Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
USE OF CANNABIDIOL IN THE TREATMENT OF TUBEROUS SCLEROSIS COMPLEX
Document Type and Number:
WIPO Patent Application WO/2018/234811
Kind Code:
A1
Abstract:
The present invention relates to the use of cannabidiol (CBD) for the treatment of tumours associated with Tuberous Sclerosis Complex (TSC). In particular the CBD was able to decrease the number and size of marker cells, pS6, in a zebrafish model of TSC. This is5 suggestive of a disease modifying effect whereby treatment with CBD could result in the reduction or prevention of the benign tumours that occur in TSC patients. Preferably the CBD used is in the form of a highly purified extract of cannabis such that the CBD is present at greater than 98% of the total extract (w/w) and the other components of the extract are characterised. In particular the cannabinoid tetrahydrocannabinol (THC) has been substantially10 removed, to a level of not more than 0.15% (w/w) and the propyl analogue of CBD, cannabidivarin, (CBDV) is present in amounts of up to 1%. Alternatively, the CBD may be a synthetically produced CBD. In use the CBD is given concomitantly with one or more other drugs used in the treatment of TSC. Such drugs may include rapamycin and / or everolimus.

Inventors:
WHALLEY BENJAMIN (GB)
HIND WILLIAM (GB)
GRAY ROYSTON (GB)
BAZELOT MICHAEL (GB)
DE SILVA SERRA INES (GB)
WILLIAMS CLAIRE (GB)
TEE ANDREW (GB)
Application Number:
PCT/GB2018/051733
Publication Date:
December 27, 2018
Filing Date:
June 21, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
GW RES LTD (GB)
International Classes:
A61K45/06; A61K31/352; A61K31/436; A61P35/00
Domestic Patent References:
WO2016191651A12016-12-01
WO2013168131A12013-11-14
WO2018002637A12018-01-04
WO2018002665A12018-01-04
WO2017204986A12017-11-30
Foreign References:
CN104490873A2015-04-08
Attorney, Agent or Firm:
HGF LIMITED (GB)
Download PDF:
Claims:
CLAIMS

1. Cannabidiol (CBD) for use in the treatment of tumours associated with Tuberous

Sclerosis Complex (TSC).

2. CBD for use according to claim 1 , wherein the tumours associated with TSC are benign tumours.

3. CBD for use according to any of the preceding claims, wherein the size of the tumours associated with TSC are reduced.

4. CBD for use according to any of the preceding claims, wherein the CBD is provided at a dose suitable to inhibit the formation of the tumours associated with TSC.

5. CBD for use according to claim 1 , wherein the CBD is used in combination with one or more concomitant drugs used in the treatment of TSC.

6. CBD for use according to claim 2, wherein the one or more concomitant drugs used in the treatment of TSC is an mTOR inhibitor.

7. CBD for use according to claim 3, wherein the mTOR inhibitor is rapamycin or

everolimus.

8. CBD for use according to any of the preceding claims, wherein the CBD is present as a highly purified extract of cannabis which comprises at least 95% (w/w) CBD.

9. CBD for use according to claim 5, wherein the extract comprises less than 0.15% THC.

10. CBD for use according to claim 5 or 6, wherein the extract further comprises up to 1 % CBDV.

11. CBD for use according to claim 1 , wherein the CBD is present as a synthetic compound.

12. CBD for use according to any of the preceding claims, wherein the dose of the one or more concomitant drugs used in the treatment of TSC that are used in combination with the CBD is reduced.

13. CBD for use according to any of the preceding claims, wherein the dose of CBD is greater than 5 mg/kg/day.

14. A method of treating a patient with Tuberous Sclerosis Complex (TSC) comprising administering cannabidiol (CBD) in a therapeutically effective amount to prevent or reduce tumours associated with TSC to the patient in need thereof.

Description:
USE OF CANNABIDIOL IN THE TREATMENT OF TUBEROUS SCLEROSIS COMPLEX

[0001] The present invention relates to the use of cannabidiol (CBD) for the treatment of tumours associated with Tuberous Sclerosis Complex (TSC). In particular the CBD was able to decrease the number and size of marker cells, pS6, in a zebrafish model of TSC. This is suggestive of a disease modifying effect whereby treatment with CBD could result in the reduction or prevention of the benign tumours that occur in TSC patients.

[0002] Preferably the CBD used is in the form of a highly purified extract of cannabis such that the CBD is present at greater than 98% of the total extract (w/w) and the other components of the extract are characterised. In particular the cannabinoid tetrahydrocannabinol (THC) has been substantially removed, to a level of not more than 0.15% (w/w) and the propyl analogue of CBD, cannabidivarin, (CBDV) is present in amounts of up to 1 %. Alternatively, the CBD may be a synthetically produced CBD.

[0003] In use the CBD is given concomitantly with one or more other drugs used in the treatment of TSC. Such drugs may include rapamycin and / or everolimus. Alternatively the CBD may be formulated for administration separately, sequentially or simultaneously with one or more drugs used in the treatment of TSC or the combination may be provided in a single dosage form. Where the CBD is formulated for administration separately, sequentially or simultaneously it may be provided as a kit or together with instructions to administer the one or more components in the manner indicated. It may also be used as the sole medication, i.e. as a monotherapy.

BACKGROUND TO THE INVENTION

[0004] Tuberous sclerosis complex (TSC) is an autosomal dominant neurocutaneous disorder characterized by the formation of hamartomas in multiple organ systems, most notably the brain, skin, kidneys, heart and eyes (Leung 2007). A hamartoma is a benign tumour composed of an overgrowth of mature cells and tissues normally found at the site that are grown in a disorganized mass. The benign tumours, also referred to as tubers, are descriptive of the potato-like nodules in the brain, which calcify with age and become sclerotic (Leung 2007).

[0005] The disease caused by a mutation in either the TSC1 , coding the protein hamartin, or the TSC2, coding tuberin, genes (Curatolo 2015). Hamartin and tuberin form a complex that, due to its GTPase-activating protein (GAP) domain, keeps Rheb bound to GDP. When a mutation in either of these proteins is present, the inhibitory function of this complex is impaired, allowing constitutive phosphorylation of mTOR (Laplante & Sabatini 2012; Dibble et al. 2012).

[0006] Symptoms of the disorder vary depending on which system and which organs are involved. The natural course of TSC varies from individual to individual, with symptoms ranging from very mild to quite severe. Tumours can grow in nearly any organ, but they most commonly occur in the brain, kidneys, heart, lungs, and skin.

[0007] When present, neurologic complications are the most common cause of mortality and morbidity and are the most likely to affect quality of life. Seizures and epilepsy are the most common forms of neurologic complication and occur in 75-90% of patients (Leung 2007). Sixty- three percent of patients experience seizure onset within the first year of life (Wang & Fallah 2014).

[0008] The most common types of seizures in TSC patients are infantile spasms, complex partial seizures, and generalized tonic clonic seizures (Leung 2007). Refractory epilepsy develops in 55-62.5% of patients (Wang & Fallah 2014).

[0009] Three types of brain lesions are seen in TSC: cortical tubers, for which the disease is named, generally form on the surface of the brain but may also appear in the deep areas of the brain; subependymal nodules (SEN), which form in the walls of the ventricles, the fluid-filled cavities of the brain; and subependymal giant-call astrocytomas (SEGA), which develop from SEN and grow such that they may block the flow of fluid within the brain, causing a build-up of fluid and pressure and leading to headaches and blurred vision.

[0010] Tumours called cardiac rhabdomyomas are often found in the hearts of infants and young children with TSC. If the tumours are large or there are multiple tumours, they can block circulation and cause death.

[0011] Benign tumours called phakomas are sometimes found in the eyes of individuals with TSC, appearing as white patches on the retina. Generally they do not cause vision loss or other vision problems, but they can be used to help diagnose the disease. Additional tumours and cysts may be found in other areas of the body, including the liver, lung, and pancreas. Bone cysts, rectal polyps, gum fibromas, and dental pits may also occur.

[0012] Patients with TSC are commonly treated with anti-seizure medications to control epilepsy; in addition tumours are treated with mTOR inhibitors.

[0013] Only one drug, an mTOR inhibitor (everolimus) has been FDA approved for the treatment of TSC in the 165 years the disease has been identified, and it is specifically indicated for the treatment of sub-ependymal giant cell astrocytoma associated with TSC, where treatment is required, and where surgery is not considered appropriate. The therapeutic dose of everolimus is 4.5mg/m 2 .

[0014] Everolimus has a high risk side effect profile and may not be suitable for use in all patients with TSC. As cited in the Prescribing Information, "clinical benefit such as improvement in disease-related symptoms has not been demonstrated" (Novartis 2015).

[0015] The side effect profile of everolimus includes non-infectious pneumonitis;

immunosuppression; increased risk of infections; angioedema; oral ulceration; renal failure; impaired wound healing; elevations of serum creatinine, urinary protein, blood glucose, and lipids; decreases in haemoglobin, neutrophils, and platelets.

[0016] The most common adverse reactions (incidence≥30%) include stomatitis, infections, rash, fatigue, diarrhoea, oedema, abdominal pain, nausea, fever, asthenia, cough, headache and decreased appetite (Novartis 2015).

[0017] Tuber growth is a hallmark of TSC presentation. In addition to mTOR inhibition, which may contribute to reduction in tuber size in patients with TSC, overexpression of hypoxia- inducible factor-1 (HIF1 a) and vascular endothelial growth factor (VEGF) are thought to be the cause of these tubers. These growth factors are involved in the angiogenesis pathway and correlate with the mutation of TSC1 and TSC2 as well as the extent of tuber growth in animal models of TSC. It is unknown if these actions are downstream of the modulation of mTOR by CBD, or by a distinct signalling pathway. Rapamycin or related inhibitors of mTOR may have therapeutic benefit in TSC both by direct tumour cell killing and by inhibiting the development of TSC lesions through impairment of VEGF production.

[0018] The compound cannabidiol (CBD) has recently been used in several treatment resistant paediatric epilepsy clinical trials, where significant reductions in seizure frequency were reported (Devinsky et al. 2014). Furthermore CBD has been shown to be beneficial in the treatment of refractory epilepsy in TSC patients having focal onset seizures (Geffrey et al., 2014).

[0019] Whilst the potential of cannabis and the cannabinoids, including CBD, to treat epilepsy has been rekindled, to date the ability of the compound to treat the tumours associated with TSC or indeed to provide a disease modifying effect has not been shown.

[0020] The applicant has found that CBD was able to reduce TSC tumour cells in both an in vitro TSC cell line and in vivo in a zebrafish model. Furthermore CBD has been shown to act in a synergistic manner with an mTOR inhibitor, rapamycin, in a tumour cell line.

[0021] Therefore CBD has been shown to treat the benign tumours associated with TSC. Such an ability to reduce or prevent the formation of these benign tumours results in a disease modifying effect. This effect comes about due to the reduction of the size or the prevention of formation of the benign tumours thereby prevents the co-morbid symptoms associated with TSC such as seizures which are caused by the benign tumours growing in the brain.

BRIEF SUM MARY OF THE DISCLOSURE

[0022] In accordance with a first aspect of the present invention there is provided cannabidiol (CBD) for use in the treatment of tumours associated with Tuberous Sclerosis Complex (TSC). [0023] Preferably the tumours associated with TSC are benign tumours. More preferably, the size of the tumours associated with TSC are reduced. Alternatively, the CBD is provided at a dose suitable to inhibit the formation of the tumours associated with TSC.

[0024] Preferably the CBD is used in combination with one or more concomitant drugs used in the treatment of TSC. More preferably the one or more concomitant drugs used in the treatment of TSC is an mTOR inhibitor. More preferably still the mTOR inhibitor is rapamycin or everolimus.

[0025] Preferably the CBD is present as a highly purified extract of cannabis which comprises at least 95% (w/w) CBD. Preferably the extract comprises less than 0.15% THC. More preferably the extract further comprises up to 1 % CBDV.

[0026] Alternatively the CBD is present as a synthetic compound.

[0027] Preferably the dose of the one or more concomitant drugs used in the treatment of

TSC that are used in combination with the CBD is reduced.

[0028] Preferably the dose of CBD is greater than 5 mg/kg/day.

[0029] In accordance with a second aspect of the present invention there is provided a method of treating a patient with Tuberous Sclerosis Complex (TSC) comprising administering cannabidiol (CBD) in a therapeutically effective amount to prevent or reduce tumours associated with TSC to the patient in need thereof. BRIEF DESCRIPTION OF THE DRAWINGS

[0030] Embodiments of the invention are further described hereinafter with reference to the accompanying drawings, in which:

[0031] Figure 1 shows the effect of CBD on wound healing in Tsc2-/-Mouse Embryonic Fibroblast (A) and AML cell lines (B);

[0032] Figure 2 shows the effect of CBD on migration and invasion in Tsc2-/-Mouse

Embryonic Fibroblast (A) and AML (B) cell lines;

[0033] Figure 3 shows the effect of CBD on tumour formation in Tsc2-/-Mouse Embryonic Fibroblast (A) and AML (B) cell lines;

[0034] Figure 4 shows the effect of CBD on the number of pS6 positive cells in a zebrafish model of TSC;

[0035] Figure 5 shows the effect of CBD on the size of pS6 cells in a zebrafish model of TSC; and

[0036] Figure 6 shows the effect of CBD on inhibition of growth in PANC1 (A) and

MIAPACA(B) cell lines.

DEFINITIONS

[0037] Definitions of some of the terms used to describe the invention are detailed in Table 1 below:

[0038] The cannabinoids described in the present application are listed below along with their standard abbreviations. Table 1. Cannabinoids and their abbreviations

[0039] The table above is not exhaustive and merely details the cannabinoids which are identified in the present application for reference. So far over 60 different cannabinoids have been identified and these cannabinoids can be split into different groups as follows:

Phytocannabinoids; Endocannabinoids and Synthetic cannabinoids (which may be novel cannabinoids or synthetically produced phytocannabinoids or endocannabinoids). [0040] "Phytocannabinoids" are cannabinoids that originate from nature and can be found in the cannabis plant. The phytocannabinoids can be isolated from plants to produce a highly purified extract or can be reproduced synthetically.

[0041] "Highly purified cannabinoids" are defined as cannabinoids that have been extracted from the cannabis plant and purified to the extent that other cannabinoids and non-cannabinoid components that are co-extracted with the cannabinoids have been removed, such that the highly purified cannabinoid is greater than or equal to 95% (w/w) pure.

[0042] "Synthetic cannabinoids" are compounds that have a cannabinoid or cannabinoid-like structure and are manufactured using chemical means rather than by the plant.

[0043] Phytocannabinoids can be obtained as either the neutral (decarboxylated form) or the carboxylic acid form depending on the method used to extract the cannabinoids. For example it is known that heating the carboxylic acid form will cause most of the carboxylic acid form to decarboxylate into the neutral form. DETAILED DESCRIPTION

[0044] The following examples provide evidence for the efficacy of CBD in the ability to treat tumours in TSC. Example 1 tests CBD in two different TSC cell lines: (i) Angiomyolipomas 621-101 cell-line (derived from a Tuberous Sclerosis Complex patient with a known loss of function of TSC2), and (ii) Tsc2-/- and Tsc2+/+ mouse embryonic fibroblast (MEF) cell lines.

[0045] Example 2 describes a TSC model in zebrafish (Kim et al. 2011). CNS development in zebrafish follows the same pattern as in other vertebrates, with both neurons and glial cells having been identified.

[0046] Lastly, Example 3 demonstrates the synergistic action of CBD with an mTOR

inhibitor, everolimus, in tumour cell lines.

EXAMPLE 1 : EFFICACY OF CAIMIMABIDIOL IN REDUCTION OF TUMOURS IN TWO TUBEROUS SCLEROSIS COMPLEX CELL LINES

Materials and Methods

Cell lines and maintenance

[0047] (i) Angiomyolipomas (AML) cells derived from a Tuberous Sclerosis Complex patient (621-101 cell-line), with a known loss of function of TSC2. To generate a rescue control cell line, these 621-101 cells were stably transfected with a TSC2-expressing plasmid possessing Zeocin™ resistance (pcDNA3.1zeo-hTSC2). Zeocin™ was purchased from LifeTechnologies (cat no: R25001) and was used to originally generate the stable cell line (by Prof. Lisa Henske) over two weeks of Zeocin™ selection and then for continued maintenance of the cells in tissue culture (at 100μg/ml). [0048] (ii) Tsc2-/- p53-/- and Tsc2+/+ p53-/- mouse embryonic fibroblast (MEF) cell lines (now referred to as Tsc2-/- and Tsc2+/+, respectively).

Cell culture

[0049] Tsc2-/- and Tsc2+/+ cell lines were cultured and maintained in DMEM supplemented with 10% (v/v) FBS and 1 % (v/v) penicillin-streptomycin in a humidified incubator (5% CO2 at 37°C). The AML -/- cell lines were cultured and maintained in DMEM supplemented with 15% (v/v) FBS and 1 % (v/v) penicillin-streptomycin in a humidified incubator (5% CO2 at 37oC). For hypoxia, cells were put into a Binder CB150 hypoxic chamber set at 1 % O2 for the indicated time points

Drug treatments

[0050] Pure CBD was used in this example. Rapamycin was obtained from MerkMillipore.

[0051] The stock concentration of CBD was made to 20 mM in dimethyl sulfoxide (DMSO). CBD was added to culture media to final concentrations of either 5 μΜ or 10 μΜ, as indicated). The final concentration of the DMSO in the cell culture medium was at a maximum level of 0.05% (v/v).

Wound Healing Assays

[0052] Cells were seeded in 60 mm plates and left to reach 100% confluency. Cells were then synchronised in 1 % (v/v) FBS DMEM for 24 h and "wounded" with a pipette tip. Dead cells were removed with PBS wash and then subsequently replaced with DMEM (10% (v/v) FBS). Cells were pre-treated for 30 min with either rapamycin, c-MET or STAT3 inhibitors (where indicated) before cytokine stimulation. Pictures were taken before treatment and 12-18 h after treatment using an inverted AMG EVOS microscope equipped with an Olympus camera.

[0053]

Migration and invasion assays

[0054] Transwell permeable supports with 6.5 mm diameter inserts, 8.0 μηι pore size, and a polycarbonate membrane were used to perform migration assays. Cells were grown in a 75- cm2 flask with standard medium (10% (v/v) FBS) until confluent. Cells were then harvested using Trypsin-EDTA. Cells were counted using a haemocytometer. A total of 1x10 6 cells and were resuspended in DMEM containing 1 % (v/v) FBS. These cells were then seeded in the upper chamber of the Transwell; the lower chamber was filled with 600 mL of standard culture medium (10% (v/v) FBS) and 5 mg/mL fibronectin, as an adhesive substrate. Cells were incubated at 37°C 5% CO2 for 24 hours. The percentage of adherent cells was then determined by fixing the cells with methanol and acetone (1 : 1) for 20 minutes at 20°C. Cells were then stained with Crystal Violet (5 mg/mL) in ethanol for 10 minutes, followed by a stringent wash with dH20 until the water ran clear. Crystal Violet stained cells were eluted with 1 % (w/v) SDS and the absorbance was read at 550 nm on a Genova MK3 Lifescience. [0055] For invasion assays, a similar protocol was used; however, the top chamber of the Transwell was filled with 300 ml_ of BD Matrigel Basement Membrane Matrix (1 mg/mL). The Matrigel was incubated at 37oC for 4 hours to allow it to gel. Cells were then seeded and incubated as described for migration assay for 3 days. The number of invaded cells was determined by fixation staining and elution of crystal violet with 1 % (w/v) SDS, as before.

Tumour spheroid growth assays

[0056] Two-layered soft agar assays were carried out in 6-well plates. All cell lines were plated in complete DMEM media in 0.35% (v/v) agar at (3 *10 5 ) over a 0.6% (v/v) agar layer. The agar was then overlaid with complete DMEM media and spheroids were grown for 14 days at 37°C in 5% CO2. Media were changed twice a week and new drug was added to media. Pictures were taken using an inverted AMG EVOS microscope equipped with an Olympus camera. Volume of tumour spheroids was measured using ImageJ software.

Lysing and western Blot

[0057] Cells were washed in Phosphate Buffered Saline (PBS) and then lysed directly in sample buffer (62.5 mM Tris HCL, 50 mM DTT, 2 % SDS (w/v), 10 % Glycerol (w/v), 0.1 % Bromophenol blue (w/v) pH 7.6) and sonicated for 5 x 20 s cycles on full power (30 amplitude microns). Samples were boiled at 95 °C for 10 min. Lysates were resolved by SDS-PAGE and proteins were transferred to polyvinylidene difluoride membranes.

Membranes were blocked in 5 % (w/v) dry milk powder dissolved in Tris-buffered saline containing 0.1 % (v/v) Tween, then probed with primary antibody and horse radish peroxidase- conjugated secondary antibody. Proteins were visualized using Enhanced Chemiluminescent solution and Hyperfilm. Antibodies towards ribosomal protein S6 (rpS6), phospho-rpS6

(Ser235/236) were purchased from Cell Signaling Technology. Anti-HIF-1a was obtained from BD transduction laboratories. Statistical Analysis

[0058] Experiments were carried out at least 3 times (unless otherwise indicated). Where applicable, results are expressed as mean ± standard deviation (SD). Student's t-test and oneway ANOVA with Bonferroni's post hoc test were used and significance reported at p≤ 0.05. Data shown in the figures represent **p<0.01 , ***p<0.001. Results

Wound healing assay

[0059] Wound closure is an indication of both cell migration as well as proliferation of the leading edge of the wound, and a decrease of this is considered beneficial in this setting as it indicates a reduction of potential tumour growth. [0060] Figure 1 (A) demonstrates that CBD at 10 and 20 μΜ was as effective as rapamycin to block wound closer in the Tsc2-/- Mouse Embryonic Fibroblast (MEF) cell lines. Tsc2-/- MEFs have a higher capacity to close the wound than the Tsc2+/+ control cells.

[0061] Figure 1 (B) demonstrates the data for the wound closure assays in the second cell line. CBD was effective at reducing wound closure with 5, 10 and 20 μΜ CBD. 20 μΜ CBD was as significant as rapamycin in reducing wound closure by 60%.

[0062] When CBD and rapamycin were given in combination there combined effect was similar to that of either the compounds CBD or rapamycin. Cell migration and invasion assay

[0063] Cell migration and invasion assays were carried out in the Tsc2-/- cells, using Tsc2+/+ as a control cell. There was a non-significant increase in the number of migrated cells in Tsc2-/- cells, and whilst CBD did not cause a significant reduction in these cells, it did reduce cell migration to control level as is demonstrated in Figure 2 (A).

[0064] At concentrations of 5 μΜ and 10 μΜ, CBD had a marked effect at blocking cell invasion in the Tsc2-/- MEFs, with 5 μΜ CBD restoring invasion to the level of the Tsc2+/+ control cells, and 10 μΜ CBD having a more robust effect, as is shown in Figure 2 (B).

[0065] In the AML 621-101 cells, CBD at both 5 μΜ and 10 μΜ was sufficient to

significantly block both cell migration and invasion. Indeed, 10 μΜ CBD reduced cell migration to a significantly greater extent than rapamycin.

Tumour formation assay

[0066] Tumour formation assays were carried out in soft agar in the Tsc2-/- MEFs.

Significance to inhibit tumour spheroid diameter was observed with 10 μΜ CBD, but not 5 μΜ, and with rapamycin. Similar observations were found in the AML 621-101 cells, where 10 μΜ was also sufficient to block tumour spheroid formation). Figure 3 detail these data.

Western blot analysis

[0067] Western blotting was carried out to determine whether CBD could impair mTORCI signalling (using rpS6 phosphorylation as a readout of mTORCI signalling). CBD at 5 μΜ inhibited rpS6 phosphorylation after 6 h of treatment in the Tsc2-/- MEFs.

[0068] Under hypoxia HIF-1a protein is stabilised and ensures an angiogenic response and is necessary for tumour formation, metabolic transformation and malignancy. Of interest, CBD at 10 μΜ was sufficient to block hypoxia induced expression of HIF-1 a protein in both Tsc2-/- MEFs and AML 621-101.

Conclusions [0069] CBD can block tumour formation, cell migration and cell invasion in two cell models of TSC. CBD also blocks mTORCI signalling after 6 h of treatment, and is a potent repressor of HIF-1 a, which indicates that CBD could have potential as an anti-angiogenic agent.

EXAMPLE 2: EFFICACY OF CANNABIDIOL IN REDUCTION OF TUMOURS A ZEBRAFISH MODEL OF TUBEROUS SCLEROSIS COMPLEX

Materials and Methods

Zebrafish Husbandry

[0070] A zebrafish model of TSC with a tsc2 gene deletion was used. This model has been previously published and validated (Kim et al. 201 1). By mating the gene deletion animals with wild-type tsc2+/+ animals, heterozygote (tsc2+/-) zebrafish were also obtained and tested in this example. Imaging

[0071] For both pS6 and TUNEL staining, non-consecutive sections were imaged using a Zeiss Axiolmager microscope. Exposure time was kept constant during image acquisition and

determined by the observation of a slide where primary antibody or enzyme solution was

omitted. Pictures were taken with a 20 X objective, using the AxioVision software, and coloured using Fiji ImageJ. All counting and measuring of cells was done in the original black and white pictures.

Immunohistochemistry

[0072] For TUNEL staining (1 : 10; Roche, Sigma, UK), 10 μηι sections were cut and collected onto microscope slides, and stored at -80°C until used. Sections were incubated for 2 hours, at room temperature, in a 2% BSA, 10% horse serum and 0.05% TX-100 buffer. After the final rinsing step, sections were incubated with TUNEL solution, in the dark, for 1 hour, at 37°C.

[0073] Negative control was performed by omitting the enzyme solution, while positive control was performed by previous incubation of sections with 5 mg/mL DNAse for 10 minutes, at 37°C.

[0074] For TUNEL assay, qualitative observation was done solely on one animal per genotype and per group, due to the low presence of labelled cells.

Statistical Analysis

[0075] Statistical analysis was performed in SPSS (IBM SPSS Statistics 22), except for the chi- square test, which was done using GraphPad Prism 5. Normality and sphericity were tested using the Kolmogorov-Smirnov and Mauchly's tests, respectively. Repeated measures two-way ANOVA tests were used to analyse the locomotor assay data and cell number, while a three- way ANOVA test was used to analyse cell size. Touch response (TR) between genotypes and treatments was analysed by chi-square test. Tests were followed by Tukey or Bonferroni post- hoc tests.

[0076] Data are expressed as mean ±SEM unless stated otherwise, and significant values were considered when p< 05. All graphs were prepared with GraphPad Prism 5.

Materials

[0077] Pure CBD was used in this example. Danieau's solution was used as the embryo medium for the zebrafish.

Results

Cell Number:

[0078] CBD was able to decrease the number of pS6 positive cells as is shown in Figure 4. To explore mTOR pathway activation in the different genotypes and treatments,

immunohistochemistry was performed using a pS6 (Ser235/236) antibody.

[0079] Upon initial microscope observation, a stronger immunoreactivity was detected in tsc2-/- brain tissue compared to tsc2+/+ and tsc2+/- zebrafish sections. DMSO sections also showed increased reactivity when compared to the Danieau's group, while sections from CBD incubated animals showed a marked decrease in the number of pS6 positive cells.

[0080] To better understand the apparent differences between groups, pS6 positive cells were counted for each genotype and treatment. A significant main effect of genotype was found, with tsc2-/- zebrafish sections showing more pS6 positive cells in the brain than tsc2+/+ and tsc2+/- (+173.3%) (Figure 4A).

[0081] A significant main effect of treatment was also found, indicating that CBD reduced the average number of positive cells compared to both Danieau's (-63.2%) and DMSO (-77.1 %) groups. Additionally, DMSO incubated slides had a significant increase in the average number of positive cells (+61.2%) compared to Danieau's (Figure 4B).

[0082] A significant interaction between genotype and treatment was also found, revealing that, while CBD had no significant effect on the average number of pS6 positive cells in tsc2+/+ nor tsc2+/- Danieau's incubated zebrafish, it did have a significant effect on tsc2-/- zebrafish (- 55.4% for the Danieau's and -58.9% for the DMSO group) (Figure 4C).

Cell Size

[0083] CBD decreases the area of pS6 positive cells as is shown in Figure 5. [0084] A significant main effect of genotype was found, indicating that tsc2-/- zebrafish had significantly larger pS6 positive brain cells than tsc2+/+ and tsc2+/- (+29%) zebrafish (Figure 5A).

[0085] Additionally, a significant main effect of treatment was also present, revealing that CBD incubated zebrafish had smaller pS6 positive cells than the ones present in the Danieau's or DMSO groups (-20.1 %) (Figure 5B).

[0086] Finally, an interaction between genotype and treatment was found. While Danieau's incubated zebrafish had progressively bigger pS6 positive cells, according to mutation severity, DMSO incubated animals had increased pS6 positive cell size in tsc2+/+ (+18.1 %) but not tsc2+/- nor tsc2-/- cells, compared to the Danieau's group.

[0087] As for CBD, incubation did not affect cell size in tsc2+/+ zebrafish, but it did significantly decrease the size of pS6 positive cells in tsc2+/- and tsc2-/- zebrafish (-27.6% and -23% compared to Danieau's, and -18.1 % and -20.8%, compared to the DMSO groups, respectively) (Figure 5C).

Conclusions

[0088] The impact of CBD treatment upon the mTOR pathway was assessed by conducting immunohistochemistry against pS6, which is a commonly used marker for mTOR activity. When mTOR is active, pS6 has been shown to increase, in in vitro and in vivo models, as well as in human tissue (Roux et al. 2007).

[0089] Zebrafish with a tsc2-/- mutation exhibit more pS6 positive cells in the brain, compared to tsc2+/+ and tsc2+/- groups. The number of pS6 positive cells was not significantly different between tsc2+/+ and tsc2+/- animals.

[0090] Unexpectedly, it was found that DMSO increased the number of pS6 positive cells in the brain. However, although CBD was dissolved in this same vehicle, we saw a marked reduction in the number of pS6 positive cells in tsc2-/- zebrafish brain, compared to the other two genotypes.

[0091] Measurement of pS6 positive cells revealed that tsc2-/- cells had a larger area compared to tsc2+/+ and tsc2+/- positive cells, while these two groups did not statistically differ from each other.

[0092] When pS6 positive cell area was analysed by treatment, a significant main effect of CBD, but not of DMSO, on size was seen, revealing that CBD treated larvae had smaller cells, compared to Danieau's and DMSO treated groups.

[0093] Additionally, while the size of tsc2+/+ cells was not affected by CBD, as we had previously seen for the number of positive cells, this time, CBD also reduced the size of tsc2+/- cells, suggesting that the effect of CBD on size is mutation dependent. [0094] In conclusion, CBD produced a positive effect in a zebrafish model of TSC, reducing both cell size and cell number of pS6 cells. This is suggestive of a disease modifying effect whereby treatment with CBD can reduce the benign tumours that occur in virtually all TSC patients.

EXAMPLE 3: EFFICACY OF CANNABIDIOL IN COMBINATION WITH AN mTOR INHIBITOR IN A TUMOUR CELL LINE

Materials and Methods

Cell Lines

[0095] PANC-1 is a human pancreatic carcinoma, epithelial-like cell line; PANC-1 cells are used as an in vitro model of non-endocrine pancreatic cancer for tumorigenicity studies. The cells possess the type B phenotype for glucose-6-phosphate dehydrogenase G6PD and

overexpress heregulin/human epidermal growth-factor receptor 2 (HER2/neu) oncogene.

[0096] MIAPACA is a homo sapiens pancreas carcinoma. This is a hypotriploid human cell line.

Materials

[0097] Everolimus (RAD001 , Afinitor, Novartis), is a rapamycin analogue. It is an oral

mammalian target of rapamycin (mTOR) inhibitor and belongs to the PI3K related family of protein kinases and is activated by phosphorylation at serine 2448 (S2448).

[0098] Pure CBD was used in this example.

Cell Viability Assay

[0099] Cell viability was analyzed by the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl

tetrazolium bromide] assay. Pancreatic cancer cells were seeded onto each well of a 96-well plate. After 72h of treatment with CBD, MTT solution 5mg/ml in PBS was added to each well.

The plates were then incubated at 37°C for an additional 4 h to allow MTT to form formazan crystals by reacting with metabolically active cells. The formazan crystals were solubilized in a 1 N isopropanol/HCI 10% solution at 37°C, on a shaking table for 20 min. The absorbance values of the solution in each well were measured at 570 nm using a microplate reader. Cell viability was determined by the formula: cell viability (%) = (absorbance of the treated wells - absorbance of the blank control wells) / (absorbance of the negative control wells - absorbance of the blank control wells) x 100%. All MTT experiments were performed in triplicate and

repeated at least three times.

Drug Combination Assay [00100] For the study of the synergism between RadOOl and CBD on pancreatic cell lines Miapaca was tested, the cells were seeded in 96-multiwell plates at the density of 3,8 10 3 cells/well. After 24 hr incubation at 37°C, the cells were treated with different concentrations of RadOOl and CBD. Different molar ratios between two drugs in different sequences of administration were tested, (RadOOl for 48 h and CBD for 72h or RadOOl for 72h and CBD for 48h and in co-administration for 72 h.

[00101] The proliferative response was estimated by colorimetric 3-(4,5 di-methylthiazol-2- yl)-2,5-diphenyltetrazolium bromide (MTT) test. MTT conversion to formazan by metabolically viable cells was monitored by spectrophotometer at an optical density of 570 nm. Each data point represents the average of three separate experiments with each experiment containing four wells. Drug combination studies were based on concentration-effect curves generated as a plot of the fraction of unaffected (surviving) cells versus drug concentration after treatment.

[00102] To explore the relative contribution of each agent to the synergism equi-active doses of Rad001/Chloroquine were tested (IC50). Assessment of synergy was performed quantitating drug interaction by Calcusyn computer program (Biosoft, Ferguson, MO).

Combination index (CI) values of <1 , 1 , and >1 indicate synergy, additivity and antagonism, respectively.

[00103] Dose reduction index (DRI) representing the measure of how much the dose of each drug in a combination may be reduced at a given effect level compared with the doses of each drug alone.

Potentiation Factor (PF)

[00104] The specific contribution of CBD and RadOOl on the cytotoxic effect of the combination of drugs was analysed by calculating the potentiation factor (PF) in both cell lines, defined as the ratio of the IC50 of either CBD, RadOOl alone to the IC50 of CBD and RadOOl in combination; a higher PF indicates a greater cytotoxicity

Results

Cell Viability Assay

[00105] The growth inhibition induced by different concentrations (0,04-50 μΜ) of CBD on human Panc-1 at 72 h from the beginning of the treatment was evaluated with MTT assay.

[00106] In Panc-1 cells, CBD reached a 41 % growth inhibition, at the concentration of 50 μΜ as is shown in Figure 6A.

[00107] In Miapaca cells 50% growth inhibition was reached with CBD at the concentration of 15 μΜ as is shown in Figure 6B. Drug Combination Assay

[00108] The effects of the pharmacological combination between CBD and RAD001 on the proliferation MIAPACA cell lines, using MTT assay were studied. The data obtained were processed with dedicated program, CalcuSyn (Chou and Talalay, Biosoft, Oregon, USA), which measures synergism.

[00109] The combination of RadOOl and CBD produced a strong synergism when the drugs were administered in combination for 72 h as shown in Table 2 below.

Table 2. Synergy data for the combination of CBD with RadOOl

Potentiation Factor (PF)

[00110] The specific contribution of CBD and RadOOl on the cytotoxic effect of the combination of drugs was analyzed by calculating the potentiation factor (PF) defined as the ratio of the IC50 of either CBD or RadOOl alone to the IC50 of CBD and RadOOl in

combination. A higher PF indicates a greater cytotoxicity.

[00111] When CBD was tested in combination with RadOOl , the PF for CBD was 8 and for RadOOl the PF was 4, suggesting that CBD contributed a higher degree of cytotoxicity than the mTOR inhibitor RadOOl

Conclusions

[00112] The data shown in this example demonstrates the ability of CBD to work in synergy with the mTOR inhibitor RadOOl (everolimus) in decreasing the viability of tumour cells. Such a combination may be beneficial in the treatment of TSC where mTOR inhibitors are commonly used medications.