WO2001058445A1 - Therapy with cannabinoids in the treatment of cerebral tumor - Google Patents

Therapy with cannabinoids in the treatment of cerebral tumor Download PDF

Info

Publication number
WO2001058445A1
WO2001058445A1 PCT/ES2000/000450 ES0000450W WO0158445A1 WO 2001058445 A1 WO2001058445 A1 WO 2001058445A1 ES 0000450 W ES0000450 W ES 0000450W WO 0158445 A1 WO0158445 A1 WO 0158445A1
Authority
WO
WIPO (PCT)
Prior art keywords
cannabinoids
cannabinoid
tumors
malignant
natural
Prior art date
Application number
PCT/ES2000/000450
Other languages
Spanish (es)
French (fr)
Inventor
Manuel Guzman Pastor
Cristina Sanchez Garcia
Ismael Galve Roperh
Original Assignee
Universidad Complutense De Madrid
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 Universidad Complutense De Madrid filed Critical Universidad Complutense De Madrid
Priority to EP00976087A priority Critical patent/EP1177790B1/en
Priority to AT00976087T priority patent/ATE295162T1/en
Priority to AU13979/01A priority patent/AU1397901A/en
Priority to DE60020111T priority patent/DE60020111T2/en
Priority to ES00976087T priority patent/ES2241670T3/en
Publication of WO2001058445A1 publication Critical patent/WO2001058445A1/en

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
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • 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
    • A61K31/05Phenols
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/473Quinolines; Isoquinolines ortho- or peri-condensed with carbocyclic ring systems, e.g. acridines, phenanthridines
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53831,4-Oxazines, e.g. morpholine ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • the present invention falls within the field of brain tumor therapy.
  • the present invention as set forth in this specification refers to the therapeutic use of cannabinoids for the treatment of brain tumors.
  • the therapies used today for the treatment of these tumors usually be ineffective or at most palliative
  • the invention involves a technically simple approach, lacking noticeable side effects and highly effective for the treatment of brain tumors including the most malignant (glioblastomas)
  • glioblastomas are the most frequent (1 per 50 000 people and year) malignant (mortality close to 100%) and of more rapid evolution (life expectancy of weeks / months after diagnosis )
  • the treatment of glioblastomas is usually ineffective or merely palliative, and involves techniques such as surgery, radiotherapy, chemotherapy and immunotherapy (Louis, DN & Gusella, JF, Trenas Genet.
  • C sativa cannabinoids (Fig 1) carry ac Above its effects because they are similar to certain molecules produced by animals (including humans) that probably play important roles in the nervous system.
  • Cannabinoids both natural and synthetic, act by binding to specific membrane receptors (cannabinoid receptors or CB type), of which two different subtypes CB ⁇ and CB 2 are known today (Pertwee, op cit, Howlett, A er al, in The lUPHAR Compendium of Receptor Characte ⁇ zation and Classification, eds Godframd, T, Humphrey , P Ruffolo, R & Vanhoutte, P, lUPHAR Media, pp 97-104, 1998) Not all The body's tissues possess these receptors are located mainly in the nervous system, and that is why cannabinoids may exert their effects on the brain (Pertwee, op at, Childers, SR & Breivo
  • cannabinoids inhibit the proliferation of MCF-7 breast tumor cells (De Petrocellis, L et al. , Proc Nati Acad. Sci.
  • the present invention makes use of cannabinoids for the treatment of brain tumors for the first time, and is based on our original observations that cannabinoids induce a marked regression (which leads to a lengthening of life) and even eradication (which entails Cure) of ghoblastomas in laboratory animals
  • This invention involves the use of a technically simple therapy, devoid of noticeable side effects and, more importantly, very effective for the treatment of brain tumors, which as mentioned above cannot be treated satisfactorily today with other techniques or compounds
  • the experiments that have led to the present invention are detailed below.
  • C6 glioblastoma cells in the rat brain are widely used as an experimental model of malignant brain tumor (Barth, RF, J Neur ⁇ ncol 36, 91-102, 1998) C6 glioblastoma cells were inoculated directly into the brain of Wistar rats, and tumors were visualized by magnetic resonance imaging All animals that were left untreated died uniformly 12-18 days after cell inoculation (Fig 3a).
  • a group of animals was administered for 7 days THC or WIN-55,212-2 through a cannula located at the site of inoculation.
  • C6 glioblastoma cells were inoculated subcutaneously in mice deficient in RAG-2 recombinase (RAG-2 ' 1 ' ), lacking mature T and B lymphocytes (Shinkai, Y et al, Ce // 68, 855-867 1992)
  • RAG-2 ' 1 ' RAG-2 recombinase
  • Fig 4b the tumor size was extraordinarily smaller in animals treated with THC or WIN-55,212-2 than in control animals
  • Examples of tumor-bearing mice and dissected tumors after treatment with or without cannabinoids for 7 days are shown in Fig 4b
  • cannabinoids did not induce any significant alteration of behavioral parameters such as motor coordination and physical activity. Water and food intake, as well as weight gain, were also not affected by cannabinoids.
  • biochemical parameters glucose and food intake, as well as weight gain, were also not affected by cannabinoids.
  • the biochemical parameters glucose, urea, uric acid, creatinine, cholesterol, bilirubin
  • the markers of tissue damage alanine and aspartate ammotransferases, ⁇ -glutamyltransferase, creatine kinase, lactate dehydrogenase
  • cannabinoids are not only not toxic compounds for nerve cells, but even protect them of toxic stimuli such as glutamatergic agonists (Skaper, SD et al, Pr oc Nati Acad S ⁇ USA 93, 3984-3989, 1996, Shen, M & Thayer,
  • WIN-55,212-2, CP-55,940 and HU-210 induced the death of these cells at doses lower than THC, as expected from its higher affinity for cannabinoid receptors (Pertwee, op at)
  • SR141716 a selective CB antagonist nor SR144528 (a selective CB 2 antagonist)
  • Shire, D et al, Life Sci 65, 627-635, 1999 were able separately to prevent THC-induced cell death.
  • mice have been carried out with glioblastomas as a brain tumor model.
  • the invention can be applied to the treatment of other tumors.
  • brain diseases for example medulloepitheliomas, medulloblastomas, neuroblastomas, germinomas, embryonic carcinomas, astrocytomas, astroblastomas, ependymomas, oligodendrog omas, plexal carcinomas, neuroepitehomas, pineoblastomas, ependyloblastomas, neuroectodermal tumors, malignant melanomas, malignant melanomas
  • the experiments that have led to the present invention have been carried out with two paradigmatic cannabinoids, one natural (THC) and one synthetic (WIN-55,212-2)
  • THC natural
  • WIN-55,212-2 synthetic
  • the cannabinoid with more potent antiproliferative effect will be used for a given
  • C sativa for example ⁇ 8 -tetrah ⁇ drocannab ⁇ nol, cannabinol, cannabidiol
  • Fig 1 synthetic cannabinoids (for example HU-210, CP-55,940, CP-50,556)
  • Fig 2 Pertwee, op ⁇ t, F Barth, op cit)
  • medications that contain any cannabinoid in their composition
  • the experiments that have led to the present invention have been carried out by intratumorally administering the cannabinoid.
  • this will be the chosen route of administration, since it allows a high accessibility of the cannabinoid to the tumor.
  • the route of administration can also be systemic, for example intrape ⁇ toneal, intravenous or oral
  • the experiments that have led to the present invention have been carried out by the continuous administration of a dose of cannabinoid for a certain time.
  • these parameters will be optimized depending on the specific requirements of the treatment patient condition, size and tumor location, number of tumors, etc.
  • the mode of application may be continuous (preferred mode) or sequential in one or more dose per day This will obviously vary the doses of compound administered and the total duration of treatment.
  • Cannabinoid administration to rats began 12 days after cell inoculation.
  • the average tumor size was 70 mm 3 (range 25-100 mm 3 ) estimated by magnetic resonance imaging (Izquierdo et al, op cit, Cortés, by Felipe, Martin Hughes & Izquierdo, op ⁇ t)
  • the cannabinoids were administered by a cannula located at the site of inoculation of the tumor and fixed to the skull with dental cement, a small stainless steel thyme secured the cannula and cement Dental
  • the cannula was connected subcutaneously through a catheter to an osmotic mini-pump (Alzet 2001) that operated at a flow of 1 ⁇ l / h for 7 days
  • the osmotic pump was filled with 500-2500 ⁇ g THC or 50-250 ⁇ g WIN-55,212-2 in 200 ⁇ l of PBS supplemented with 5 mg / ml of ordered and dialyze
  • Fig 3a As seen in Fig 3a, all animals that were left untreated died uniformly 12-18 days after cell inoculation. Animals treated with cannabinoids had a significantly longer life than control animals. Moreover, cannabinoids eradicated completely the tumor in a significant percentage of animals
  • Fig 3b an MRI image of one of the animals cured with THC is shown, after the administration of the cannabinoid the tumor mass disappears completely, and a residual hypointense zone is observed that is interpreted as a fibrous scar at the site of inoculation No recurrence was observed in animals cured with cannabinoids
  • Tumors were induced in RAG-2 ' " mice by subcutaneous inoculation of 5x10 6 cells of C6 ghoblastoma in 100 ⁇ l of PBS supplemented with 0 1% glucose About 10 days later, when the average volume of the tumors was 250 mm 3 (range 200-300 mm 3 ), the animals were randomly divided into 3 groups and injected for 7 days vehicle,
  • the tumor size was extraordinarily smaller in animals treated with THC or WIN-
  • mice bearing tumors and dissected tumors after treatment with or without cannabinoids for 7 days are shown in Fig 4b
  • THC was capable of inducing the death of C6 glioblastoma cells also when added simultaneously SR141716 medium (selective antagonist of CBi) and SR144528 ( a selective antagonist of CB 2) prevented THC-induced cell death
  • Cannabmoids (2500 ⁇ g THC or 250 ⁇ g WIN-55,212-2) were administered to rats without tumors for 7 days as described above. Rats were then sacrificed and their brains fixed. with 4% paraformaldehyde in PBS Death by apoptosis was determined in 40 ⁇ m thick sections of the brain using a "TUNNEL staining kit" according to the supplier's instructions (Boeh ⁇ nger, Mannheim, Germany) DNA stranding with Deoxyundin triphosphate labeled with fluorescein was visualized by a confocal microscope (excitation wavelength 488 nm, length of 525 nm emission wave) The laser intensity and photodetector sensitivity were kept constant to allow comparison between treatments At least 5 optical fields were analyzed per animal
  • TUNNEL stains were performed in the subvent ⁇ cular area of the rat brain, which continues to proliferate in the adult animal.
  • the administration of cannabinoids not only did not produce any significant apoptotic effect on the living brain, but also the slight dizziness observed in the caudate putamen of control animals was not observed in animals treated with cannabinoids

Abstract

The therapy with cannabinoids in the treatment of cerebral tumors involves (intracranial or systematic) administration of (natural or synthetic) cannabinoids to (human or non-human) mammals having cerebral tumors. Activation of the specific receptors of the cannabinoids leads to selective death of the transformed cells. Regression or eradication of the cerebral tumors is achieved without any significant side-effects.

Description

TítuloTitle
TERAPIA CON CANNABINOIDES PARA ELTRATAMIENTO DE TUMORES CEREBRALESCANNABINOID THERAPY FOR THE TREATMENT OF BRAIN TUMORS
Campo técnico de la invenciónTechnical Field of the Invention
La presente invención se encuadra en el campo de la terapia de tumores cerebralesThe present invention falls within the field of brain tumor therapy.
Objeto de la invenciónObject of the invention
La presente invención según se recoge en esta memoria descriptiva se refiere a la utilización terapéutica de los cannabinoides para el tratamiento de tumores cerebrales Las terapias utilizadas hoy en día para el tratamiento de estos tumores (cirugía, radioterapia, quimioterapia, inmunoterapia, terapia génica) suelen ser ineficaces o a lo sumo paliativas La invención supone una aproximación técnicamente simple, carente de efectos colaterales notorios y altamente eficaz para el tratamiento de tumores cerebrales incluidos los más malignos (glioblastomas)The present invention as set forth in this specification refers to the therapeutic use of cannabinoids for the treatment of brain tumors. The therapies used today for the treatment of these tumors (surgery, radiotherapy, chemotherapy, immunotherapy, gene therapy) usually be ineffective or at most palliative The invention involves a technically simple approach, lacking noticeable side effects and highly effective for the treatment of brain tumors including the most malignant (glioblastomas)
AntecedentesBackground
Dentro de los tumores cerebrales que afectan a los seres humanos los glioblastomas son los más frecuentes (1 por 50 000 personas y año) malignos (mortalidad cercana al 100%) y de evolución más rápida (esperanza de vida de semanas/meses tras su diagnóstico) Hoy en día, el tratamiento de los glioblastomas suele ser ineficaz o meramente paliativo, e implica técnicas tales como cirugía, radioterapia, quimioterapia e inmunoterapia (Louis, D N & Gusella, J F , Trenas Genet. 11, 412-415 1995 Avgeropoulos, N.G & Batchelor, T T , Oncologist 4, 209-224, 1999) Además, la terapia génica ha comenzado a ser empleada como tratamiento experimental de los glioblastomas, aunque hasta ahora ha proporcionado escasos resultados positivos (Martuza, R L , Nature Med 3, 1323, 1997) El ya de por sí poco probable éxito de estas aproximaciones terapéuticas suele además verse complicado por factores como el rápido crecimiento, la notoria heterogeneidad, el alto nivel de infiltración y la extrema resistencia a quimioterapia que exhiben los glioblastomas (Maintz, D et al , J Neuropathol Exp Neurol 56, 1098-1 104, 1997, Masón, W , Louis, D N & Cairncross, J G , J Clin Oncol 15, 3423-3426, 1997, Martuza, op cit , Avgeropoulos & Batchelor, op cit ) Parece claro por tanto que es altamente recomendable el desarrollo de terapias alternativas para el tratamiento de tumores cerebrales Los cannabinoides son compuestos que deben su nombre a que son sintetizados por la planta Cannabis sativa L Estos compuestos, entre los que el Δ9-tetrahιdrocannabιnol (THC) destaca por sus elevadas potencia y abundancia, son los responsables de los efectos centrales y periféricos del consumo de marihuana (Pertwee, R G , Pharmacol Ther 74, 129-180,1997, Felder, C C & Glass, M Annu Rev Pharmacol Toxicol 38, 179-200, 1998) Los cannabinoides de C sativa (Fig 1) llevan a cabo sus efectos debido a que son similares a determinadas moléculas producidas por los animales (incluidos los humanos) que probablemente desempeñan funciones importantes en el sistema nervioso Estas moléculas se denominan por ello cannabmoides endógenos o endocannabinoides, y la anandamida (=araquιdonoιletanolamιda) es su principal representante (Di Marzo, V , Melck, D , Bisogno, T & De Petrocellis, L , Trends Neurosa 21, 521-528 1998, Martin, B R , Mechoulam, R & Razdan, R K , Ufe S 65, 573-595, 1999) Es más, se han conseguido obtener en el laboratorio compuestos que mimetizan la acción de los cannabinoides naturales pero con una potencia mucho más elevada Son los denominados cannabinoides sintéticos uno de cuyos representantes es el WIN-55,212-2 (Fig 2) (Pertwee, op cit Barth, F , Expert Opm Ther Patents 8, 301-313, 1998) Los cannabinoides, tanto los naturales como los sintéticos, actúan mediante su unión a receptores específicos de membrana (receptores de cannabinoides o de tipo CB), de los cuales se conocen hoy en día dos subtipos diferentes CBÍ y CB2 (Pertwee, op cit , Howlett, A er al , en The lUPHAR Compendium of Receptor Characteπzation and Classification, eds Godframd, T , Humphrey, P Ruffolo, R & Vanhoutte, P , lUPHAR Media, pp 97-104, 1998) No todos los tejidos del organismo poseen estos receptores se localizan principalmente en el sistema nervioso, y por eso los cannabinoides ejercen mayoπtaπamente sus efectos en el cerebro (Pertwee, op at , Childers, S R & Breivogel, C S , Drug Alcohol Depen 51, 173-187, 1998) Existe hoy en día un gran número de estudios acerca de las posibles aplicaciones terapéuticas de los cannabinoides Es más, en la actualidad ya se permite a los médicos en el Remo Unido y en diversos estados de los Estados Unidos prescribir THC o determinados cannabinoides sintéticos como estimulantes del apetito e inhibidores del vómito en pacientes con SIDA o cáncer tratados crónicamente con quimioterapia (Gπnspoon, L & Bakalar, J B , JAMA 273, 1875-1876,1995, Voth, E & Schwartz, R , Ann Intem Med 126, 791-798, 1997) Entre los usos terapéuticos potenciales de los cannabinoides destacan los siguientes (a) como agentes analgésicos se ha demostrado que actúan de forma altamente eficaz en la atenuación del dolor agudo y crónico, (b) como agentes que reducen la actividad motora se están probando hoy en día en el tratamiento de los trastornos asociados a la enfermedad de Parkinson, el corea de Huntington y la esclerosis múltiple, (c) como agentes anticonvulsivantes se estudia su aplicación en el tratamiento de la epilepsia, (d) como agentes que disminuyen la presión infraocular podrían utilizarse en el tratamiento del glaucoma (Voth y Schwartz, op cit , Manzanares, J er a/., Trends Pharmacol Sci 20, 287-294, 1999, Pop, E , Curr Opm CPNS Invest Drugs 1, 587-596, 1999, Sanudo-Pena, M C , Tsou, K & Walker, J M , Ufe Sci 65, 703-713, 1999) Algunos de estos usos terapéuticos potenciales de los cannabinoides ya han sido patentados (ver por ejemplo US4189491 , US5939429, WO9711668, WO9832441 y WO9957106)Among the brain tumors that affect humans, glioblastomas are the most frequent (1 per 50 000 people and year) malignant (mortality close to 100%) and of more rapid evolution (life expectancy of weeks / months after diagnosis ) Today, the treatment of glioblastomas is usually ineffective or merely palliative, and involves techniques such as surgery, radiotherapy, chemotherapy and immunotherapy (Louis, DN & Gusella, JF, Trenas Genet. 11, 412-415 1995 Avgeropoulos, NG & Batchelor, TT, Oncologist 4, 209-224, 1999) In addition, gene therapy has begun to be used as an experimental treatment of glioblastomas, although so far it has provided few positive results (Martuza, RL, Nature Med 3, 1323, 1997) The already unlikely success of these therapeutic approaches usually It is also complicated by factors such as rapid growth, notorious heterogeneity, high level of infiltration and extreme resistance to chemotherapy exhibited by glioblastomas (Maintz, D et al, J Neuropathol Exp Neurol 56, 1098-1 104, 1997, Masón , W, Louis, DN & Cairncross, JG, J Clin Oncol 15, 3423-3426, 1997, Martuza, op cit, Avgeropoulos & Batchelor, op cit) It seems clear therefore that the development of alternative therapies for treatment is highly recommended of brain tumors Cannabinoids are compounds that owe their name to the fact that they are synthesized by the Cannabis sativa L plant. These compounds, among which Δ 9 -tetrahιdrocannabιnol (THC) stands out for its high potency and abundance, are responsible for the central effects and peripherals of marijuana use (Pertwee, RG, Pharmacol Ther 74, 129-180,1997, Felder, CC & Glass, M Annu Rev Pharmacol Toxicol 38, 179-200, 1998) C sativa cannabinoids (Fig 1) carry ac Above its effects because they are similar to certain molecules produced by animals (including humans) that probably play important roles in the nervous system. These molecules are therefore called endogenous cannabmoids or endocannabinoids, and anandamide (= araquιdonoιletanolamιda) is their main Representative (Di Marzo, V, Melck, D, Bisogno, T & De Petrocellis, L, Trends Neurosa 21, 521-528 1998, Martin, BR, Mechoulam, R & Razdan, RK, Ufe S 65, 573-595, 1999 ) Moreover, it has been possible to obtain in the laboratory compounds that mimic the action of natural cannabinoids but with a much higher potency. They are called synthetic cannabinoids, one of whose representatives is WIN-55,212-2 (Fig 2) (Pertwee, op cit Barth, F, Expert Opm Ther Patents 8, 301-313, 1998) Cannabinoids, both natural and synthetic, act by binding to specific membrane receptors (cannabinoid receptors or CB type), of which two different subtypes CB Í and CB 2 are known today (Pertwee, op cit, Howlett, A er al, in The lUPHAR Compendium of Receptor Characteπzation and Classification, eds Godframd, T, Humphrey , P Ruffolo, R & Vanhoutte, P, lUPHAR Media, pp 97-104, 1998) Not all The body's tissues possess these receptors are located mainly in the nervous system, and that is why cannabinoids may exert their effects on the brain (Pertwee, op at, Childers, SR & Breivogel, CS, Drug Alcohol Depen 51, 173-187, 1998 ) There are today a large number of studies about the possible therapeutic applications of cannabinoids. Moreover, doctors are now allowed in the United Kingdom and in various states of the United States to prescribe THC or certain synthetic cannabinoids such as appetite stimulants and vomit inhibitors in patients with AIDS or cancer chronically treated with chemotherapy (Gπnspoon, L & Bakalar, JB, JAMA 273, 1875-1876,1995, Voth, E & Schwartz, R, Ann Intem Med 126, 791- 798, 1997) Among the potential therapeutic uses of cannabinoids, the following (a) stand out as analgesic agents have been shown to act highly effectively in the attenuation of acute pain o and chronic, (b) as agents that reduce motor activity are being tested today in the treatment of disorders associated with Parkinson's disease, Huntington's chorea and multiple sclerosis, (c) as anticonvulsant agents its study is studied application in the treatment of epilepsy, (d) as agents that decrease infraocular pressure could be used in the treatment of glaucoma (Voth and Schwartz, op cit, Manzanares, J er a /., Trends Pharmacol Sci 20, 287-294, 1999, Pop, E, Curr Opm CPNS Invest Drugs 1, 587-596, 1999, Sanudo-Pena, MC, Tsou, K & Walker, JM, Ufe Sci 65, 703-713, 1999) Some of these potential therapeutic uses of the cannabinoids have already been patented (see for example US4189491, US5939429, WO9711668, WO9832441 and WO9957106)
Uno de los efectos más intrigantes e inexplorados de los cannabinoides es su capacidad para inhibir el crecimiento de células transformadas in vitro Así, se ha demostrado que diversos cannabinoides inhiben la proliferación de células de tumor de mama MCF-7 (De Petrocellis, L et al, Proc Nati Acad. Sci. USA 95, 8375-8380, 1998), de células de glioblastoma C6 (Sánchez, C , Galve-Roperh, I , Canova, C , Brachet, P & Guzmán, M , FEBS Lett 436, 6-10, 1998) y de células de tumor de próstata PC-3 (Ruiz, L , Miguel, A & Díaz-Laviada, I , FEBS Lett 458, 400-404, 1999) Sin embargo, estos hallazgos en sistemas de cultivo celular nunca han sido hasta ahora observados in vivo, por lo que su significado biomédico es desconocidoOne of the most intriguing and unexplored effects of cannabinoids is their ability to inhibit the growth of transformed cells in vitro. Thus, it has been shown that various cannabinoids inhibit the proliferation of MCF-7 breast tumor cells (De Petrocellis, L et al. , Proc Nati Acad. Sci. USA 95, 8375-8380, 1998), of C6 glioblastoma cells (Sánchez, C, Galve-Roperh, I, Canova, C, Brachet, P & Guzmán, M, FEBS Lett 436, 6-10, 1998) and PC-3 prostate tumor cells (Ruiz, L, Miguel, A & Díaz-Laviada, I, FEBS Lett 458, 400-404, 1999) Without However, these findings in cell culture systems have never been observed in vivo so far, their biomedical significance is unknown.
Descripción de la invenciónDescription of the invention
La presente invención hace uso por primera vez de los cannabinoides para el tratamiento de tumores cerebrales, y se basa en nuestras observaciones originales de que los cannabinoides inducen una marcada regresión (que conlleva un alargamiento de la vida) e incluso la erradicación (que conlleva la curación) de ghoblastomas en animales de laboratorio Esta invención implica la utilización de una terapia técnicamente simple, carente de efectos colaterales notorios y, lo que es más importante, muy eficaz para el tratamiento de tumores cerebrales, que como se ha comentado anteriormente no pueden ser tratados hoy en día de manera satisfactoria con otras técnicas o compuestos Los experimentos que han conducido a la presente invención se detallan a continuaciónThe present invention makes use of cannabinoids for the treatment of brain tumors for the first time, and is based on our original observations that cannabinoids induce a marked regression (which leads to a lengthening of life) and even eradication (which entails Cure) of ghoblastomas in laboratory animals This invention involves the use of a technically simple therapy, devoid of noticeable side effects and, more importantly, very effective for the treatment of brain tumors, which as mentioned above cannot be treated satisfactorily today with other techniques or compounds The experiments that have led to the present invention are detailed below.
Acción antitumoral de los cannabinoides en ratasAntitumor action of cannabinoids in rats
La inyección de células de glioblastoma C6 en el cerebro de rata se utiliza ampliamente como modelo experimental de tumor cerebral maligno (Barth, R F , J Neur∞ncol 36, 91-102, 1998) Se inocularon células de glioblastoma C6 directamente en el cerebro de ratas Wistar, y los tumores se visualizaron por resonancia magnética Todos los animales que se dejaron sin tratar murieron uniformemente 12-18 días después de la inoculación de las células (Fig 3a). Para evaluar el potencial antitumoral de los cannabinoides, 12 días después de la inoculación de las células se administró a un grupo de animales durante 7 días THC o WIN-55,212-2 a través de una cánula localizada en el sitio de inoculación Los animales tratados con cannabinoides tuvieron una vida significativamente más larga que los animales control (Fig 3a). Así, la administración de cannabinoides consiguió incrementar la supervivencia a 19-35 días en 9/15 animales (tratamiento con THC) o a 19-43 días en 4/15 animales (tratamiento con WIN-55,212-2) Es más, los cannabinoides erradicaron completamente el tumor en 3/15 animales (tratamiento con THC) o en 5/15 animales (tratamiento con WIN-55,212-2) En la Fig 3b se muestra una imagen de resonancia magnética de uno de los animales curados con THC, tras la administración del cannabinoide la masa tumoral desaparecía completamente, y en su lugar se observaba una zona hipomteπsa residual que se interpreta como una cicatriz fibrosa en el sitio de inoculación No se observó recurrencia alguna en los 8 animales curados con caπnabmoidesInjection of C6 glioblastoma cells in the rat brain is widely used as an experimental model of malignant brain tumor (Barth, RF, J Neur∞ncol 36, 91-102, 1998) C6 glioblastoma cells were inoculated directly into the brain of Wistar rats, and tumors were visualized by magnetic resonance imaging All animals that were left untreated died uniformly 12-18 days after cell inoculation (Fig 3a). To assess the antitumor potential of cannabinoids, 12 days after cell inoculation, a group of animals was administered for 7 days THC or WIN-55,212-2 through a cannula located at the site of inoculation. Animals treated with Cannabinoids had a significantly longer life than control animals (Fig 3a). Thus, the administration of cannabinoids managed to increase survival to 19-35 days in 9/15 animals (treatment with THC) or 19-43 days in 4/15 animals (treatment with WIN-55,212-2) Moreover, cannabinoids completely eradicated the tumor in 3 / 15 animals (treatment with THC) or in 5/15 animals (treatment with WIN-55,212-2) An image of magnetic resonance imaging of one of the animals cured with THC is shown in Fig 3b, after administration of the cannabinoid tumor mass disappeared completely, and instead a residual hypomteπsa area was observed that is interpreted as a fibrous scar at the site of inoculation No recurrence was observed in the 8 animals cured with caπnabmoides
Acción antitumoral de los cannabinoides en ratones inmunodeficientesAntitumor action of cannabinoids in immunodeficient mice
Para discernir si la acción antiproliferativa de los cannabinoides se debe a un efecto directo sobre las células tumorales o a un efecto indirecto mediado por una respuesta inmune, se inocularon subcutáneamente células de glioblastoma C6 en ratones deficientes en la recombinasa RAG-2 (RAG- 2'1'), que carecen de linfocitos T y B maduros (Shinkai, Y et al , Ce// 68, 855- 867 1992) Como se muestra en la Fig 4a, el tamaño de los tumores era extraordinariamente inferior en los animales tratados con THC o WIN- 55,212-2 que en los animales control En la Fig 4b se muestran ejemplos de ratones portadores de tumores y de tumores diseccionados después del tratamiento con o sin cannabinoides durante 7 díasTo discern whether the antiproliferative action of cannabinoids is due to a direct effect on tumor cells or an indirect effect mediated by an immune response, C6 glioblastoma cells were inoculated subcutaneously in mice deficient in RAG-2 recombinase (RAG-2 ' 1 ' ), lacking mature T and B lymphocytes (Shinkai, Y et al, Ce // 68, 855-867 1992) As shown in Fig 4a, the tumor size was extraordinarily smaller in animals treated with THC or WIN-55,212-2 than in control animals Examples of tumor-bearing mice and dissected tumors after treatment with or without cannabinoids for 7 days are shown in Fig 4b
Segundad del tratamiento con cannabinoides in vivo Se examinaron a continuación los posibles efectos secundarios del tratamiento con cannabinoides Las ratas sin tumor y tratadas con cannabinoides no veían afectada en absoluto su supervivencia (Fig 3a) Al igual que en los 8 animales antes mencionados cuyos tumores fueron erradicados con cannabinoides, el análisis minucioso por resonancia magnética de todos los animales sin tumor revelaba que el tratamiento con cannabinoides no producía ninguna señal de daño por necrosis, edema, infección, inflamación o trauma Para descartar la posibilidad de que los cannabinoides fueran tóxicos para las células nerviosas en división, se realizaron tinciones de TÚNEL en la zona subventπcular del cerebro de las ratas, que continúa en proliferación en el animal adulto La administración de cannabinoides no sólo no produjo ningún efecto apoptótico significativo en el cerebro in vivo, sino que además el ligero mareaje observado en el caudado putamen de los animales control no era evidente en los animales tratados con cannabinoidesSecondary treatment with cannabinoids in vivo The possible side effects of cannabinoid treatment were examined below Rats without tumor and treated with cannabinoids were not affected at all their survival (Fig 3a) As in the 8 animals mentioned above whose tumors were eradicated with cannabinoids, thorough magnetic resonance analysis of all animals without tumor revealed that treatment with cannabinoids produced no sign of damage due to necrosis, edema, infection, inflammation or trauma to rule out the possibility that Cannabinoids were toxic to dividing nerve cells, TUNNEL stains were performed in the subventπcular area of the rat's brain, which continues to proliferate in the adult animal. Cannabinoid administration not only did not produce any significant apoptotic effect on the brain in vivo. , but also the slight dizziness observed in the caudate putamen of the control animals was not evident in the animals treated with cannabinoids
Tanto en los animales sin tumor como en los portadores de tumor, los cannabinoides no indujeron ninguna alteración significativa de parámetros comportamentales como coordinación motora y actividad física La ingesta de agua y alimento, así como la ganancia de peso, tampoco se vieron afectadas por los cannabinoides Así mismo, en los análisis de sangre los parámetros bioquímicos (glucosa, urea, ácido úrico, creatinina, colesterol, bilirrubina) y los marcadores de daño tisular (alanina y aspartato ammotransferasas, γ-glutamiltransferasa, creatina quinasa, lactato deshidrogenasa) no resultaron afectados ni a lo largo del periodo de 7 días de administración ni hasta 2 meses después de la finalización del tratamiento con cannabinoides Datos de otros autores apoyan la idea de que los cannabinoides no sólo no son compuestos tóxicos para las células nerviosas, sino que incluso las protegen de estímulos tóxicos tales como los agonistas glutamaérgicos (Skaper, S D et al , Proc Nati Acad Sα USA 93, 3984-3989, 1996, Shen, M & Thayer, S A , Mol Pharmacol 54, 459-462, 1998), los agentes oxidantes (Hampson, A J , Gπmaldi, M Axelrod, J & Wink, D , Proc Nati Acad Sci USA 95, 8268-8273, 1998) y la isquemia (Nagayama, T er a/ , J Neurosa 19, 2987-2995, 1999)In both animals without tumor and in tumor carriers, cannabinoids did not induce any significant alteration of behavioral parameters such as motor coordination and physical activity. Water and food intake, as well as weight gain, were also not affected by cannabinoids. Likewise, in the blood tests the biochemical parameters (glucose, urea, uric acid, creatinine, cholesterol, bilirubin) and the markers of tissue damage (alanine and aspartate ammotransferases, γ-glutamyltransferase, creatine kinase, lactate dehydrogenase) were not affected neither over the period of 7 days of administration nor up to 2 months after the end of cannabinoid treatment Data from other authors support the idea that cannabinoids are not only not toxic compounds for nerve cells, but even protect them of toxic stimuli such as glutamatergic agonists (Skaper, SD et al, Pr oc Nati Acad Sα USA 93, 3984-3989, 1996, Shen, M & Thayer, SA, Mol Pharmacol 54, 459-462, 1998), oxidizing agents (Hampson, AJ, Gπmaldi, M Axelrod, J & Wink, D , Proc Nati Acad Sci USA 95, 8268-8273, 1998) and ischemia (Nagayama, T er a /, J Neurosa 19, 2987-2995, 1999)
Caracterización farmacológica de la acción antitumoral de los cannabinoidesPharmacological characterization of the antitumor action of cannabinoids
Se llevaron a cabo experimentos encaminados a caracterizar farmacológicamente la muerte inducida por cannabinoides de células de glioblastoma C6 en cultivo Agonistas sintéticos de elevada potencia comoExperiments aimed at pharmacologically characterizing the cannabinoid-induced death of C6 glioblastoma cells in culture High potency synthetic agonists such as
WIN-55,212-2, CP-55,940 y HU-210 inducían la muerte de estas células a dosis menores que el THC, como cabía esperar de su mayor afinidad por los receptores de cannabinoides (Pertwee, op at ) Así, tras 5 días de exposición a cannabinoides, la viabilidad del glioblastoma C6 se veía reducida en un 50% a concentraciones de 20 nM WIN-55, 212-2, 45 nM CP- 55,940, 10 nM HU-210 y 480 nM THC (n=4) Ni el SR141716 (un antagonista selectivo de CB ni el SR144528 (un antagonista selectivo de CB2) (Shire, D et al , Life Sci 65, 627-635, 1999) eran capaces por separado de prevenir la muerte celular inducida por el THC Sin embargo, cuando los dos antagonistas se añadían conjuntamente a las incubaciones se observaba una prevención eficaz de la muerte celular inducida por THC (Fig 5a) De acuerdo con ello, el análisis por Western blot demostró que las células de glioblastoma C6 expresaban tanto el receptor CBi como el CB2 (Fig 5b)WIN-55,212-2, CP-55,940 and HU-210 induced the death of these cells at doses lower than THC, as expected from its higher affinity for cannabinoid receptors (Pertwee, op at) Thus, after 5 days of exposure to cannabinoids, the viability of glioblastoma C6 was reduced by 50% at concentrations of 20 nM WIN-55, 212-2, 45 nM CP- 55,940, 10 nM HU-210 and 480 nM THC (n = 4) Neither SR141716 (a selective CB antagonist nor SR144528 (a selective CB 2 antagonist)) Shire, D et al, Life Sci 65, 627-635, 1999) were able separately to prevent THC-induced cell death. However, when the two antagonists were added together to the incubations an effective prevention of death was observed. THC-induced cell (Fig 5a) Accordingly, Western blot analysis showed that C6 glioblastoma cells expressed both the CBi and CB 2 receptors (Fig 5b)
Aplicación de la invención en otros supuestos Los experimentos que han conducido a la presente invención se han llevado a cabo con ratas y ratones como animales portadores de tumores Sin embargo, tanto por el diseño experimental empleado como por la similitud que exhiben los tumores cerebrales en distintos mamíferos (R F Barth, op cit ), la invención puede aplicarse al tratamiento de tumores cerebrales en otros mamíferos, incluido el ser humanoApplication of the invention in other cases The experiments that have led to the present invention have been carried out with rats and mice as tumor-bearing animals. However, both for the experimental design employed and for the similarity that brain tumors exhibit in different mammals (RF Barth, op cit), the invention can be applied to the treatment of brain tumors in other mammals, including humans
Los experimentos que han conducido a la presente invención se han llevado a cabo con glioblastomas como modelo de tumor cerebral Sin embargo, por el diseño experimental que se ha utilizado para la inducción y tratamiento de los tumores, la invención puede aplicarse al tratamiento de otros tumores cerebrales, por ejemplo meduloepiteliomas, meduloblastomas, neuroblastomas, germinomas, carcinomas embrionarios, astrocitomas, astroblastomas, ependimomas, oligodendrog omas, carcinomas plexales, neuroepitehomas, pineoblastomas, ependimoblastomas, tumores neuroectodérmicos, meningiomas malignos, condrosarcomas, sarcomatosomas meningeales, melanomas malignos o schwanomas malignos Los experimentos que han conducido a la presente invención se han llevado a cabo con dos cannabinoides paradigmáticos, uno natural (THC) y otro sintético (WIN-55,212-2) En una realización preferida de la invención se utilizará el cannabinoide con efecto antiproliferativo más potente para un determinado tumor Sin embargo, puesto que el efecto antiproliferativo de estos compuestos está mediado por los receptores de cannabinoides (receptores de tipo CB, Howlett et al , op αt ), la invención es aplicable a cualquier otro agonista de estos receptores, tanto cannabmoides de C sativa (por ejemplo Δ8-tetrahιdrocannabιnol, cannabinol, cannabidiol) (Fig 1) como cannabinoides sintéticos (por ejemplo HU-210, CP-55,940, CP-50,556) (Fig 2) (Pertwee, op αt , F Barth, op cit ) También se incluyen en este apartado los medicamentos que contengan en su composición cualquier cannabinoideThe experiments that have led to the present invention have been carried out with glioblastomas as a brain tumor model. However, because of the experimental design that has been used for the induction and treatment of tumors, the invention can be applied to the treatment of other tumors. brain diseases, for example medulloepitheliomas, medulloblastomas, neuroblastomas, germinomas, embryonic carcinomas, astrocytomas, astroblastomas, ependymomas, oligodendrog omas, plexal carcinomas, neuroepitehomas, pineoblastomas, ependyloblastomas, neuroectodermal tumors, malignant melanomas, malignant melanomas The experiments that have led to the present invention have been carried out with two paradigmatic cannabinoids, one natural (THC) and one synthetic (WIN-55,212-2) In a preferred embodiment of the invention, the cannabinoid with more potent antiproliferative effect will be used for a given tumor However, since the antiproliferative effect of these compounds is mediated by cannabinoid receptors (CB type receptors, Howlett et al, op αt), the invention is applicable to any other agonist of these receptors, both cannabmoids. of C sativa (for example Δ 8 -tetrahιdrocannabιnol, cannabinol, cannabidiol) (Fig 1) as synthetic cannabinoids (for example HU-210, CP-55,940, CP-50,556) (Fig 2) (Pertwee, op αt, F Barth, op cit) Also included in this section are medications that contain any cannabinoid in their composition
Los experimentos que han conducido a la presente invención se han llevado a cabo administrando intratumoralmente el cannabinoide En una realización preferida de la invención ésta será la vía de administración elegida, ya que permite una elevada accesibilidad del cannabinoide al tumor Sin embargo, puesto que la acción del cannabinoide es directa sobre el tumor y no parece afectar sustancialmente a sistemas periféricos, la vía de administración puede ser también sistémica, por ejemplo intrapeπtoneal, intravenosa u oralThe experiments that have led to the present invention have been carried out by intratumorally administering the cannabinoid. In a preferred embodiment of the invention this will be the chosen route of administration, since it allows a high accessibility of the cannabinoid to the tumor. However, since the action of the cannabinoid is direct on the tumor and does not seem to substantially affect peripheral systems, the route of administration can also be systemic, for example intrapeπtoneal, intravenous or oral
Los experimentos que han conducido a la presente invención se han llevado a cabo mediante la administración continua de una dosis de cannabinoide durante un tiempo determinado En una realización preferida de la invención se optimizarán estos parámetros dependiendo de los requerimientos específicos del tratamiento estado del paciente, tamaño y localización del tumor, número de tumores, etc Así, por ejemplo, el modo de aplicación podrá ser continuo (modo preferido) o secuencial en una o varias dosis por día Esto hará obviamente variar las dosis de compuesto administradas y el tiempo total de duración del tratamientoThe experiments that have led to the present invention have been carried out by the continuous administration of a dose of cannabinoid for a certain time. In a preferred embodiment of the invention these parameters will be optimized depending on the specific requirements of the treatment patient condition, size and tumor location, number of tumors, etc. Thus, for example, the mode of application may be continuous (preferred mode) or sequential in one or more dose per day This will obviously vary the doses of compound administered and the total duration of treatment.
Breve descripción de las figurasBrief description of the figures
Figura 1Figure 1
Fórmula química de los principales cannabmoides de C sativaChemical formula of the main C sativa cannabmoids
Figura 2Figure 2
Fórmula química de los principales cannabinoides sintéticosChemical formula of the main synthetic cannabinoids
Figura 3Figure 3
Acción antitumoral de los cannabinoides en ratas (a) Curvas de supervivencia de ratas con tumores cerebrales Se indujeron glioblastomas en 45 ratas (día 0), 15 animales no fueron tratados con cannabinoides (---), mientras que otros 15 se trataron con THC ( — ) y otros 15 con WIN-55,212-2 ( ) entre los días 12 y 19 Los animales tratados con cannabinoides vivieron significativamente más tiempo que los controles (P<0 01 por el test log-rank) Se administró así mismo THC y WIN-55,212-2 a 5 ratas cada uno en las que no se indujo tumor alguno (- - )Antitumor action of cannabinoids in rats (a) Survival curves of rats with brain tumors Glioblastomas were induced in 45 rats (day 0), 15 animals were not treated with cannabinoids (---), while another 15 were treated with THC (-) and another 15 with WIN-55,212-2 () between days 12 and 19 Animals treated with cannabinoids lived significantly longer than controls (P <0 01 by the log-rank test) THC was also administered and WIN-55,212-2 to 5 rats each in which no tumor was induced (- -)
(b) Imagen de resonancia magnética en proyecciones axial (superior) y coronal (inferior) del cerebro de una rata antes (izquierda) y después(b) Magnetic resonance imaging in axial (upper) and coronal (lower) projections of a rat's brain before (left) and after
(derecha) del tratamiento con THC Un glioblastoma de 100 mm3 (flecha) fue erradicado con 500 μg de THC La imagen fue tomada 7 días después de la finalización del tratamiento con THC(right) of THC treatment A 100 mm 3 glioblastoma (arrow) was eradicated with 500 μg of THC The image was taken 7 days after the end of THC treatment
Figura 4Figure 4
Acción antitumoral de los cannabinoides en ratones inmunodeficientes (a) Se indujeron glioblastomas en 18 ratones Cuando los tumores alcanzaron el tamaño deseado (día 0), 6 animales fueron tratados con vehículo (o), mientras que otros 6 se trataron con THC (•) y otros 6 con WIN- 55,212-2 (z) durante 7 días El tamaño de los tumores en los animales tratados con cannabinoides era significativamente menor que en los controles a todos los tiempos (P<0 01 por el test f de Student) (b) Ejemplos de glioblastomas en ratones (arriba) y diseccionadosAnti-tumor action of cannabinoids in immunodeficient mice (a) Glioblastomas were induced in 18 mice When the tumors reached the desired size (day 0), 6 animals were treated with vehicle (o), while another 6 were treated with THC (•) and another 6 with WIN- 55,212-2 (z) for 7 days The size of tumors in animals treated with cannabinoids was significantly smaller than in controls a all times (P <0 01 by Student's f test) (b) Examples of glioblastomas in mice (above) and dissected
(abajo, barra 1 cm) tras el tratamiento durante 7 días con vehículo, THC o WIN-55,212-2 (WIN)(bottom, 1 cm bar) after treatment for 7 days with vehicle, THC or WIN-55,212-2 (WIN)
Figura 5Figure 5
Implicación de los receptores de cannabinoides en la muerte celularInvolvement of cannabinoid receptors in cell death
(a) Se cultivaron células de glioblastoma C6 durante 5 días en ausencia o presencia de 1 μM THC, 1 μM SR141716 (SR1 ) y/o 1 μM SR144528 (SR2) (n=6) *Sιgnιfιcatιvamente diferente de las incubaciones sin adiciones (P<0 01 por el test r de Student)(a) C6 glioblastoma cells were cultured for 5 days in the absence or presence of 1 μM THC, 1 μM SR141716 (SR1) and / or 1 μM SR144528 (SR2) (n = 6) * Sιgnιfιcatιvably different from incubations without additions ( P <0 01 by Student's r test)
(b) Presencia de receptores de cannabinoides CBT y CB2 en células de glioblastoma C6 La detección de los receptores se realizó por Western ó/or con anticuerpos específicos para cada uno de los dos receptores(b) Presence of CB T and CB 2 cannabinoid receptors in C6 glioblastoma cells. The detection of the receptors was performed by Western or / or with specific antibodies for each of the two receptors.
Modo de realización de la invenciónEmbodiment of the invention
La presente invención se ilustra adicionalmente con los ejemplos que se exponen a continuaciónThe present invention is further illustrated by the examples set forth below.
E|emplo 1E | emplo 1
Curación de glioblastomas en ratasGlioblastoma cure in rats
Se anestesiaron ratas Wistar macho (250-300 g de peso corporal) conMale Wistar rats (250-300 g body weight) were anesthetized with
3% isofluorano en una mezcla de oxígeno (0 8 l/mm) y protóxido (0 4 l/mm)3% isofluorane in a mixture of oxygen (0 8 l / mm) and protoxide (0 4 l / mm)
Se prepararon 5x106 células de glioblastoma C6 en 100 μl de solución salina tamponada con fosfato (PBS) suplementada con 0 1% glucosa y se inyectaron estereotaxicamente en el lóbulo fronto-paπetal del hemisferio cerebral derecho (4 mm a la derecha de bregma, 4 5 mm de profundidad desde el cráneo) (Izquierdo, M et al , Gene Ther 2, 66-69, 1995) Las ratas recibieron dexametasona (2 mg/l) y tetraciclina (75 mg/kg de peso corporal) en el agua durante 3 días antes y 7 días después de la inoculación de las células Se llevó a cabo un exhaustivo seguimiento de los tumores mediante resonancia magnética por métodos descritos por otros autores (Izquierdo et al , op αt , Cortés, M L , de Felipe, P , Martín, V , Hughes, M A & Izquierdo, M , Gene Ther 5, 1499-1507, 1998)5x10 6 cells of C6 glioblastoma were prepared in 100 µl phosphate buffered saline (PBS) supplemented with 0 1% glucose and injected stereotaxically into the frontal-paputal lobe of the hemisphere right brain (4 mm to the right of bregma, 4.5 mm deep from the skull) (Left, M et al, Gene Ther 2, 66-69, 1995) Rats received dexamethasone (2 mg / l) and tetracycline ( 75 mg / kg of body weight) in the water for 3 days before and 7 days after the inoculation of the cells A thorough follow-up of the tumors was performed by magnetic resonance by methods described by other authors (Izquierdo et al, op αt, Cortés, ML, by Felipe, P, Martín, V, Hughes, MA & Izquierdo, M, Gene Ther 5, 1499-1507, 1998)
La administración de cannabinoides a las ratas comenzó 12 días después de la inoculación de las células En este momento, el tamaño medio de los tumores era de 70 mm3 (intervalo 25-100 mm3) estimado por resonancia magnética (Izquierdo et al , op cit , Cortés, de Felipe, Martin Hughes & Izquierdo, op αt ) Los cannabinoides se administraron mediante una cánula situada en el lugar de inoculación del tumor y fijada al cráneo con cemento dental, un pequeño tomillo de acero inoxidable aseguraba la cánula y el cemento dental La cánula se conectó subcutáneamente mediante un catéter a una mini-bomba osmótica (Alzet 2001 ) que operaba a un flujo de 1 μl/h durante 7 días La bomba osmótica se rellenó con 500-2500 μg de THC o 50-250 μg de WIN-55,212-2 en 200 μl de PBS suplementado con 5 mg/ml de albúmina de suero bovino (BSA) des pidizada y dializada Las dosis de cannabinoides utilizadas dependieron de las características del tumor a tratar, las dosis mayores se emplearon para tumores grandes, densos e invasivosCannabinoid administration to rats began 12 days after cell inoculation. At this time, the average tumor size was 70 mm 3 (range 25-100 mm 3 ) estimated by magnetic resonance imaging (Izquierdo et al, op cit, Cortés, by Felipe, Martin Hughes & Izquierdo, op αt) The cannabinoids were administered by a cannula located at the site of inoculation of the tumor and fixed to the skull with dental cement, a small stainless steel thyme secured the cannula and cement Dental The cannula was connected subcutaneously through a catheter to an osmotic mini-pump (Alzet 2001) that operated at a flow of 1 μl / h for 7 days The osmotic pump was filled with 500-2500 μg THC or 50-250 μg WIN-55,212-2 in 200 μl of PBS supplemented with 5 mg / ml of ordered and dialyzed bovine serum albumin (BSA) The doses of cannabinoids used depended on the characteristics of the tumor to be treated, the higher doses were used aron for large, dense and invasive tumors
Como se observa en la Fig 3a, todos los animales que se dejaron sin tratar murieron uniformemente 12-18 días después de la inoculación de las células Los animales tratados con cannabinoides tuvieron una vida significativamente más larga que los animales control Es más, los cannabinoides erradicaron completamente el tumor en un porcentaje significativo de animales En la Fig 3b se muestra una imagen de resonancia magnética de uno de los animales curados con THC, tras la administración del cannabinoide la masa tumoral desaparece completamente, y se observa una zona hipointensa residual que se interpreta como una cicatriz fibrosa en el sitio de inoculación No se observó recurrencia alguna en los animales curados con cannabinoidesAs seen in Fig 3a, all animals that were left untreated died uniformly 12-18 days after cell inoculation. Animals treated with cannabinoids had a significantly longer life than control animals. Moreover, cannabinoids eradicated completely the tumor in a significant percentage of animals In Fig 3b, an MRI image of one of the animals cured with THC is shown, after the administration of the cannabinoid the tumor mass disappears completely, and a residual hypointense zone is observed that is interpreted as a fibrous scar at the site of inoculation No recurrence was observed in animals cured with cannabinoids
Ejemplo 2Example 2
Curación de qlioblastomas en ratones mmunodeficientesHealing of qlioblastomas in immunodeficient mice
Se indujeron tumores en ratones RAG-2 '" mediante inoculación subcutánea de 5x106 células de ghoblastoma C6 en 100 μl de PBS suplementado con 0 1% glucosa Unos 10 días después, cuando el volumen medio de los tumores era de 250 mm3 (intervalo 200-300 mm3), los animales se dividieron al azar en 3 grupos y se les inyectó durante 7 días vehículo,Tumors were induced in RAG-2 '" mice by subcutaneous inoculation of 5x10 6 cells of C6 ghoblastoma in 100 µl of PBS supplemented with 0 1% glucose About 10 days later, when the average volume of the tumors was 250 mm 3 (range 200-300 mm 3 ), the animals were randomly divided into 3 groups and injected for 7 days vehicle,
500 μg de THC o 50 μg de WIN-55,212-2 por día en 100 μl de PBS suplementado con 5 mg/ml de BSA deslipidizada y dializada Las dimensiones de los tumores se midieron con un calibrador y se calculó su volumen como (4π/3) x (anchura/2)2 x (longιtud/2)500 mg of THC or 50 μ g of WIN 55,212-2-day in 100 .mu.l of PBS supplemented with 5 mg / ml of delipidized and dialyzed BSA Dimensions of the tumors were measured with a caliper and its volume was calculated as (4π / 3) x (width / 2) 2 x (length / 2)
Como se muestra en la Fig 4a, el tamaño de los tumores era extraordinariamente inferior en los animales tratados con THC o WIN-As shown in Fig 4a, the tumor size was extraordinarily smaller in animals treated with THC or WIN-
55,212-2 que en los animales control En la Fig 4b se muestran ejemplos de ratones portadores de tumores y de tumores diseccionados después del tratamiento con o sin cannabinoides durante 7 días55,212-2 than in control animals Examples of mice bearing tumors and dissected tumors after treatment with or without cannabinoids for 7 days are shown in Fig 4b
Eiemplo 3Example 3
Implicación de los receptores de cannabinoides en la muerte de células de qlioblastoma Se cultivaron a 37°C y 5% CO2 células de glioblastoma C6 en medioImplication of cannabinoid receptors in cell death qlioblastoma were cultured at 37 ° C and 5% CO 2 C6 glioblastoma cells in medium
F12 suplementado con suero fetal de ternera al 10%, 24 h antes del comienzo del experimento las células se transfirieron a medio F12 libre suero y suplementado con insulina (5 μg/ml), transfernna (5 μg/ml), selenito sódico (5 μg/ml) y BSA deslipidizada y dializada (10 mg/ml) El medio se renovó cada 48 h y la viabilidad celular se determinó mediante el método del MTT (Sánchez, C , Galve-Ropem, I , Canova, C , Brachet, P & Guzman M , op at ) Como se observa en la Fig 5a, el THC era capaz de inducir la muerte de las células de glioblastoma C6 Además, cuando se añadían simultáneamente al medio SR141716 (antagonista selectivo de CBi) y SR144528 (antagonista selectivo de CB2) se prevenía la muerte celular inducida por el THCF12 supplemented with 10% fetal calf serum, 24 h before the start of the experiment, the cells were transferred to serum free F12 medium and supplemented with insulin (5 μg / ml), transfernna (5 μg / ml), sodium selenite (5 μg / ml) and elliptical and dialyzed BSA (10 mg / ml) The medium was renewed every 48 h and cell viability was determined by the MTT method (Sánchez, C, Galve-Ropem, I, Canova, C, Brachet, P & Guzman M, op at) As shown in Fig 5a, THC was capable of inducing the death of C6 glioblastoma cells also when added simultaneously SR141716 medium (selective antagonist of CBi) and SR144528 ( a selective antagonist of CB 2) prevented THC-induced cell death
Para comprobar que ambos receptores estaban presentes en las células C6, las células se lavaron con PBS, se rascaron las placas en medio de lisis y se obtuvo la fracción particulada mediante centrifugación a 40 OOOg durante 60 min (Sánchez, C , Galve-Roperh, I , Canova, C , Brachet, P & Guzmán, M , op at ) Se sometieron las muestras a electroforesis en geles de poliacπlamida con dodecil sulfato sódico y las proteínas se transfirieron de los geles a membranas de nitrocelulosa Las membranas se bloquearon con BSA deshpidizada y dializada al 1%, y se incubaron con un anticuerpo frente a los residuos 1-14 del receptor CBi de rata (diluido 1 5000) o con un anticuerpo frente a los residuos 350-361 del receptor CB2 humano (diluido 1 2000) Las muestras se sometieron finalmente a revelado con un "kit" de electroquimioluminiscencia (Amersham, Bucks, Reino Unido) Como muestra la Fig 5b, las células de glioblastoma C6 expresaban tanto el receptor CBÍ como el CB2 To verify that both receptors were present in C6 cells, the cells were washed with PBS, the plates were scratched in lysis medium and the particulate fraction was obtained by centrifugation at 40 OOOg for 60 min (Sánchez, C, Galve-Roperh, I, Canova, C, Brachet, P & Guzmán, M, op at) The samples were electrophoresed in polyacrylamide gels with sodium dodecyl sulfate and the proteins were transferred from the gels to nitrocellulose membranes The membranes were blocked with dehpidized BSA and 1% dialyzed, and incubated with an antibody against residues 1-14 of the rat CBi receptor (diluted 1 5000) or with an antibody against residues 350-361 of the human CB 2 receptor (diluted 1 2000) The samples were finally developed with an electrochemiluminescence "kit" (Amersham, Bucks, United Kingdom) As shown in Fig 5b, C6 glioblastoma cells expressed both the CB Í receptor and CB 2 receptor
Eiemplo 4Example 4
Segundad del tratamiento con cannabinoides m vivo Se administraron los cannabmoides (2500 μg de THC o 250 μg de WIN-55,212-2) a ratas sin tumores durante 7 días tal y como se ha descrito anteriormente Las ratas fueron entonces sacrificadas y sus cerebros se fijaron con 4% paraformaldehído en PBS La muerte por apoptosis se determinó en secciones del cerebro de 40 μm de espesor utilizando un "kit" de tinción TÚNEL de acuerdo con las instrucciones del suministrador (Boehπnger, Mannheim, Alemania) El mareaje de hebras de DNA con desoxiundina trifosfato marcada con fluoresceina se visualizó mediante un microscopio confocal (longitud de onda de excitación 488 nm, longitud de onda de emisión 525 nm) La intensidad del láser y la sensibilidad del fotodetector se mantuvieron constantes para permitir la comparación entre tratamientos Se analizaron al menos 5 campos ópticos por animalSecondary treatment with live m cannabinoids Cannabmoids (2500 μg THC or 250 μg WIN-55,212-2) were administered to rats without tumors for 7 days as described above. Rats were then sacrificed and their brains fixed. with 4% paraformaldehyde in PBS Death by apoptosis was determined in 40 µm thick sections of the brain using a "TUNNEL staining kit" according to the supplier's instructions (Boehπnger, Mannheim, Germany) DNA stranding with Deoxyundin triphosphate labeled with fluorescein was visualized by a confocal microscope (excitation wavelength 488 nm, length of 525 nm emission wave) The laser intensity and photodetector sensitivity were kept constant to allow comparison between treatments At least 5 optical fields were analyzed per animal
Se realizaron tinciones de TÚNEL en la zona subventπcular del cerebro de las ratas, que continúa en proliferación en el animal adulto La administración de cannabinoides no sólo no produjo ningún efecto apoptótico significativo en el cerebro m vivo, sino que además el ligero mareaje observado en el caudado putamen de los animales control no se observaba en los animales tratados con cannabinoides TUNNEL stains were performed in the subventπcular area of the rat brain, which continues to proliferate in the adult animal. The administration of cannabinoids not only did not produce any significant apoptotic effect on the living brain, but also the slight dizziness observed in the caudate putamen of control animals was not observed in animals treated with cannabinoids

Claims

ReivindicacionesClaims
1 - Uso de cannabinoides naturales y sintéticos en la fabricación de un medicamento para el tratamiento terapéutico, en mamíferos incluyendo seres humanos, de tumores cerebrales seleccionados del grupo que comprende glioblastomas, meduloepitehomas, meduloblastomas, neuroblastomas, germinomas, carcinomas embrionarios, astrocitomas, astroblastomas, ependimomas, ohgodendrogliomas, carcinomas plexales, neuroepiteliomas, pineoblastomas, epandimoblastomas, tumores neuroectodérmicos, meningiomas malignos, condrosarcomas, sarcomatosomas meningeales, melanomas malignos y schwanomas malignos1 - Use of natural and synthetic cannabinoids in the manufacture of a medicament for therapeutic treatment, in mammals including humans, of brain tumors selected from the group comprising glioblastomas, medulloepitehomas, medulloblastomas, neuroblastomas, germinomas, embryonic carcinomas, astrocytomas, astroblastomas, ependymomas, ohgodendrogliomas, plexal carcinomas, neuroepitheliomas, pineoblastomas, epandimoblastomas, neuroectodermal tumors, malignant meningiomas, chondrosarcomas, meningeal sarcomatosomes, malignant melanomas and malignant schwannomas
2 - Uso según la reivindicación 1 , en el que los tumores cerebrales son ghoblastomas2 - Use according to claim 1, wherein the brain tumors are ghoblastomas
3 - Uso según las reivindicaciones 1 y 2, en el que los cannabinoides naturales se seleccionan del grupo formado por Δ9-tetrahιdrocannabιnol (THC), Δ8-tetrahιdrocannabιnol, cannabinol y cannabidiol3 - Use according to claims 1 and 2, wherein the natural cannabinoids are selected from the group consisting of Δ 9 -tetrahιdrocannabιnol (THC), Δ 8 -tetrahιdrocannabιnol, cannabinol and cannabidiol
4 - Uso según una cualquiera de las reivindicaciones precedentes, en el que el cannabinoide natural es Δ9-tetrahιdrocannabιnol (THC)4 - Use according to any one of the preceding claims, wherein the natural cannabinoid is Δ 9 -tetrahιdrocannabιnol (THC)
5 - Uso según las reivindicaciones 1 y 2, en el que los cannabinoides sintéticos se seleccionan del grupo formado por WIN-55,212-2, HU-210, CP-5 - Use according to claims 1 and 2, wherein the synthetic cannabinoids are selected from the group consisting of WIN-55,212-2, HU-210, CP-
55,940 y CP-50,556 (levonantradol)55,940 and CP-50,556 (levonantradol)
6 - Uso según las reivindicaciones 1 y 5, en el que el cannabinoide sintético es WIN-55,212-2 7 - Medicamento para el tratamiento, en mamíferos incluyendo seres humanos, de tumores cerebrales seleccionados del grupo que comprende glioblastomas, meduloepite omas, meduloblastomas, neuroblastomas germinomas, carcinomas embrionarios, astrocitomas, astroblastomas ependimomas, oligodendroghomas, carcinomas plexales, neuroepitehomas pineoblastomas, epandimobiastomas, tumores neuroectodérmicos meningiomas malignos, condrosarcomas, sarcomatosomas meningeales melanomas malignos y schwanomas malignos, caracterizado porque comprende, como principio activo, un cannabinoide natural o sintético, y un excipiente farmacéuticamente aceptable6 - Use according to claims 1 and 5, wherein the synthetic cannabinoid is WIN-55,212-2 7 - Medication for the treatment, in mammals including humans, of brain tumors selected from the group comprising glioblastomas, meduloepite omas, medulloblastomas, neuroblastomas germinomas, embryonic carcinomas, astrocytomas, ependymoma astroblastomas, oligodendroghomas, plexal carcinomas, neuroepitehomas, pineoblastbiaomas, pineoblastbiaomas tumors neuroectodermal malignant meningiomas, chondrosarcomas, meningeal sarcomatosomes, malignant melanomas and malignant schwannomas, characterized in that it comprises, as an active ingredient, a natural or synthetic cannabinoid, and a pharmaceutically acceptable excipient
8 - Medicamento según la reivindicación 7, en el que el cannabinoide natural se selecciona del grupo formado por Δ9-tetrahιdrocannabιnol (THC) Δ8-tetrahιdrocannabιnol, cannabinol y cannabidiol8 - A medicament according to claim 7, wherein the natural cannabinoid is selected from the group consisting of Δ 9 -tetrahιdrocannabιnol (THC) Δ 8 -tetrahιdrocannabιnol, cannabinol and cannabidiol
9 - Medicamento según la reivindicación 7, en el que el cannabinotde sintético se selecciona del grupo formado por WIN-55,212-2, HU-210, CP- 55,940 y CP-50,556 (levonantradol)9 - A medicament according to claim 7, wherein the synthetic cannabinotde is selected from the group consisting of WIN-55,212-2, HU-210, CP-55,940 and CP-50,556 (levonantradol)
10 - Medicamento según una cualquiera de las reivindicaciones 7 a 9 en el que el excipiente es uno adecuado para administración intratumoral (intracraneal) o sistémica, tal como oral, intravenosa o intrapentoneal10 - A medicament according to any one of claims 7 to 9 wherein the excipient is one suitable for intratumoral (intracranial) or systemic administration, such as oral, intravenous or intrapentoneal
11 - Medicamento según la reivindicación 10, en el que el excipiente para administración intratumoral es una solución salina tamponada con fosfato (PBS) suplementada con albúmina de suero de bovino (BSA) deslipizada y dializada11 - A medicament according to claim 10, wherein the excipient for intratumoral administration is a phosphate buffered saline (PBS) supplemented with bovine serum albumin (BSA) slipped and dialyzed
12 - Medicamento según una cualquiera de las reivindicaciones 10 a 11 , en el que la concentración del cannabinoide en el líquido de administración intratumoral es de 10 a 10000 μg/ml para el cannabinoide natural y de 1 a 1000 μg/ml para el cannabinoide sintético12 - A medicament according to any one of claims 10 to 11, wherein the concentration of the cannabinoid in the liquid of intratumoral administration is 10 to 10,000 μg / ml for the natural cannabinoid and 1 to 1000 μg / ml for the synthetic cannabinoid
13 - Procedimiento para el tratamiento terapéutico, en mamíferos incluyendo seres humanos, de tumores cerebrales seleccionados del grupo que comprende glioblastomas, meduloepitehomas, meduloblastomas, neuroblastomas, germinomas, carcinomas embrionarios, astrocitomas, astroblastomas, ependimomas, oligodendrogliomas, carcinomas plexales, neuroepiteliomas, pineoblastomas, epandimoblastomas tumores neuroectodérmicos, meningiomas malignos, condrosarcomas, sarcomatosomas meningeales, melanomas malignos y schwanomas malignos, caracterizado porque comprende administrar, al animal afectado por uno de dichos tumores, una cantidad terapéuticamente eficaz de un medicamento como el definido en una cualquiera de las reivindicaciones 7 a 1213 - Procedure for therapeutic treatment, in mammals including humans, of brain tumors selected from the group comprising glioblastomas, meduloepitehomas, medulloblastomas, neuroblastomas, germinomas, embryonic carcinomas, astrocytomas, astroblastomas, ependymomas, oligodendrogliomas, plexal carcinomas, pineappleoomas epandimoblastomas neuroectodermal tumors, malignant meningiomas, chondrosarcomas, meningeal sarcomatosomes, malignant melanomas and malignant schwannomas, characterized in that it comprises administering, to the animal affected by one of said tumors, a therapeutically effective amount of a medicament as defined in any one of claims 7 to 12
14 - Procedimiento según la reivindicación 13, caracterizado porque la administración se efectúa por vía intratumoral14 - Method according to claim 13, characterized in that the administration is carried out intratumorally
15 - Procedimiento según la reivindicación 14, caracterizado porque la cantidad administrada de cannabinoide (principio activo) varia de 100 a 50000 μg para los cannabinoides naturales y de 10 a 5000 μg para los cannabinoides sintéticos 15 - Method according to claim 14, characterized in that the administered amount of cannabinoid (active ingredient) varies from 100 to 50,000 μg for natural cannabinoids and from 10 to 5000 μg for synthetic cannabinoids
PCT/ES2000/000450 2000-02-11 2000-11-22 Therapy with cannabinoids in the treatment of cerebral tumor WO2001058445A1 (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
EP00976087A EP1177790B1 (en) 2000-02-11 2000-11-22 Therapy with cannabinoids in the treatment of cerebral tumor
AT00976087T ATE295162T1 (en) 2000-02-11 2000-11-22 THERAPEUTIC USE OF CANNABINOIDS FOR BRAIN TUMOR TREATMENT
AU13979/01A AU1397901A (en) 2000-02-11 2000-11-22 Therapy with cannabinoids in the treatment of cerebral tumor
DE60020111T DE60020111T2 (en) 2000-02-11 2000-11-22 THERAPEUTIC APPLICATION OF CANNABINOIDES FOR BRAIN TUMOR TREATMENT
ES00976087T ES2241670T3 (en) 2000-02-11 2000-11-22 CANNABINOID THERAPY FOR THE TREATMENT OF CEREBRAL TUMORS.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ESP200000323 2000-02-11
ES200000323U ES1045342Y (en) 2000-02-11 2000-02-11 ROTATING EXHIBITOR FOR POSTCARDS, PHOTOS AND SIMILAR.

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US09958960 A-371-Of-International 2001-11-27
US10/647,739 Division US20040039048A1 (en) 2000-02-11 2003-08-25 Therapy with cannabinoid compounds for the treatment of brain tumors

Publications (1)

Publication Number Publication Date
WO2001058445A1 true WO2001058445A1 (en) 2001-08-16

Family

ID=8492271

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/ES2000/000450 WO2001058445A1 (en) 2000-02-11 2000-11-22 Therapy with cannabinoids in the treatment of cerebral tumor

Country Status (2)

Country Link
ES (1) ES1045342Y (en)
WO (1) WO2001058445A1 (en)

Cited By (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1307186A1 (en) * 2000-05-17 2003-05-07 Atlantic Technology Ventures, Inc. Methods for decreasing cell proliferation based on (3r, 4r)-delta8-tetrahydrocannabinol-11-oic acids
GB2478074A (en) * 2008-06-04 2011-08-24 Gw Pharma Ltd THC and CBD for use in the treatment of tumours
GB2478072A (en) * 2008-06-04 2011-08-24 Gw Pharma Ltd THC and CBD for use in the treatment of brain tumours
US8632825B2 (en) 2008-06-04 2014-01-21 Gw Pharma Limited Anti-tumoural effects of cannabinoid combinations
US8790719B2 (en) 2010-03-12 2014-07-29 Gw Pharma Limited Phytocannabinoids in the treatment of cancer
US9084771B2 (en) 2007-05-17 2015-07-21 Sutter West Bay Hospitals Methods and compositions for treating cancer
US10758514B2 (en) 2013-06-19 2020-09-01 Gw Pharma Limited Use of tetrahydrocannabinol and/or cannabidiol for increasing radiosensitivity in the treatment of a brain tumour

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BAEK SEUNG-HWA, ET AL.: "Boron trifluoride etherate on silica-A modified Lewis acid reagent (VII). Antitumor activity of cannabigerol against human oral epitheloid carcinoma cells", ARCHIVES OF PHARMACOL. RES., vol. 21, no. 3, June 1998 (1998-06-01), SEOUL, pages 353 - 356, XP000957635 *
BAEK SEUNG-HWA, ET AL.: "Synthesis and antitumor activity of cannabigerol", ARCHIVES OF PHARMACOL. RES., vol. 19, no. 3, 1996, SEOUL, pages 228 - 230, XP000979791 *
BRAUDE M.C. & STEPHEN SZARA: "Pharmacology of marihuana", 1976, RAVEN PRESS, NEW YORK, NY, USA, XP000957683 *
HARRIS L.S. ET AL.: "Retardation of tumor growth by delta-9 tetrahydrocannabinol", THE PHARMACOLOGIST, vol. 16, no. 2, 1974, pages 259, XP000957658 *
RUIZ L. ET AL.: "Delta-9 tetrahydrocannabinol induces apoptosis in human prostate PC-3 cells via a receptor-independent mechanism", FEBS LETT., vol. 458, no. 3, 1999, pages 400 - 404, XP000981226 *

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1307188A1 (en) * 2000-05-17 2003-05-07 Atlantic Technology Ventures, Inc. Cannabinoid drugs
EP1307188A4 (en) * 2000-05-17 2005-07-06 Atlantic Technology Ventures I Cannabinoid drugs
EP1307186A4 (en) * 2000-05-17 2005-10-19 Atlantic Technology Ventures I Methods for decreasing cell proliferation based on (3r, 4r)-delta8-tetrahydrocannabinol-11-oic acids
EP1307186A1 (en) * 2000-05-17 2003-05-07 Atlantic Technology Ventures, Inc. Methods for decreasing cell proliferation based on (3r, 4r)-delta8-tetrahydrocannabinol-11-oic acids
US9084771B2 (en) 2007-05-17 2015-07-21 Sutter West Bay Hospitals Methods and compositions for treating cancer
US11344527B2 (en) 2007-05-17 2022-05-31 Sutter Bay Hospitals Methods and compositions for treating cancer
US11260043B2 (en) 2007-05-17 2022-03-01 Sutter Bay Hospitals Methods and compositions for treating cancer
GB2478074B (en) * 2008-06-04 2012-12-26 Gw Pharma Ltd Anti-tumoural effects of cannabinoid combinations
US8632825B2 (en) 2008-06-04 2014-01-21 Gw Pharma Limited Anti-tumoural effects of cannabinoid combinations
GB2478072B (en) * 2008-06-04 2012-12-26 Gw Pharma Ltd Anti-tumoural effects of cannabinoid combinations
GB2478072A (en) * 2008-06-04 2011-08-24 Gw Pharma Ltd THC and CBD for use in the treatment of brain tumours
GB2478074A (en) * 2008-06-04 2011-08-24 Gw Pharma Ltd THC and CBD for use in the treatment of tumours
US8790719B2 (en) 2010-03-12 2014-07-29 Gw Pharma Limited Phytocannabinoids in the treatment of cancer
US9675654B2 (en) 2010-03-12 2017-06-13 Gw Pharma Limited Phytocannabinoids in the treatment of cancer
US10758514B2 (en) 2013-06-19 2020-09-01 Gw Pharma Limited Use of tetrahydrocannabinol and/or cannabidiol for increasing radiosensitivity in the treatment of a brain tumour

Also Published As

Publication number Publication date
ES1045342Y (en) 2001-02-16
ES1045342U (en) 2000-08-16

Similar Documents

Publication Publication Date Title
Wang et al. Salidroside attenuates neuroinflammation and improves functional recovery after spinal cord injury through microglia polarization regulation
Liu et al. RIP3 deficiency protects against traumatic brain injury (TBI) through suppressing oxidative stress, inflammation and apoptosis: dependent on AMPK pathway
Zhang et al. Modulation of brain cation-Cl− cotransport via the SPAK kinase inhibitor ZT-1a
Sang et al. NGF and PI3K/Akt signaling participate in the ventral motor neuronal protection of curcumin in sciatic nerve injury rat models
De Petrocellis et al. Non‐THC cannabinoids inhibit prostate carcinoma growth in vitro and in vivo: pro‐apoptotic effects and underlying mechanisms
Chen et al. TGF-β1 neuroprotection via inhibition of microglial activation in a rat model of Parkinson’s disease
Chen et al. Activating transcription factor 4 mediates hyperglycaemia-induced endothelial inflammation and retinal vascular leakage through activation of STAT3 in a mouse model of type 1 diabetes
Munson et al. Anti-invasive adjuvant therapy with imipramine blue enhances chemotherapeutic efficacy against glioma
Huang et al. Capsaicin protects cortical neurons against ischemia/reperfusion injury via down-regulating NMDA receptors
Lombard et al. CB2 cannabinoid receptor agonist, JWH-015, triggers apoptosis in immune cells: potential role for CB2-selective ligands as immunosuppressive agents
Velasco et al. Cannabinoids and gliomas
Abdelfadil et al. Thymoquinone induces apoptosis in oral cancer cells through p38β inhibition
Rummel et al. Parthenolide attenuates LPS-induced fever, circulating cytokines and markers of brain inflammation in rats
Guan et al. Puerarin ameliorates retinal ganglion cell damage induced by retinal ischemia/reperfusion through inhibiting the activation of TLR4/NLRP3 inflammasome
Elsherbiny et al. ABT-702, an adenosine kinase inhibitor, attenuates inflammation in diabetic retinopathy
US20040039048A1 (en) Therapy with cannabinoid compounds for the treatment of brain tumors
Peng et al. Oxyfadichalcone C inhibits melanoma A375 cell proliferation and metastasis via suppressing PI3K/Akt and MAPK/ERK pathways
Ko et al. Polydeoxyribonucleotide ameliorates lipopolysaccharide-induced acute lung injury via modulation of the MAPK/NF-κB signaling pathway in rats
Chen et al. Dl-3-n-butylphthalide exerts dopaminergic neuroprotection through inhibition of neuroinflammation
Sarmah et al. Sirtuin-1-mediated NF-κB pathway modulation to mitigate inflammasome signaling and cellular apoptosis is one of the neuroprotective effects of intra-arterial mesenchymal stem cell therapy following ischemic stroke
Wei et al. 17β-estradiol ameliorates oxidative stress and blue light-emitting diode-induced retinal degeneration by decreasing apoptosis and enhancing autophagy
Ranieri et al. Endocannabinoid system in neurological disorders
Xue et al. Phosphorylation at S548 as a functional switch of sterile alpha and TIR motif-containing 1 in cerebral ischemia/reperfusion injury in rats
Li et al. Effect of photodynamic therapy combined with torasemide on the expression of matrix metalloproteinase 2 and sodium-potassium-chloride cotransporter 1 in rat peritumoral edema and glioma
WO2001058445A1 (en) Therapy with cannabinoids in the treatment of cerebral tumor

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU BR CA CN JP MX NO NZ SG US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR

121 Ep: the epo has been informed by wipo that ep was designated in this application
WWE Wipo information: entry into national phase

Ref document number: 2000976087

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 09958960

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 2000976087

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 2000976087

Country of ref document: EP