CBD

Left Top

Introduction

CBD is the second most common cannabinoid from the plant of cannabis. In contrast to THC, CBD is not psychoactive and the reduced side effects associated to its administration makes it as important as THC regarding its therapeutic properties. CBD is already used to treat epilepsy and to reduce symptoms of Multiple Sclerosis. Also, CBD can counteract the psychoactive effects of THC, making THC more easy to tolerate when high doses are required. CBD also has a huge therapeutic potential in a wide range of diseases due to its neuroprotective and immunomodulatory properties.

Chemical Name

Cannabidiol

IUPHAR entry

Wikipedia Entry

Tags

Bottom

Literature Discussion

Alzheimer's

Mesenchymal stem cells may form a promising new method to regenerate nerve cells in degenerative diseases like Alzheimer’s. Treatment of mesenchymal stem cells with CBD prevented hallmarks of Alzheimer’s like β Amyloid plaques and neurofibrillary tangles (Libro et al., 2016) suggesting therapeutic potential. This effect was mediated by TRPV1 and the PI3K/Akt/GSK3β pathway. In rats, 10mg/kg CBD prevented reactive astrogliosis and neuronal degeneration after β Amyloid injection. This effect was mediated by PPARΥ (Esposito et al., 2011). In Alzheimer-prone AβPP/PS1 transgenic mice, application of THC in combination with CBD. In early symptomatic stages, THC/CBD could revert both β Amyloid plaque formation and memory impairment. In late stages, plaque formation could not be prevented but memory could still be preserved by administration of THC and CBD (Aso et al., 2016). GPR3 has been previously linked to Alzheimer's disease. This receptor share about 35% of amino acid sequence with CB1 and CB2. CBD acts as an inverse agonist for this receptor as it has been shown in a β-arrestin2 recruitment assay (Laun & Song, 2017).

anxiety

In healthy volunteers, oral THC (10mg), but not CBD (600 mg) produced anxiety, dysphoria, positive psychotic symptoms, physical and mental sedation and subjective intoxication (Martin-Santos et al., 2012). In Spontaneously Hypertensive rats, intraperitoneal doses of CBD as low as 1 mg/kg could prevent anxiety but not the development of positive psychotic symptoms (Almeida et al., 2013), suggesting an inter-species difference with humans or a differential dose-effect with the study above. In rats, intraperitoneal CBD (10 mg/kg) improved extinction of fearful memories (Song et al., 2016). Similarly, THC injected intraperitoneally at 0.3-10 mg/kg disrupted consolidation of fearful memories but produced positive psychotic effects. Co-application of CBD at 10/1 (CBD/THC) preserved anxiolysis while abolishing psychotic symptoms (Stern et al., 2015). Direct injection of CBD into the substantia nigra suppressed anxiety but not innate fear-induced antinociception (da Silva et al., 2015).

cancer

breast cancer
In cell culture 5 μM CBD killed (MBA-MB-231) breast cancer cells but not normal cells through cell-autonomous apoptosis and autophagy. This effect was independent of CB1, CB2 and TRPV1 (Shrivastava et al., 2011). Similarly, 3 μM CBD killed 50% of MDA-MB231 breast cancer cells in culture (Ward et al., 2014). In addition to blocking proliferation, CBD also inhibits breast cancer cell invasion and metastasis(McAllister et al., 2011; Murase et al., 2014).

glioblastoma
In glioma xenografts 7.5 mg/kg/day CBD decreased tumor growth by about 20%. 7.5 mg/kg/day THC produced similar results and combined application of CBD and THC reduced tumor growth by approximately 50% suggesting synergy between both pathways (Torres et al., 2011). In mice, a THC/CBD mixture can inhibit tumor growth by blocking angiogenesis and cell proliferation (Hernán Pérez de la Ossa et al., 2013). Similarly, CBD was found to inhibit glioma stem-like cell proliferation and thus tumor initiation through a TRPV2-dependent autophagic process (Nabissi et al., 2015). In mice, 15 mg/kg CBD significantly reduced glioblastoma growth (Soroceanu et al., 2013). In mice a combination of CBD and THC was found to work synergistically with radiation therapy to reduce tumor size (Scott et al., 2014). In human glioblastoma cell lines CBD reduces cancer cell viability and proliferation (Deng et al., 2016). CBD modulates Id-1 gene and targets receptors CB1, CB2, TRPV-1 and TRPV-2 (Solinas et al., 2013). Importantly, CBD improves effectiveness of THC and is also effective in glioblastoma THC-resistant cells (Marcu et al., 2010; Solinas et al., 2013). CBD also improves effectiveness of other anti cancer drugs as temozolomide, carmustine or dodorubicin through TRPV-2 receptor (Nabissi et al., 2013).

leukemia

CBD showed cell activation modulation, but the mechanism failed to show G protein-coupled receptor pathways, suggesting unknown internal mechanisms (Giudice et al., 2007).

Lung Cancer
In three different cancer cell lines (A549, H358 and H460) CBD dose dependently (1nM-3μM) increased ICAM-1 and TIMP-1 through TRPV1. In mice carrying human Lung Cancer xenografts, CBD increased ICAM-1 and TIMP-1 2.6-3.0-fold, inhibiting Lung Cancer cell invasion and metastasis (Ramer et al., 2012). In cancer cell lines (A549 and H460) and human metastatic Lung Cancer cells CBD as well as THC promote ICAM-mediated Lymphokine-Activated Killer cell adhesion and cancer cell lysis (Haustein et al., 2014). In cancer cell lines (A549 and H460) and human metastatic Lung Cancer cells CBD induced apoptosis via COX-2 and pparγ. In A549-xenografted mice CBD caused tumor regression (Ramer et al., 2013).

Melanoma
In mice, xenografted melanomas (A375 and SK-MEL-28 cells) a mixture of 1.5μM THC and 1.5 μM CBD in extract essentially reduced cancer cell viability and tumor growth to zero. THC alone (3 μM) reduced cell viability to approximately 50% and temozolomide left viability unchanged (Armstrong et al., 2015).

Myeloma
In cancer cells derived from Multiple Myeloma patients 20 μM CBD alone, and in combination with bortezomib strongly inhibited cell division/tumor growth. This effect at least partially through TRPV2 (Morelli et al., 2014). The pro-apoptotic effect of CBD works synergistically with THC and carfilzomib (Nabissi et al., 2016).

Neuroblastoma
In neuroblastoma cell lines ±5 μg/ml CBD reduced cancer cell viability by ±50% whereas ±15 μg/ml THC was required for the same effect. In mice CBD was also more effective than THC and 20 mg/kg CBD reduced tumor volume by about 60% (Fisher et al., 2016).

Prostate cancer
In cell culture 1-10 μM CBD (like THC) induced apoptosis of Prostate cancer cells /xenograft tumors (LNCaP, 22RV1, DU-145 and PC-3)(De Petrocellis et al., 2013).

Lung Cancer

In three different cancer cell lines (A549, H358 and H460) CBD dose dependently (1nM-3μM) increased ICAM-1 and TIMP-1 through TRPV1. In mice carrying human lung cancer xenografts, CBD increased ICAM-1 and TIMP-1 2.6-3.0-fold, inhibiting lung cancer cell invasion and metastasis (Ramer et al., 2012). In cancer cell lines (A549 and H460) and human metastatic lung cancer cells CBD as well as THC promote ICAM-mediated Lymphokine-Activated Killer cell adhesion and cancer cell lysis (Haustein et al., 2014). In cancer cell lines (A549 and H460) and human metastatic lung cancer cells CBD induced apoptosis via COX-2 and PPARγ. In A549-xenografted mice CBD caused tumor regression (Ramer et al., 2013).

COPD

In the ovalbumin rat model of asthma, 5 mg/kg i.p. CBD decreased the levels of cytokines IL-4 (T helper cell differentiation and IgE production), IL-5 (eosinophil maturation), IL-6 (T cell proliferation), IL-13 (mucus hypersecretion) and TNFα (asthma mediator) but not IL-10, suggesting therapeutic potential in suppressing lung inflammation (Vuolo et al., 2015). In the LPS mouse model of lung inflammation, 20 mg/kg i.p. CBD induced pparγ-dependent G-CSF secretion from mast cells and subsequent myeloid-derived suppressor cell mobilization thus suppressing inflammation (Hegde et al., 2015).
In another study, CBD was partially effective in suppressing coughing (Makwana et al., 2015). 

Functional Gastro-Intestinal Disorders

Apart from THC, (relatively) non-psychotropic cannabinoids such as THCVCBD and CBG were found to have anti-inflammatory effects in experimental intestinal inflammation (Alhouayek and Muccioli, 2012). In the TNBS mouse model of colitis, 5 mg/kg CBD i.p. twice daily for three days attenuated colitis and promoted endothelial and epithelial wound healing (Krohn et al., 2016). In the DNBS mouse model of colitis, both oral and i.p. CBD decreased tissue damage and intestinal hypermotility. CBD in extract was more effective than pure CBD, suggesting a significant entourage effect (Pagano et al., 2016). In the LPS mouse model of colitis, 10 mg/kg i.p. CBD decreased reactive gliosis, mast cell and macrophage recruitment, TNFα expression and intestinal apoptosis. In ulcerative colitis patient rectal biopsies also reduced reactive gliosis, at least partially through pparγ (De Filippis et al., 2011). In mice, CBD seems to inhibit intestinal motility in both CB1 dependent and independent ways (Fride et al., 2005).

depression

In rats, 10-60 nMol injected CBD straight into the prefrontal cortex reduced depressive behavior, possibly through 5-HT1A (Sartim et al., 2016). Similarly, CBD (200 mg/kg) and CBC (20 mg/kg) displayed significant anti-depressant activity. The anti-depressant effects of the different cannabinoids display different dose-dependency and are probably reached through different receptors (El-Alfy et al., 2010). 

Diabetes

In non-obese diabetic mice, CBD treatment reduces Diabetes incidence from 86% to 30%. CBD reduced pro-inflammatory IFNγ, TNFα and Th1-associated cytokines while increasing anti-inflammatory Th2-associated cytokines, resulting in reduced insulitis (Weiss et al., 2006, 2007). In isolated arteries from Zucker diabetic fatty rats 10 μM CBD enhanced maximal vasorelaxation via enhanced COX-1/2 signalling (Wheal et al., 2014).

Eczema

A comparative study into the topical anti-inflammatory activity of cannabinoids (on croton oil-inflamed skin in mice) showed that Δ8THC, Δ9THC and THCV are about half as effective in reducing inflammation as Indometacin (a commonly used Non-steroid anti-inflammatory drug), but approximately 5 times more effective than CBCV and CBDCBC and CBDV had no appreciable anti-inflammatory activity (Tubaro et al., 2010)

epilepsy

Cultured HEK293 cells carrying human epilepsy-associated mutations in Nav1.6 display increased resurgent sodium currents and increased excitability. 1 μM CBD reduced resurgent sodium currents and increased the refractory period. In cultured mouse striatal neurons CBD reduced overall action potential firing suggesting therapeutic potential (Patel et al., 2016). In heterologous cells (HEK293), THC and CBD were found to inhibit T-type calcium channels with an IC50 of approximately 1μM (Ross et al., 2008). THC-mediated inhibition was frequency dependent where CBD-mediated inhibition was not. As T-type calcium channels function in thalamus-mediated synchronization of brain regions and are implicated in various types of epilepsy, THC and CBD are likely to suppress seizure generation. In human neuroblastoma cells (SH-SY5Y) and mouse cortical neurons CBD and CBG both blocked sodium channels Nav1.1, 1.2 and 1.5 (Hill et al., 2014). Interestingly, CBD but not CBG protected against pentyleneterzole (PTZ)-induced seizures in rat, suggesting that the anti-convulsant effect of CBD is not just through blocking sodium channels.

Graft rejection

In mice with experimental autoimmune myocarditis, 10 mg/kg/day i.p. CBD reduced the expression of pro-inflammatory IL-6, IL1β and IFNγ and consequent (T-cell-mediated) inflammation, oxidative stress and tissue fibrosis (Lee et al., 2016).

Hypoxic-Ischemic Encelphalopathy

CBD showed neuroprotective effects with functional and behavioral recovery in hypoxia-ischemic animal models (Alvarez et al., 2008; Lafuente et al., 2011). CBD increased neuronal and astrocyte survival and reduced brain damage and reactive astrogliosis (Hayakawa et al., 2009; Schiavon et al., 2014). CBD mechanisms would involve the modulation of excitotoxicity, oxidative stress and inflammation through CB2, 5HT1A, Adenosine A2A and PPAR-γ receptors (Castillo et al., 2010; Hind et al., 2015; Pazos et al., 2012, 2013). CBD shows neuroprotective effects in a rat model of HI in a wider time window than any other neuroprotective treatment for this pathology (Mohammed, Ceprián, Jimenez, Pazos, & Martínez-Orgado, 2016). Similar to previous studies in HI, rat models of Arterial Isquemic stroke showed improved neurobehavioral functioning after CBD treatment, including modulation of astrogliosos and microglial proliferation while showed reduced excitotocicity, neuronal loss and apoptosis (Ceprián et al., 2016). CBD, combined with hypotermia (typical treatment for HI), improves effects of exitotoxicity, inflammation, oxidative stress and cell damage compared to the treatment of hypothermia or cannabidiol alone (Lafuente et al., 2016).

Inflammation

In rats 2.5 mg/kg/day for 14 days selectively increased the number of Natural Killer cells and Natural Killer T-cells. At 5 mg/kg/day, lymphopenia was induced by reducing B-, T- and T helper cells but not NK or NTK cells. This suggests that CBD can selectively boost innate immunity and suppress the acquired immune system (Ignatowska-Jankowska et al., 2009). In the LPS mouse model of acute lung inflammation 20 and 80 mg/kg i.p. reduced lung resistance and elastance, leucocyte recruitment and the expression of TNF, IL-6, MCP-1 and MIP-2 resulting in potent suppression of inflammation (Ribeiro et al., 2014). In cultured mouse T-cells, 4-8 μM CBD induced apoptosis of CD4+ and CD8+ by increasing ROS and caspase3/8 (Lee et al., 2008).

insomnia

CBD would act as an inhibitor of Anandamide uptake through TPRV1 receptor, suggesting a role in sleep (Bisogno et al., 2001; Mechoulam et al., 1997). The effects of CBD in sleep appear to be related to a reduction of Anxiety-induced REM sleep instead of sleep regulation processes (Hsiao et al., 2012). CBD would eliminate those residual effects but subjects reported sleepiness after CBD administration (Nicholson et al., 2004).

Ischemia

In newborn piglets with brain hypoxia-ischemia, hypothermia and 1mg/kg i.v. CBD both reduced the number of necrotic neurons. The effects were additive suggesting complementary pathways (Lafuente et al., 2016). In mice, hypoxic-ischemic brain damage were reduced (90% less apoptosis, 50% less astrocyte damage) when 1 mg/kg CBD was administered subcutaneously within 15 minutes, 1, 3, 6, 12 or 18 hours after the insult. CBD application 24 hours after the insult was ineffective (Mohammed et al., 2016).

Liver fibrosis

In Human Hepatocyte Line-5 cells, 10 μM CBD or 5 μM THCV reduced intracellular lipid levels. In obese mice 3 mg/kg oral CBD reduced the level of liver triglycerides suggestive of increased lipolysis (Silvestri et al., 2015).

Malaria

In mice infected with Plasmodium Berghei ANKA 30 mg/kg/day i.p. CBD decreased pro-inflammatory IL-6 and TNFα, prevented memory deficits and anxiety and increased survival suggesting a neuroprotective effect (Campos et al., 2015).

Morphine Interaction

CBD shows a synergistic interaction with Morphine only in the acetic acid-stimulated stretching assay (Neelakantan et al., 2015).

Multiple Sclerosis

In the rat MOG35-55 model of experimental autoimmune encephalitis / Multiple Sclerosis 5 μM CBD upregulated CD69 and Lag3 on CD4+CD25- T-cells and anti-inflammatory markers IL-10 and STAT5 promoting T-cell anergy and cell-cycle arrest. CBD also reduced MHC2, CD25 and CD69 on CD19+ cells reducing their antigen-presenting and pro-inflammatory potential (Kozela et al., 2015). Gene profiling showed that CBD generally suppresses pro-inflammatory genes, T-cell proliferation and potentially T-cell memory while enhancing anti-inflammatory genes (affected genes listed in article) (Kozela et al., 2016).
In MOG35-55 mice topical treatment with a 1% CBD cream was neuroprotective, reducing release of CD4 and CD8 T-cells and the expression of pro-inflammatory cytokines and inflammatory markers p-selectin, IL-10, GFAP, TGF-β, IFN-γ, nitrotyrosin, iNOS, PARP and caspase-3 (Giacoppo et al., 2015a). In MOG35-55 mice, CBD i.p. also reduced apoptosis and neurodegeneration (Giacoppo et al., 2015b).
In MOG treated mice 5 mg/kg i.p. CBD or PEA reduced neurobehavioral deficits, inflammation, demyelination, axonal damage and expression of pro-inflammatory cytokines. Co-administration of CBD and PEA reduced the therapeutic potential suggesting an antagonistic interaction (Rahimi et al., 2015). In a mouse model of MS (Theiler's murine encephalomyelitis), Sativex (50/50% THC/CBD oromucoso spray was compared with CBD-enriched or THC-enriched cannabis extract. Motor deterioration and inflammation (astrogliosis) were equally reduced by Sativex and CBD-enriched extract but THC-enriched extract was less effective. The effects of CBD were PPARγ-mediated whereas THC signaling was CB1/2 dependent (Feliú et al., 2015).

obesity

THCV and CBD reduce the amount of circulating lipids and enable weight-loss (Silvestri et al., 2015). THCV induced hypophagia and reduction in body weight at low doses (from 3mg/kg), suggesting a possible treatment for Obesityand metabolic syndrome. THC combination with THCV would delete these effects, but they are rescued by combining them with CBD (Riedel et al., 2009; Silvestri et al., 2015; Wargent et al., 2013)

Obsessive Compulsive Disorder

In rats oral application of 120 mg/kg CBD inhibited obsessive-compulsive behavior (1. Dose is incredibly high. 2. Article also shows preferential oral absorption for CBD and CBDV and i.p. absorption for THCV and CBG) (Deiana et al., 2012). 

OCD

Several studies have pointed out a correlation between the occurrence of OCD and cannabis use (De Alwis et al., 2014; Bidwell et al., 2014; Loflin et al., 2014). However, whether cannabis use precipitates OCD or cannabis is used to self-medicate against the symptoms of OCD remains to be elucidated.

pain

In mice 2 mg/kg i.p. (like THC and gabapentin) CBD reduced, but did not prevent, neuropathic pain induced by cis-platin (Harris et al., 2016).

Parkinson’s disease

In the 6-OH-DOPA mouse model of Parkinson’s daily administration of either THC or CBD provided lasting neuroprotection (Lastres-Becker et al., 2005). In a rat model of Parkinson’s Disease, THCV and CBD were neuroprotective in a CB2-independent way (García et al., 2011). In cultured midbrain neurons, CBDTHCA and THC had anti-oxidative properties. Moreover, THCA and THC were shown to be neuroprotective (Moldzio et al., 2012). GPR6 has been previously linked to Parkinson's disease. This receptor share about 35% of amino acid sequence with CB1 and CB2. CBD acts as an inverse agonist for this receptor as it has been shown in a β-arrestin2 recruitment assay (Laun & Song, 2017).

Psoriasis

In cultured hyper-proliferating human keratinocytes CBD (as well as THC, CBN and CBG) inhibited cell proliferation in a dose-dependent, CB1/2-independent manner suggesting therapeutic potential (Wilkinson and Williamson, 2007).

psychosis and schizophrenia

Anti-psychotic drugs often antagonize dopamine D2 receptors. In apoMorphine- or amphetamine treated rats 15-60 mg/kg CBD reduced stereotyped behavior and hyperlocomotion similar to haloperidol in a dose-dependent manner. At 120-480 mg/kg (!!!) CBD increased prolactin levels (like haloperidol) but did not induce catalepsy (unlike haloperidol) suggesting CBD has anti-psychotic potential at high doses (reviewed in Zuardi et al., 2006). In patients with acute schizophrenia, AEA levels are elevated and inversely correlated with psychotic symptoms, suggesting involvement of the endocannabinoid system in the regulation of psychosis (Giuffrida et al., 2004). At 27.5 μM CBD can AEA inactivation and indirectly increase AEA levels (Bisogno et al., 2001). In a small-scale clinical trial up to 4 doses of 200 mg CBD/day suppressed psychotic symptoms as effectively as amisulpride but with fewer side-effects (Leweke et al., 2012). Similarly, in several case reports CBD doses of up to 1500 mg/day for up to 4 weeks produced similar anti-psychotic effects as observed with classical anti-psychotics but with fewer side-effects (reviewed in Zuardi et al., 2006). It is known that THC can induce psychosis-like effects in healthy volunteers. Using fMRI it was shown that 600 mg (oral capsule) could prevent psychosis-like behavior induced by 10 mg THC (Bhattacharyya et al., 2010).

Rheumatoid arthritis

In mice with collagen-induced arthritis 5 mg/kg/day i.p. or 25 mg/kg/day oral effectively blocked disease progression and suppressed joint damage, lymphocyte proliferation and IFNγ and TNF expression (Malfait et al., 2000).

Stroke

Several clinical trials have tested the therapeutic potential of cannabinoids after stroke. Meta-analysis revealed that both endocannabinoids like AEA, OEA or PEA and plant cannabinoids like THC or CBD can significantly reduce neuronal degeneration after stroke (England et al., 2015).

Tinnitus

Studies with animals suggest that synthetic cannabinoid agonists or mixed administration of phytocannabinoids THC and CBDcould exacerbate induced Tinnitus symptoms (Zheng et al., 2010, 2015).

References:

Alhouayek, M., and Muccioli, G.G. (2012). The endocannabinoid system in inflammatory bowel diseases: from pathophysiology to therapeutic opportunity. Trends Mol. Med. 18, 615–625.

Almeida, V., Levin, R., Peres, F.F., Niigaki, S.T., Calzavara, M.B., Zuardi, A.W., Hallak, J.E., Crippa, J.A., and Abílio, V.C. (2013). Cannabidiol exhibits anxiolytic but not antipsychotic property evaluated in the social interaction test. Prog. Neuropsychopharmacol. Biol. Psychiatry 41, 30–35.

Alvarez, F.J., Lafuente, H., Rey-Santano, M.C., Mielgo, V.E., Gastiasoro, E., Rueda, M., Pertwee, R.G., Castillo, A.I., Romero, J., and Martínez-Orgado, J. (2008). Neuroprotective effects of the nonpsychoactive cannabinoid cannabidiol in hypoxic-ischemic newborn piglets. Pediatr. Res. 64, 653–658.

Armstrong, J.L., Hill, D.S., McKee, C.S., Hernandez-Tiedra, S., Lorente, M., Lopez-Valero, I., Eleni Anagnostou, M., Babatunde, F., Corazzari, M., Redfern, C.P., et al. (2015). Exploiting cannabinoid-Induced Cytotoxic Autophagy to Drive Melanoma Cell Death. J. Invest. Dermatol.

Aso, E., Andrés-Benito, P., and Ferrer, I. (2016). Delineating the Efficacy of a Cannabis-Based Medicine at Advanced Stages of Dementia in a Murine Model. J. Alzheimers Dis. JAD.

Bhattacharyya, S., Morrison, P.D., Fusar-Poli, P., Martin-Santos, R., Borgwardt, S., Winton-Brown, T., Nosarti, C., O’ Carroll, C.M., Seal, M., Allen, P., et al. (2010). Opposite effects of delta-9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 35, 764–774.

Bidwell, L.C., Henry, E.A., Willcutt, E.G., Kinnear, M.K., and Ito, T.A. (2014). Childhood and current ADHD symptom dimensions are associated with more severe cannabis outcomes in college students. Drug Alcohol Depend. 135, 88–94.

Bisogno, T., Hanus, L., De Petrocellis, L., Tchilibon, S., Ponde, D.E., Brandi, I., Moriello, A.S., Davis, J.B., Mechoulam, R., and Di Marzo, V. (2001). Molecular targets for cannabidiol and its synthetic analogues: effect on vanilloid VR1 receptors and on the cellular uptake and enzymatic hydrolysis of Anandamide. Br. J. Pharmacol. 134, 845–852.

Campos, A.C., Brant, F., Miranda, A.S., Machado, F.S., and Teixeira, A.L. (2015). Cannabidiol increases survival and promotes rescue of cognitive function in a murine model of cerebral Malaria. Neuroscience 289, 166–180.

Castillo, A., Tolón, M.R., Fernández-Ruiz, J., Romero, J., and Martinez-Orgado, J. (2010). The neuroprotective effect of cannabidiol in an in vitro model of newborn hypoxic-ischemic brain damage in mice is mediated by CB(2) and adenosine receptors. Neurobiol. Dis. 37, 434–440.

Ceprián, M., Jiménez-Sánchez, L., Vargas, C., Barata, L., Hind, W., & Martínez-Orgado, J. (2016). Cannabidiol reduces brain damage and improves functional recovery in a neonatal rat model of arterial ischemic stroke. Neuropharmacology, 116, 151-159. https://doi.org/10.1016/j.neuropharm.2016.12.017

De Alwis, D., Agrawal, A., Reiersen, A.M., Constantino, J.N., Henders, A., Martin, N.G., and Lynskey, M.T. (2014). ADHD symptoms, autistic traits, and substance use and misuse in adult Australian twins. J. Stud. Alcohol Drugs 75, 211–221.

De Filippis, D., Esposito, G., Cirillo, C., Cipriano, M., De Winter, B.Y., Scuderi, C., Sarnelli, G., Cuomo, R., Steardo, L., De Man, J.G., et al. (2011). Cannabidiol reduces intestinal inflammation through the control of neuroimmune axis. PloS One 6, e28159.

De Petrocellis, L., Ligresti, A., Schiano Moriello, A., Iappelli, M., Verde, R., Stott, C.G., Cristino, L., Orlando, P., and Di Marzo, V. (2013). Non-THC cannabinoids inhibit prostate carcinoma growth in vitro and in vivo: pro-apoptotic effects and underlying mechanisms. Br. J. Pharmacol. 168, 79–102.

Deiana, S., Watanabe, A., Yamasaki, Y., Amada, N., Arthur, M., Fleming, S., Woodcock, H., Dorward, P., Pigliacampo, B., Close, S., et al. (2012). Plasma and brain pharmacokinetic profile of cannabidiol (CBD), cannabidivarine (CBDV), Δ9-tetrahydrocannabivarin (THCV) and cannabigerol (CBG) in rats and mice following oral and intraperitoneal administration and CBD action on obsessive-compulsive behaviour. Psychopharmacology (Berl.) 219, 859–873.

Deng, L., Ng, L., Ozawa, T., and Stella, N. (2016). Quantitative analyses of synergistic responses between cannabidiol and DNA-damaging agents on the proliferation and viability of glioblastoma and neural progenitor cells in culture. J. Pharmacol. Exp. Ther.

England, T.J., Hind, W.H., Rasid, N.A., and O’Sullivan, S.E. (2015). cannabinoids in experimental stroke: a systematic review and meta-analysis. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 35, 348–358.

Esposito, G., Scuderi, C., Valenza, M., Togna, G.I., Latina, V., De Filippis, D., Cipriano, M., Carratù, M.R., Iuvone, T., and Steardo, L. (2011). Cannabidiol reduces Aβ-induced neuroinflammation and promotes hippocampal neurogenesis through pparγ involvement. PloS One 6, e28668.

Feliú, A., Moreno-Martet, M., Mecha, M., Carrillo-Salinas, F.J., de Lago, E., Fernández-Ruiz, J., and Guaza, C. (2015). A sativex-like combination of phytocannabinoids as a disease-modifying therapy in a viral model of Multiple Sclerosis.Br. J. Pharmacol.

Fisher, T., Golan, H., Schiby, G., PriChen, S., Smoum, R., Moshe, I., Peshes-Yaloz, N., Castiel, A., Waldman, D., Gallily, R., et al. (2016). In vitro and in vivo efficacy of non-psychoactive cannabidiol in neuroblastoma. Curr. Oncol. Tor. Ont 23, S15-22.

Fride, E., Ponde, D., Breuer, A., and Hanus, L. (2005). Peripheral, but not central effects of cannabidiol derivatives: mediation by CB(1) and unidentified receptors. Neuropharmacology 48, 1117–1129.

García, C., Palomo-Garo, C., García-Arencibia, M., Ramos, J., Pertwee, R., and Fernández-Ruiz, J. (2011). Symptom-relieving and neuroprotective effects of the phytocannabinoid Δ9-THCV in animal models of Parkinson’s disease. Br. J. Pharmacol. 163, 1495–1506.

Giacoppo, S., Galuppo, M., Pollastro, F., Grassi, G., Bramanti, P., and Mazzon, E. (2015a). A new formulation of cannabidiol in cream shows therapeutic effects in a mouse model of experimental autoimmune encephalomyelitis. Daru J. Fac. Pharm. Tehran Univ. Med. Sci. 23, 48.

Giacoppo, S., Soundara Rajan, T., Galuppo, M., Pollastro, F., Grassi, G., Bramanti, P., and Mazzon, E. (2015b). Purified Cannabidiol, the main non-psychotropic component of Cannabis sativa, alone, counteracts neuronal apoptosis in experimental Multiple Sclerosis. Eur. Rev. Med. Pharmacol. Sci. 19, 4906–4919.

Giudice, E.D., Rinaldi, L., Passarotto, M., Facchinetti, F., D’Arrigo, A., Guiotto, A., Carbonare, M.D., Battistin, L., and Leon, A. (2007). Cannabidiol, unlike synthetic cannabinoids, triggers activation of RBL-2H3 mast cells. J. Leukoc. Biol. 81, 1512–1522.

Giuffrida, A., Leweke, F.M., Gerth, C.W., Schreiber, D., Koethe, D., Faulhaber, J., Klosterkötter, J., and Piomelli, D. (2004). Cerebrospinal Anandamide levels are elevated in acute schizophrenia and are inversely correlated with psychotic symptoms. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 29, 2108–2114.

Harris, H.M., Sufka, K.J., Gul, W., and ElSohly, M.A. (2016). Effects of Delta-9-Tetrahydrocannabinol and Cannabidiol on Cisplatin-Induced Neuropathy in Mice. Planta Med.

Haustein, M., Ramer, R., Linnebacher, M., Manda, K., and Hinz, B. (2014). cannabinoids increase Lung Cancer cell lysis by lymphokine-activated killer cells via upregulation of ICAM-1. Biochem. Pharmacol. 92, 312–325.

Hayakawa, K., Irie, K., Sano, K., Watanabe, T., Higuchi, S., Enoki, M., Nakano, T., Harada, K., Ishikane, S., Ikeda, T., et al. (2009). Therapeutic time window of cannabidiol treatment on delayed ischemic damage via high-mobility group box1-inhibiting mechanism. Biol. Pharm. Bull. 32, 1538–1544.

Hegde, V.L., Singh, U.P., Nagarkatti, P.S., and Nagarkatti, M. (2015). Critical Role of Mast Cells and Peroxisome Proliferator-Activated Receptor γ in the Induction of Myeloid-Derived Suppressor Cells by Marijuana Cannabidiol In Vivo. J. Immunol. Baltim. Md 1950 194, 5211–5222.

Hernán Pérez de la Ossa, D., Lorente, M., Gil-Alegre, M.E., Torres, S., García-Taboada, E., Aberturas, M.D.R., Molpeceres, J., Velasco, G., and Torres-Suárez, A.I. (2013). Local delivery of cannabinoid-loaded microparticles inhibits tumor growth in a murine xenograft model of glioblastoma multiforme. PloS One 8, e54795.

Hill, A.J., Jones, N.A., Smith, I., Hill, C.L., Williams, C.M., Stephens, G.J., and Whalley, B.J. (2014). Voltage-gated sodium (NaV) channel blockade by plant cannabinoids does not confer anticonvulsant effects per se. Neurosci. Lett. 566, 269–274.

Hind, W.H., England, T.J., and O’Sullivan, S.E. (2015). Cannabidiol protects an in vitro model of the blood brain barrier (BBB) from oxygen-glucose deprivation via PPARγ and 5-HT1a. Br. J. Pharmacol.

Hsiao, Y.-T., Yi, P.-L., Li, C.-L., and Chang, F.-C. (2012). Effect of cannabidiol on sleep disruption induced by the repeated combination tests consisting of open field and elevated plus-maze in rats. Neuropharmacology 62, 373–384.

Ignatowska-Jankowska, B., Jankowski, M., Glac, W., and Swiergel, A.H. (2009). Cannabidiol-induced lymphopenia does not involve NKT and NK cells. J. Physiol. Pharmacol. Off. J. Pol. Physiol. Soc. 60 Suppl 3, 99–103.

Kozela, E., Juknat, A., Kaushansky, N., Ben-Nun, A., Coppola, G., and Vogel, Z. (2015). Cannabidiol, a non-psychoactive cannabinoid, leads to EGR2-dependent anergy in activated encephalitogenic T cells. J. Neuroinflammation 12, 52.

Kozela, E., Juknat, A., Gao, F., Kaushansky, N., Coppola, G., and Vogel, Z. (2016). Pathways and gene networks mediating the regulatory effects of cannabidiol, a nonpsychoactive cannabinoid, in autoimmune T cells. J. Neuroinflammation 13, 136.

Krohn, R.M., Parsons, S.A., Fichna, J., Patel, K.D., Yates, R.M., Sharkey, K.A., and Storr, M.A. (2016). Abnormal cannabidiol attenuates experimental colitis in mice, promotes wound healing and inhibits neutrophil recruitment. J. Inflamm. Lond. Engl. 13, 21.

Lafuente, H., Alvarez, F.J., Pazos, M.R., Alvarez, A., Rey-Santano, M.C., Mielgo, V., Murgia-Esteve, X., Hilario, E., and Martinez-Orgado, J. (2011). Cannabidiol Reduces Brain Damage and Improves Functional Recovery After Acute Hypoxia-Ischemia in Newborn Pigs. Pediatr. Res. 70, 272–277.

Lafuente, H., Pazos, M.R., Alvarez, A., Mohammed, N., Santos, M., Arizti, M., Alvarez, F.J., and Martinez-Orgado, J.A. (2016). Effects of Cannabidiol and Hypothermia on Short-Term Brain Damage in New-Born Piglets after Acute Hypoxia-Ischemia. Front. Neurosci. 10, 323.

Lastres-Becker, I., Molina-Holgado, F., Ramos, J.A., Mechoulam, R., and Fernández-Ruiz, J. (2005). cannabinoids provide neuroprotection against 6-hydroxydopamine toxicity in vivo and in vitro: relevance to Parkinson’s disease. Neurobiol. Dis. 19, 96–107.

Laun, A. S., & Song, Z.-H. (2017). GPR3 and GPR6, novel molecular targets for cannabidiol. Biochemical and Biophysical Research Communications, 490(1), 17-21. https://doi.org/10.1016/j.bbrc.2017.05.165

Lee, C.-Y., Wey, S.-P., Liao, M.-H., Hsu, W.-L., Wu, H.-Y., and Jan, T.-R. (2008). A comparative study on cannabidiol-induced apoptosis in murine thymocytes and EL-4 thymoma cells. Int. Immunopharmacol. 8, 732–740.

Lee, W.-S., Erdelyi, K., Matyas, C., Mukhopadhyay, P., Varga, Z.V., Liaudet, L., Haskó, G., Čiháková, D., Mechoulam, R., and Pacher, P. (2016). Cannabidiol limits Tcell-mediated chronic autoimmune myocarditis: implications to autoimmune disorders and organ transplantation. Mol. Med. Camb. Mass.

Leweke, F.M., Piomelli, D., Pahlisch, F., Muhl, D., Gerth, C.W., Hoyer, C., Klosterkötter, J., Hellmich, M., and Koethe, D. (2012). Cannabidiol enhances Anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl. Psychiatry 2, e94.

Libro, R., Diomede, F., Scionti, D., Piattelli, A., Grassi, G., Pollastro, F., Bramanti, P., Mazzon, E., and Trubiani, O. (2016). Cannabidiol Modulates the Expression of Alzheimer’s Disease-Related Genes in Mesenchymal Stem Cells. Int. J. Mol. Sci. 18.

Loflin, M., Earleywine, M., De Leo, J., and Hobkirk, A. (2014). Subtypes of attention deficit-hyperactivity disorder (ADHD) and cannabis use. Subst. Use Misuse 49, 427–434.

Makwana, R., Venkatasamy, R., Spina, D., and Page, C. (2015). The effect of phytocannabinoids on airway hyperresponsiveness, airway inflammation and cough. J. Pharmacol. Exp. Ther.

Malfait, A.M., Gallily, R., Sumariwalla, P.F., Malik, A.S., Andreakos, E., Mechoulam, R., and Feldmann, M. (2000). The nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in murine collagen-induced arthritis. Proc. Natl. Acad. Sci. U. S. A. 97, 9561–9566.

Martin-Santos, R., Crippa, J.A., Batalla, A., Bhattacharyya, S., Atakan, Z., Borgwardt, S., Allen, P., Seal, M., Langohr, K., Farré, M., et al. (2012). Acute effects of a single, oral dose of d9-tetrahydrocannabinol (THC) and cannabidiol (CBD) administration in healthy volunteers. Curr. Pharm. Des. 18, 4966–4979.

McAllister, S.D., Murase, R., Christian, R.T., Lau, D., Zielinski, A.J., Allison, J., Almanza, C., Pakdel, A., Lee, J., Limbad, C., et al. (2011). Pathways mediating the effects of cannabidiol on the reduction of breast cancer cell proliferation, invasion, and metastasis. breast cancer Res. Treat. 129, 37–47.

Mechoulam, R., Fride, E., Hanu, L., Sheskin, T., Bisogno, T., Di Marzo, V., Bayewitch, M., and Vogel, Z. (1997). Anandamide may mediate sleep induction. Nature 389, 25–26.

Mohammed, N., Ceprián, M., Jimenez, L., Pazos, M.R., and Martínez-Orgado, J. (2016). Neuroprotective Effects of Cannabidiol In Hypoxic Ischemic Insult: The Therapeutic Window In Newborn Mice. CNS Neurol. Disord. Drug Targets.

Moldzio, R., Pacher, T., Krewenka, C., Kranner, B., Novak, J., Duvigneau, J.C., and Rausch, W.-D. (2012). Effects of cannabinoids Δ(9)-tetrahydrocannabinol, Δ(9)-tetrahydrocannabinolic acid and cannabidiol in MPP+ affected murine mesencephalic cultures. Phytomedicine Int. J. Phytother. Phytopharm. 19, 819–824.

Morelli, M.B., Offidani, M., Alesiani, F., Discepoli, G., Liberati, S., Olivieri, A., Santoni, M., Santoni, G., Leoni, P., and Nabissi, M. (2014). The effects of cannabidiol and its synergism with bortezomib in multiple myeloma cell lines. A role for transient receptor potential vanilloid type-2. Int. J. cancer 134, 2534–2546.

Murase, R., Kawamura, R., Singer, E., Pakdel, A., Sarma, P., Judkins, J., Elwakeel, E., Dayal, S., Martinez-Martinez, E., Amere, M., et al. (2014). Targeting multiple cannabinoid anti-tumour pathways with a resorcinol derivative leads to inhibition of advanced stages of breast cancer. Br. J. Pharmacol. 171, 4464–4477.

Nabissi, M., Morelli, M.B., Amantini, C., Liberati, S., Santoni, M., Ricci-Vitiani, L., Pallini, R., and Santoni, G. (2015). Cannabidiol stimulates Aml-1a-dependent glial differentiation and inhibits glioma stem-like cells proliferation by inducing autophagy in a TRPV2-dependent manner. Int. J. cancer J. Int. cancer.

Nabissi, M., Morelli, M.B., Offidani, M., Amantini, C., Gentili, S., Soriani, A., Cardinali, C., Leoni, P., and Santoni, G. (2016). cannabinoids synergize with carfilzomib, reducing multiple myeloma cells viability and migration. Oncotarget.

Nabissi, M., Morelli, M.B., Santoni, M., and Santoni, G. (2013). Triggering of the TRPV2 channel by cannabidiol sensitizes glioblastoma cells to cytotoxic chemotherapeutic agents. Carcinogenesis 34, 48–57.

Nicholson, A.N., Turner, C., Stone, B.M., and Robson, P.J. (2004). Effect of Delta-9-tetrahydrocannabinol and cannabidiol on nocturnal sleep and early-morning behavior in young adults. J. Clin. Psychopharmacol. 24, 305–313.

Pagano, E., Capasso, R., Piscitelli, F., Romano, B., Parisi, O.A., Finizio, S., Lauritano, A., Marzo, V.D., Izzo, A.A., and Borrelli, F. (2016). An Orally Active Cannabis Extract with High Content in Cannabidiol attenuates Chemically-induced Intestinal Inflammation and Hypermotility in the Mouse. Front. Pharmacol. 7, 341.

Patel, R.R., Barbosa, C., Brustovetsky, T., Brustovetsky, N., and Cummins, T.R. (2016). Aberrant epilepsy-associated mutant Nav1.6 sodium channel activity can be targeted with cannabidiol. Brain J. Neurol.

Pazos, M.R., Cinquina, V., Gómez, A., Layunta, R., Santos, M., Fernández-Ruiz, J., and Martínez-Orgado, J. (2012). Cannabidiol administration after hypoxia-ischemia to newborn rats reduces long-term brain injury and restores neurobehavioral function. Neuropharmacology 63, 776–783.

Pazos, M.R., Mohammed, N., Lafuente, H., Santos, M., Martínez-Pinilla, E., Moreno, E., Valdizan, E., Romero, J., Pazos, A., Franco, R., et al. (2013). Mechanisms of cannabidiol neuroprotection in hypoxic–ischemic newborn pigs: Role of 5HT1A and CB2 receptors. Neuropharmacology 71, 282–291.

Rahimi, A., Faizi, M., Talebi, F., Noorbakhsh, F., and Naderi, N. (2015). Interaction between the protective effects of cannabidiol and palmitoylethanolamide in experimental model of Multiple Sclerosis in C57BL/6 mice. Neuroscience.

Ramer, R., Bublitz, K., Freimuth, N., Merkord, J., Rohde, H., Haustein, M., Borchert, P., Schmuhl, E., Linnebacher, M., and Hinz, B. (2012). Cannabidiol inhibits Lung Cancer cell invasion and metastasis via intercellular adhesion molecule-1. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 26, 1535–1548.

Ramer, R., Heinemann, K., Merkord, J., Rohde, H., Salamon, A., Linnebacher, M., and Hinz, B. (2013). COX-2 and PPAR-γ confer cannabidiol-induced apoptosis of human Lung Cancer cells. Mol. cancer Ther. 12, 69–82.

Ribeiro, A., Almeida, V.I., Costola-de-Souza, C., Ferraz-de-Paula, V., Pinheiro, M.L., Vitoretti, L.B., Gimenes-Junior, J.A., Akamine, A.T., Crippa, J.A., Tavares-de-Lima, W., et al. (2014). Cannabidiol improves lung function and inflammation in mice submitted to LPS-induced acute lung injury. Immunopharmacol. Immunotoxicol. 1–7.

Riedel, G., Fadda, P., McKillop-Smith, S., Pertwee, R. G., Platt, B., & Robinson, L. (2009). Synthetic and plant-derived cannabinoidreceptor antagonists show hypophagic properties in fasted and non-fasted mice. British Journal of Pharmacology156(7), 1154-1166. https://doi.org/10.1111/j.1476-5381.2008.00107.x

Sartim, A.G., Guimarães, F.S., and Joca, S.R.L. (2016). Antidepressant-like effect of cannabidiol injection into the ventral medial prefrontal cortex - possible involvement of 5-HT1A and CB1 receptors. Behav. Brain Res.

Schiavon, A.P., Soares, L.M., Bonato, J.M., Milani, H., Guimarães, F.S., and Weffort de Oliveira, R.M. (2014). Protective effects of cannabidiol against hippocampal cell death and cognitive impairment induced by bilateral common carotid artery occlusion in mice. Neurotox. Res. 26, 307–316.

Scott, K.A., Dalgleish, A.G., and Liu, W.M. (2014). The Combination of Cannabidiol and Δ9-Tetrahydrocannabinol Enhances the Anticancer Effects of Radiation in an Orthotopic Murine Glioma Model. Mol. cancer Ther.

Shrivastava, A., Kuzontkoski, P.M., Groopman, J.E., and Prasad, A. (2011). Cannabidiol induces programmed cell death in breast cancer cells by coordinating the cross-talk between apoptosis and autophagy. Mol. cancer Ther. 10, 1161–1172.

da Silva, J.A., Biagioni, A.F., Almada, R.C., de Souza Crippa, J.A., Cecílio Hallak, J.E., Zuardi, A.W., and Coimbra, N.C. (2015). Dissociation between the panicolytic effect of cannabidiol microinjected into the substantia nigra, pars reticulata, and fear-induced antinociception elicited by bicuculline administration in deep layers of the superior colliculus: The role of CB1-cannabinoid receptor in the ventral mesencephalon. Eur. J. Pharmacol. 758, 153–163.

Silvestri, C., Paris, D., Martella, A., Melck, D., Guadagnino, I., Cawthorne, M., Motta, A., and Marzo, V.D. (2015). Two non-psychoactive cannabinoids reduce intra-cellular lipid levels and inhibit hepatosteatosis. J. Hepatol.

Solinas, M., Massi, P., Cinquina, V., Valenti, M., Bolognini, D., Gariboldi, M., Monti, E., Rubino, T., and Parolaro, D. (2013). Cannabidiol, a Non-Psychoactive cannabinoid Compound, Inhibits Proliferation and Invasion in U87-MG and T98G Glioma Cells through a Multitarget Effect. PLoS ONE 8.

Song, C., Stevenson, C.W., Guimaraes, F.S., and Lee, J.L.C. (2016). Bidirectional Effects of Cannabidiol on Contextual Fear Memory Extinction. Front. Pharmacol. 7, 493.

Soroceanu, L., Murase, R., Limbad, C., Singer, E., Allison, J., Adrados, I., Kawamura, R., Pakdel, A., Fukuyo, Y., Nguyen, D., et al. (2013). Id-1 is a key transcriptional regulator of glioblastoma aggressiveness and a novel therapeutic target. cancer Res. 73, 1559–1569.

Stern, C.A.J., Gazarini, L., Vanvossen, A.C., Zuardi, A.W., Galve-Roperh, I., Guimaraes, F.S., Takahashi, R.N., and Bertoglio, L.J. (2015). Δ(9)-Tetrahydrocannabinol alone and combined with cannabidiol mitigate fear memory through reconsolidation disruption. Eur. Neuropsychopharmacol. J. Eur. Coll. Neuropsychopharmacol.

Torres, S., Lorente, M., Rodríguez-Fornés, F., Hernández-Tiedra, S., Salazar, M., García-Taboada, E., Barcia, J., Guzmán, M., and Velasco, G. (2011). A combined preclinical therapy of cannabinoids and temozolomide against glioma. Mol. cancer Ther. 10, 90–103.

Tubaro, A., Giangaspero, A., Sosa, S., Negri, R., Grassi, G., Casano, S., Della Loggia, R., and Appendino, G. (2010). Comparative topical anti-inflammatory activity of cannabinoids and cannabivarins. Fitoterapia 81, 816–819

Vuolo, F., Petronilho, F., Sonai, B., Ritter, C., Hallak, J.E.C., Zuardi, A.W., Crippa, J.A., and Dal-Pizzol, F. (2015). Evaluation of Serum Cytokines Levels and the Role of Cannabidiol Treatment in Animal Model of Asthma. Mediators Inflamm. 2015, 538670.

Ward, S.J., McAllister, S.D., Kawamura, R., Murase, R., Neelakantan, H., and Walker, E.A. (2014). Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT1A receptors without diminishing nervous system function or chemotherapy efficacy. Br. J. Pharmacol. 171, 636–645.

Wargent, E. T., Zaibi, M. S., Silvestri, C., Hislop, D. C., Stocker, C. J., Stott, C. G., … Cawthorne, M. A. (2013). The cannabinoid Δ9-tetrahydrocannabivarin (THCV) ameliorates insulin sensitivity in two mouse models of ObesityNutrition & Diabetes3(5), e68. https://doi.org/10.1038/nutd.2013.9

Weiss, L., Zeira, M., Reich, S., Har-Noy, M., Mechoulam, R., Slavin, S., and Gallily, R. (2006). Cannabidiol lowers incidence of Diabetes in non-obese diabetic mice. Autoimmunity 39, 143–151.

Weiss, L., Zeira, M., Mechoulam, R., Slavin, S., and Gallily, R. (2007). Treating or preventing Diabetes with cannabidiol.

Wheal, A.J., Cipriano, M., Fowler, C.J., Randall, M.D., and O’Sullivan, S.E. (2014). Cannabidiol improves vasorelaxation in Zucker diabetic fatty rats through cyclooxygenase activation. J. Pharmacol. Exp. Ther. 351, 457–466.

Wilkinson, J.D., and Williamson, E.M. (2007). cannabinoids inhibit human keratinocyte proliferation through a non-CB1/CB2 mechanism and have a potential therapeutic value in the treatment of Psoriasis. J. Dermatol. Sci. 45, 87–92.

Zheng, Y., Stiles, L., Hamilton, E., Smith, P.F., and Darlington, C.L. (2010). The effects of the synthetic cannabinoid receptor agonists, WIN55,212-2 and CP55,940, on salicylate-induced Tinnitus in rats. Hear. Res. 268, 145–150.

Zheng, Y., Reid, P., and Smith, P.F. (2015). cannabinoid CB1 Receptor Agonists Do Not Decrease, but may Increase Acoustic Trauma-Induced Tinnitus in Rats. Front. Neurol. 6, 60

Zuardi, A.W., Crippa, J. a. S., Hallak, J.E.C., Moreira, F.A., and Guimarães, F.S. (2006). Cannabidiol, a Cannabis sativa constituent, as an antipsychotic drug. Braz. J. Med. Biol. Res. 39, 421–429.

Synthetic Pathways

CBD is synthesized through decarboxylation of CBDA.

Degradation Pathways

CBD metabolism (mostly from: Ujváry and Hanuš, 2016)

Up to 50% of all applied CBD can be excreted either unchanged or in its glucuronidated form with about 16% found in urine and 33% in feces.

CBD can be hydroxylated and then further carboxylated by Cyp450 enzymes.

Cyp1A1 produces:

Cyp1A2 produces:

Cyp2A9 (minor contributor) produces:

Cyp2C19 produces:

Cyp2D6 produces:

Cyp3A4 produces:

Cyp3A5 produces:

Hydroxylated CBD, like 7-OH-CBD can be further metabolized to over 100 secondary metabolites, like 7-COOH-CBD.

Glucuronidation of OH-CBD as well as plain CBD occurs.

CBD effect on enzymes:

CBD and 6α/β-OH-CBD effectively inactivate Cyp2C and Cyp3A but have also been shown to induce the expression of Cyp3A (mouse), Cyp2B10 (mouse) and Cyp1A1 (human).

Similarly, 6-OH-CBD can induce Cyp2B10.

CBD (IC50 27.5 μM) and 7-OH-CBD (IC50 34 μM) inhibit FAAH, effectively raising endocannabinoid levels.

Similarly, CBD (IC50 22μM) and 7-OH-CBD (IC50 ±50 μM) inhibit cellular Anandamide uptake.

Receptor affinity:

CBD has a low affinity for CB1 and CB2 of around 5 μM. However, CBD can functionally antagonize (prevent agonists from binding) at far lower concentrations (79 nM for CB1 and 138 nM for CB2)(Pertwee, 2008), which can be transiently achieved for instance by smoking/inhalation.

Pharmacokinetics (from Ujváry and Hanuš, 2016, unless otherwise stated):

Injection of 20 mg CBD gives a peak plasma level of 686 ng/ml after 3 minutes and drops to 48 ng/ml after one hour. The average half-life of injected CBD is 24±6 hr.

Smoking 19 mg CBD gives a peak plasma level of 110 ng/ml after 3 minutes. The average half-life of smoked CBD is 31±4 hr.

Sublingual application of 20 mg CBD gives a peak plasma level of 2 ng/ml after 130 minutes.

Intranasal CBD (200 μg/kg, in PEG) results in a peak plasma level of ±30 ng/ml after about 30 minutes (Paudel et al., 2010).

Eating 40 mg CBD (plus 20 mg THC) gives a peak plasma level (for both CBD and THC) of 5 ng/ml after 1.5-3 hr.

Interestingly the presence of CBDA increases the plasma concentration of CBD 4-fold.

Daily oral doses of 700 mg CBD result in a constant (low) plasma level of 5.9-11.2 ng/ml dropping to 1.5 ng/ml one week after discontinuation.

Similarly a single oral dose of 600 mg CBD gives a blood concentration of 4.7±7 ng/ml after one hour and 17±29 ng/ml after two hours.

A single oral dose of 800 mg CBD produces a peak plasma level of 221 ±36 ng/ml after 3 hours.

Skin can hold up to 6.1 mg/g CBD

18 mg/ml gel (consisting of 80% propylene glycol and 20% water) results in a steady-state plasma level of 6.3±2.1 ng/ml CBD in guinea pigs (Paudel et al., 2010). Adding 6% v/v Transcutol HP increased the steady-state plasma level to 35.6 ±11.6 ng/ml.

References:

Paudel, K.S., Hammell, D.C., Agu, R.U., Valiveti, S., and Stinchcomb, A.L. (2010). Cannabidiol bioavailability after nasal and transdermal application: effect of permeation enhancers. Drug Dev. Ind. Pharm. 36, 1088–1097.

Pertwee, R.G. (2008). The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br. J. Pharmacol. 153, 199–215.

Ujváry, I., and Hanuš, L. (2016). Human Metabolites of Cannabidiol: A Review on Their Formation, Biological Activity, and Relevance in Therapy. Cannabis cannabinoid Res. 1, 90–101.

 

Clinical Trials

Addiction

In one case report, CBD successfully suppressed anxiety symptoms normally associated with cannabis withdrawal (Crippa et al., 2013).

In a pilot clinical trial, inhaled CBD reduced cigarette smoking by 40% (Morgan et al., 2013).

 

anxiety

In human volunteers, 32 mg of CBD enhanced fear extinction suggesting an anxiolytic effect (Das et al., 2013).

In both healthy volunteers and patients with Social anxiety Disorder an oral dose of 600 mg CBD significantly reduced anxiety, cognitive impairment and discomfort related to a public speaking assignment (Bergamaschi et al., 2011).

In a similar test 400 mg CBD reduced anxiety and activity in limbic brain regions (Crippa et al., 2004).

 

cancer

In patients with cancer-related pain an oral spray with a 1/1 THC/CBD mix significantly reduced pain without desensitization or adverse side effects (Johnson et al., 2013).

 

epilepsy

Two clinical trials in the 1980’s investigated the therapeutic properties of CBD in epilepsy. CBD was found to be effective in 50% of patients, meaning that seizure occurrence was reduced by >50%. The occurrence of seizures was reduced by less than 50% in a further 37.5% with no effect observed in the remaining 12.5% (Cunha et al., 1980; Pickering et al., 2011).

In one very public case, a girl with Dravet syndrome (loss of function mutation in the sodium channel SCN1A), went from having more than 50 convulsive seizures per day to less than 3 nocturnal seizures per month by using extract from a Cannabis variety Charlotte’s Web, which has a THC content of 0.5% and a CBD content of 17% (Maa and Figi, 2014). The authors stress that there is synergy between cannabinoids and that cannabis extracts are superior to individually purified cannabinoids.

In 8 patients 200-300 mg/day oral CBD was administered for up to 4.5 months. Fours patients became almost seizure-free, 3 patients showed partial improvement and one patient did not improve (Cunha et al., 1980).

 

Multiple Sclerosis

In a 276 patient trial an oral spray with 1/1 THC/CBD for 3-4 months provided significant symptom relief in about 75% of patients (Flachenecker et al., 2014).

In a similar trial THC/CBD spray was effective against neuropathic pain associated with Multiple Sclerosis (Rog et al., 2007).

 

pain

In patients with cancer-related pain an oral spray with a 1/1 THC/CBD mix significantly reduced pain without desensitization or adverse side effects (Johnson et al., 2013).

THC/CBD spray was effective against neuropathic pain associated with Multiple Sclerosis (Rog et al., 2007).

 

Parkinson’s disease

In a 4-patient study, CBD immediately reduced insomnia associated with Parkinson’s Disease (Chagas et al., 2014).

In another trial, patients receiving 75 or 300 mg CBD/day reported improved quality of life (Chagas et al., 2014).

In a small-scale trial, CBD was found to decrease psychotic symptoms of Parkinson’s without affecting motor function (Zuardi et al., 2009).

 

psychosis / schizophrenia

In a small-scale clinical trial up to 4 doses of 200 mg CBD/day suppressed psychotic symptoms as effectively as amisulpride but with fewer side-effects (Leweke et al., 2012).

Similarly, in several case reports CBD doses of up to 1500 mg/day for up to 4 weeks produced similar anti-psychotic effects as observed with classical anti-psychotics but with fewer side-effects (reviewed in Zuardi et al., 2006).

 

References:

Bergamaschi, M.M., Queiroz, R.H.C., Chagas, M.H.N., de Oliveira, D.C.G., De Martinis, B.S., Kapczinski, F., Quevedo, J., Roesler, R., Schröder, N., Nardi, A.E., et al. (2011). Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naïve social phobia patients. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 36, 1219–1226.

Chagas, M.H.N., Zuardi, A.W., Tumas, V., Pena-Pereira, M.A., Sobreira, E.T., Bergamaschi, M.M., dos Santos, A.C., Teixeira, A.L., Hallak, J.E.C., and Crippa, J.A.S. (2014). Effects of cannabidiol in the treatment of patients with Parkinson’s disease: an exploratory double-blind trial. J. Psychopharmacol. Oxf. Engl. 28, 1088–1098.

Crippa, J. a. S., Hallak, J.E.C., Machado-de-Sousa, J.P., Queiroz, R.H.C., Bergamaschi, M., Chagas, M.H.N., and Zuardi, A.W. (2013). Cannabidiol for the treatment of cannabis withdrawal syndrome: a case report. J. Clin. Pharm. Ther. 38, 162–164.

Crippa, J.A. de S., Zuardi, A.W., Garrido, G.E.J., Wichert-Ana, L., Guarnieri, R., Ferrari, L., Azevedo-Marques, P.M., Hallak, J.E.C., McGuire, P.K., and Filho Busatto, G. (2004). Effects of cannabidiol (CBD) on regional cerebral blood flow. Neuropsychopharmacol. Off. Publ. Am. Coll. Neuropsychopharmacol. 29, 417–426.

Cunha, J.M., Carlini, E.A., Pereira, A.E., Ramos, O.L., Pimentel, C., Gagliardi, R., Sanvito, W.L., Lander, N., and Mechoulam, R. (1980). Chronic administration of cannabidiol to healthy volunteers and epileptic patients. Pharmacology 21, 175–185.

Das, R.K., Kamboj, S.K., Ramadas, M., Yogan, K., Gupta, V., Redman, E., Curran, H.V., and Morgan, C.J.A. (2013). Cannabidiol enhances consolidation of explicit fear extinction in humans. Psychopharmacology (Berl.) 226, 781–792.

Flachenecker, P., Henze, T., and Zettl, U.K. (2014). Nabiximols (THC/CBD oromucosal spray, Sativex®) in clinical practice--results of a multicenter, non-interventional study (MOVE 2) in patients with Multiple Sclerosis spasticity. Eur. Neurol. 71, 271–279.

Johnson, J.R., Lossignol, D., Burnell-Nugent, M., and Fallon, M.T. (2013). An open-label extension study to investigate the long-term safety and tolerability of THC/CBD oromucosal spray and oromucosal THC spray in patients with terminal cancer-related pain refractory to strong opioid analgesics. J. pain Symptom Manage. 46, 207–218.

Leweke, F.M., Piomelli, D., Pahlisch, F., Muhl, D., Gerth, C.W., Hoyer, C., Klosterkötter, J., Hellmich, M., and Koethe, D. (2012). Cannabidiol enhances Anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl. Psychiatry 2, e94.

Maa, E., and Figi, P. (2014). The case for medical marijuana in epilepsy. Epilepsia 55, 783–786.

Morgan, C.J.A., Das, R.K., Joye, A., Curran, H.V., and Kamboj, S.K. (2013). Cannabidiol reduces cigarette consumption in tobacco smokers: preliminary findings. Addict. Behav. 38, 2433–2436.

Pickering, E.E., Semple, S.J., Nazir, M.S., Murphy, K., Snow, T.M., Cummin, A.R., Moosavi, S.H., Guz, A., and Holdcroft, A. (2011). cannabinoid effects on ventilation and breathlessness: a pilot study of efficacy and safety. Chron. Respir. Dis. 8, 109–118.

Rog, D.J., Nurmikko, T.J., and Young, C.A. (2007). Oromucosal delta9-tetrahydrocannabinol/cannabidiol for neuropathic pain associated with Multiple Sclerosis: an uncontrolled, open-label, 2-year extension trial. Clin. Ther. 29, 2068–2079.

Zuardi, A.W., Crippa, J. a. S., Hallak, J.E.C., Moreira, F.A., and Guimarães, F.S. (2006). Cannabidiol, a Cannabis sativa constituent, as an antipsychotic drug. Braz. J. Med. Biol. Res. 39, 421–429.

Zuardi, A.W., Crippa, J. a. S., Hallak, J.E.C., Pinto, J.P., Chagas, M.H.N., Rodrigues, G.G.R., Dursun, S.M., and Tumas, V. (2009). Cannabidiol for the treatment of psychosis in Parkinson’s disease. J. Psychopharmacol. Oxf. Engl. 23, 979–983.