al. The mycelium of the Trametes versicolor synn. Coriolus versicolor (Turkey tail mushroom) exhibit anti-melanoma activity in vitro
Amiri et al. Stimulators of immunogenic cell death for cancer therapy: focusing on natural compounds
WO2022024097A1 (fr) Compositions antivirales et leurs procédés d'utilisation
WO2022024096A1 (fr) Compositions et procédés pour favoriser la perte de poids et pour le traitement de l'obésité
Zhang et al. Compositions and anti-tumor activity of Pyropolyporus fomentarius petroleum ether fraction in vitro and in vivo
MEng et al. Anticancer activity of selected Colocasia gigantia fractions