162
Views
1
CrossRef citations to date
0
Altmetric
Review

Critical appraisal of the potential use of cannabinoids in cancer management

&
Pages 301-313 | Published online: 30 Aug 2013

Abstract

Cannabinoids have been attracting a great deal of interest as potential anticancer agents. Originally derived from the plant Cannabis sativa, there are now a number of endo-, phyto- and synthetic cannabinoids available. This review summarizes the key literature to date around the actions, antitumor activity, and mechanisms of action for this broad range of compounds. Cannabinoids are largely defined by an ability to activate the cannabinoid receptors – CB1 or CB2. The action of the cannabinoids is very dependent on the exact ligand tested, the dose, and the duration of exposure. Some cannabinoids, synthetic or plant-derived, show potential as therapeutic agents, and evidence across a range of cancers and evidence in vitro and in vivo is starting to be accumulated. Studies have now been conducted in a wide range of cell lines, including glioma, breast, prostate, endothelial, liver, and lung. This work is complemented by an increasing body of evidence from in vivo models. However, many of these results remain contradictory, an issue that is not currently able to be resolved through current knowledge of mechanisms of action. While there is a developing understanding of potential mechanisms of action, with the extracellular signal-regulated kinase pathway emerging as a critical signaling juncture in combination with an important role for ceramide and lipid signaling, the relative importance of each pathway is yet to be determined. The interplay between the intracellular pathways of autophagy versus apoptosis is a recent development that is discussed. Overall, there is still a great deal of conflicting evidence around the future utility of the cannabinoids, natural or synthetic, as therapeutic agents.

Introduction

The cannabinoids are a class of over 60 compounds derived from the plant Cannabis sativa, as well as the synthetic or endogenous versions of these compounds.Citation1 Cannabis has been used as a medicinal and recreational drug for many centuries, but its psychoactive properties have led to legal regulations around access and use in most countries.Citation2 Despite this, scientific research into both natural and synthetic cannabinoids has continued. Studies are now being conducted on the potential efficacy of cannabinoids, both natural and synthetic, as anticancer agents and their possible mechanisms of action.

The first cannabinoid to be intensively studied was transCitation9-tetrahydrocannabinol (ΔCitation9-THC) which was first isolated in the 1960s.Citation3 While several other active compounds, notably ΔCitation8-THC, cannabinol, cannabidiol, and cannabicyclol, were able to be isolated, it was not until 1992 that an analogous endogenous ligand – anandamide (AEA) – was identified ().Citation4 This discovery was closely followed by the identification of the endogenous ligands 2-arachidonoyl glycerol (2-AG), 2-arachidonyl glyceryl ether (2-AGE), O-arachidonoyl ethanolamine (virodhamine), and N-arachidonoyl dopamine (NADA).Citation5 As the names of these compounds suggest, they are derivatives of the parent compound arachidonic acid, which is an important lipid-signaling molecule and a key component of the inflammatory pathway.Citation6 The endocannabinoids mimic the actions of ΔCitation9-THC in mouse behavioral tests, interfere with learning and memory, activate the hypothalamic–pituitary–adrenal axis, decrease hypothalamus prolactin secretion, decrease intraocular pressure, cause hypotension and bradycardia, and modulate the immune system.Citation7,Citation8

Table 1 A selection of cannabinoid receptor ligands and their specificities

The first cannabinoid receptor was discovered in 1988Citation9 and then cloned in 1990.Citation10 This was followed in 1993 with the discovery of a second form of the receptor, which shares 44% amino acid identity and a distinct yet similar binding profile for cannabinoid compounds.Citation11 This development led to the current terminology of CB1, for the original receptor form, and CB2. CB1 receptors are found throughout the brain, spleen, eye, testis, and uterus,Citation10Citation12 whereas CB2 receptors are associated with the cells and organs of the immune system as well as tumor cells.Citation11,Citation13,Citation14 Both receptors are part of the G-protein-coupled receptor (GPR) superfamily. In general, cannabinoid agonists do not show a great deal of selectivity between CB1 and CB2 binding; however, newly developed synthetic antagonists are now available that allow the experimental delineation of CB1 versus CB2 effects. On a general level, CB1 binding is responsible for the psychoactive properties of the cannabinoid agonists, and CB2 binding mediates immune effects.Citation5 This is discussed in more detail in later sections. The first cannabinoid-receptor antagonist was released in 1994 – SR141716 (rimonabant).Citation15 Originally designed as a treatment for obesity, it has seen extensive use as a pharmacological tool in assessing agonist actions through the CB1 receptor; however, the classification of this compound as a pure antagonist is likely to be misleading, and its more recent classification as a selective CB1-receptor inverse agonist is more accurate. Following the discovery of SR141716, a range of antagonists and inverse agonists has been developed, including SR144528, which is an inverse agonist at the CB2 receptorCitation16Citation18 ().

In the 20 years since the discovery of the CB2-receptor isoform, there have been a number of anomalous results that suggest that cannabinoid agonists exert actions beyond those mediated by CB1 or CB2.Citation19 This has led to the suggestion that there may be more isoforms of the CB receptor still to be identified; possible candidates include GPR119, GPR55, and GPR18.Citation20 Of the three, GPR55 has the largest body of evidence, suggesting it should be renamed as a cannabinoid receptor, but low-sequence homology with CB1 and CB2 along with conflicting results in agonist-binding studies means that it has not yet been fully reclassified.Citation20,Citation21 Both GPR119 and GPR18 show evidence of cannabinoid binding, but the results are not sufficiently robust to rename either protein as a cannabinoid-receptor isoform.Citation5,Citation20,Citation21 The transient receptor potential cation channel subfamily V member 1 (TRPV1) receptor has also been proposed as a possible cannabinoid isoform. The endogenous cannabinoids AEA and NADA are potent TRPV1 agonists, and a raft of evidence has established the in vivo activity of these compounds unique to TRPV1 signaling.Citation22 However, TRPV1 is an ionotropic receptor rather than a GPR, which means it does not meet one of the five criteria established for classification as a cannabinoid receptor. The ruling body, the International Union of Basic and Clinical Pharmacology, is seeking further evidence before renaming TRPV1 as a possible ionotropic cannabinoid CB3 receptor.Citation20

Physiological functions of cannabinoids

Synthesis

The endocannabinoids (AEA and 2-AG) are generally accepted to be synthesized on demand following receipt of an intracellular signal, such as cellular depolarization or Ca2+-dependent receptor stimulation. However, there is also mounting evidence to suggest that AEA may be stored to some degree within the adiposome cellular compartment.Citation23 AEA is synthesized from lipid precursors via enzymatic hydrolysis of N-acyl-phosphatidylethanolamines (NAPEs). This reaction is catalyzed by phospholipase D (PLD), which shows selectivity for this reaction.Citation24,Citation25 PLD activity is regulated by cellular depolarization, by activation of the NMDA, and by the metabotropic classes of glutamate receptor.Citation24 The NAPE precursor is derived from the transfer of arachidonic acid from phosphatidylcholine to phosphatidylethanolamine, a reaction catalyzed by an N-acetyltransferase isoform. This transfer is modulated by the presence of Ca2+ and cyclic adenosine monophosphate (cAMP).Citation24

The synthesis of 2-AG may theoretically proceed through several putative pathways, but consensus favors hydrolysis of membrane lipids through PLC to produce 1,2-diacylglycerol.Citation26 1,2-diacylglycerol can then be converted through the action of diacylglycerol lipase to the 2-AG molecule. Although 2-AG concentrations are linked to membrane depolarization, similar to AEA, the underlying control mechanisms are thought to be different, as intracellular AEA concentrations cannot be directly linked to 2-AG concentrations.Citation24,Citation26

Degradation

Once synthesized, the endocannabinoids act locally, either by interacting with plasma-bound cannabinoid receptors on the cell where they were produced or on directly neighboring cells. Signaling is terminated by the uptake of the endocannabinoid by a regulated mechanism facilitated by the AEA membrane transporter.Citation25 Following uptake, the cannabinoid is hydrolyzed by the fatty acid amide hydrolase and the monoacylglyceride hydrolase. Fatty acid amide hydrolase is a non-specific enzyme which catalyzes the hydrolysis of numerous fatty acid molecules and as such is widely distributed around the body.Citation27 Monoacylglyceride hydrolase, in contrast, is limited in its distribution to the nerve terminals of specific brain neurons.Citation28

Intracellular actions

Both forms of the traditional cannabinoid receptor, CB1 and CB2, are G-protein-linked. This means their activation results in inhibition of adenylate cyclase which is blocked by pertussis toxin. This causes a decrease in cellular cAMP, activation of the p38 mitogen-activated protein kinase (MAPK) pathway, and/or activation of ion channels. Evidence suggests that different conformational forms of GPRs allows downstream activities to be specifically modulated based on the individual ligand that binds and activates the receptor ().Citation29 This specificity of action has been observed for the CB1 receptor whereby the antagonist WIN-55,212-2 activated different Gi/0 subtypes with differing sensitivitiesCitation25,Citation26,Citation30,Citation31 ().

Table 2 Intracellular action of cannabinoids

Systemic actions of cannabinoids

The most obvious action of the cannabinoids is in the brain. Receptors and synthesis and degradation enzymes of the cannabinoids are found distributed throughout the central nervous system.Citation32 Ingestion of cannabinoids results in mood alterations, sedation, increased appetite, hallucinations, and impairment of memory, coordination, and executive function.Citation33 At higher concentrations, cannabinoids produce analgesia.Citation34 The majority of these effects are purported to be mediated through alterations of signaling through the glutamatergic neurons or γ-aminobutyric acid (GABA) neurons located in different areas of the brain.Citation35 Cannabinoid action in the motor cortex, basal ganglia, or cerebellum leads to impaired motor performance and ataxia.Citation36,Citation37 The extreme effect of catalepsy is thought to be mediated by action at the globus pallidus, striatal spiny neurons, and cortical glutamate neurons.Citation38,Citation39 The analgesic properties of the cannabinoids are linked to action on GABA neurons in the periaqueductal grey and rostroventral medulla regions and the ventral posterolateral nucleus of the thalamus.Citation40 CB1 receptors are also present in the spinal cord and on peripheral nociceptors, where they can mediate neuropathic pain and inflammation.Citation41Citation43 However, recently there has been a degree of controversy around the specificity of antibodies used in several cannabinoid-receptor immunolocalization studies.Citation44 Therefore, it is possible that the results regarding the exact distribution of the receptor in anatomical substructures are likely to be challenged and refined over the coming years.

The role of cannabinoids as modulators of the immune system is becoming more obvious. CB1 receptors are located on T lymphocytesCitation45 whereas CB2 receptors, historically associated with immune function, are located in human B cells, natural killer cells, monocytes, polymorphonuclear neutrophils, and T cells.Citation46 In general, cannabinoid agonists are associated with decreased immune function and decreased release of inflammatory mediators, such as interleukin (IL)-1, IL-2, IL-6, and IL-12.Citation47 However, this effect is complicated by duration of exposure and dose.Citation46,Citation48

Role of cannabinoids in cancer

Two therapeutic avenues exist for the development of cannabinoids as anticancer agents. As antiemetic and analgesic compounds, this class of compounds has been explored in terms of palliative care. More recently, cannabinoid agonists and antagonists have been screened for potential direct antitumorigenic properties.

Palliative care

Cannabinoids can play an important role in the palliation of pain, nausea, vomiting, and appetite for cancer patients; however, this is beyond the scope of this review. The palliative uses of cannabinoids have been reviewed elsewhere.Citation49Citation51

Antitumorigenic properties

Cannabinoids are not yet approved for the treatment of tumor progression, although their antitumorigenic effects have been known for over 30 years.Citation52 Cannabinol and ΔCitation8-THC inhibited tumor growth in a mouse model of Lewis lung adenocarcinoma after 20 days of treatment, whereas cannabidiol or ΔCitation9-THC failed to show any effect.Citation52 Since this pioneering study, a vast range of cancer cell and tumor models have been used to evaluate the possible efficacy and mechanisms of cannabinoid antitumor activity. This work is supported by findings that the endocannabinoid system may be altered during disease states. Significant levels of the cannabinoid receptor are found in prostate, breast, leukemia, melanoma, and thyroid cell lines, as well as colorectal and hepatocellular carcinoma tissue.Citation53Citation58 Of particular significance is the fact that in prostate cancer cell lines, there is evidence that the expression of both CB1 and CB2 is elevated compared to normal prostate cells.Citation53 Similarly, in lymphoma and breast cancer tissue, as well as some derived cell lines, CB1 and CB2 are overexpressed.Citation59,Citation60 The degree of increased expression correlates with tumor aggression and progression, an effect also reported in human astrocytes.Citation54,Citation61 In contrast, McKallip et al found that Michigan Cancer Foundation (MCF)-7 and MDA-MB-231 breast cancer cell lines showed very low levels of CB1 expression, and CB2 was below detectable levels, as determined by reverse-transcription polymerase chain reaction.Citation62 One reason for the reported discrepancies may be inaccuracies in reporting the subcellular localization of the receptor. It is probable that extensive trafficking between the plasma membrane and internal compartments, such as lysosomes, occurs, and that this is altered by exposure to cannabinoid agonistsCitation59,Citation63Citation69 ().

Table 3 Cannabinoid action in various cancer cell lines

Anticancer actions of specific compounds

Endo- and phytocannabinoids (ΔCitation9-THC, AEA, cannabidiol)

The primary active constituent of cannabis, ΔCitation9-THC, has been investigated in a number of in vitro- and in vivo-based systems. Overall, some efficacy has been recorded in breast, prostate, glioma, lymphoma, and pancreatic cancer cell lines. In contrast, there are reports of pro-cancerous activity in breast, bronchial, hepatoma, and lung cell lines ().

Results are not clear-cut as to whether ΔCitation9-THC causes pro- or antiproliferative effects in breast cancer cells. A study of ΔCitation9-THC in MCF-7 and MDA-MB-231 cells (≤5 μM) reported proliferation in response to cannabinoid treatment.Citation62 This finding is supported by the work of Takeda et al, who also documented a proliferative response to ΔCitation9-THC in MCF-7 cells.Citation70 In contrast, McAllister et al reported decreased proliferation in MDA-MB-231 and MDA-MB-468 cells, while studies in EVSA-T cells also showed ΔCitation9-THC inhibited cell growth.Citation71 In MCF-7 cells, von Bueren et al reported that ΔCitation9-THC did not induce cell proliferation at concentrations up to 1 mM, but did inhibit 17β-estradiol (100 nM)-induced proliferation at concentrations above 1 μM.Citation72 In mouse models of breast cancer, contradictory results have also been reported. In a xenotransplant model of 4T1 paw cells in BALB/c mice, an increase in tumor size was recorded following ΔCitation9-THC (25 mg/kg, intraperitoneally, 21 days).Citation62 In contrast, in MMTV-neu mice that showed spontaneous development of mammary tumors, ΔCitation9-THC (0.5 mg/animal/day, peritumorally, 90 days) decreased the size of mammary tumors, the occurrence of new spontaneous tumors, and the degree of metastasis to the lungs.Citation73 These results indicate that the role of compounds such as ΔCitation9-THC is not well understood, and its effects are possibly regulated by a range of factors that are yet to be determined.

In prostate cancer cell lines, ΔCitation9-THC and 2-AG increased cellular proliferation, whereas AEA had the opposite effect. Sánchez et al documented increased proliferation of LNCaP and PC3 cells following treatment with ΔCitation9-THC (50 nM).Citation74,Citation75 In the same cell lines, AEA decreased proliferation. Mimeault et al evaluated AEA in PC3, LNCaP, and DU145 cell lines, and showed a significant decrease in cell viability at concentrations above 2 μM.Citation76 Similarly, Nithipatikom et al documented a decrease in PC-3 cellular proliferation at AEA concentrations above 1 μM, whereas 2-AG at similar concentrations caused an increase in cellular growth.Citation77

In glioma cell lines, almost all studies show that cannabinoids decrease cell proliferation. ΔCitation9-THC has been investigated in C6, SF126, U87-MG, U251, SF188, and U373-MG cell lines at concentrations of 1–2 μM. Results showed a consistent decrease in cell viability independent of cell line.Citation78,Citation79 In U87 and U373 cells, cannabidiol inhibited cell proliferation, but the half-maximal inhibitory concentration (IC50) value for this effect was unusually high at 25 μM.Citation80 Both AEA and 2-AG also demonstrated antiproliferative effects in the C6 cell line with IC50 values of 1.6 and 1.8 μM, respectively.Citation81

ΔCitation9-THC shows inhibitory effects in a range of pancreatic cell lines; however, this effect appears to be biphasic, with evidence of proliferation at concentrations under 1 μM and inhibition at concentrations above 2 μM. This biphasic trend was most obvious in the Panc1 and Capan2 cell lines and less obvious in MiaPaCa2 and BxPc3 cells.Citation82 When MiaPaCa2 cells were implanted subcutaneously into nude mice and left to form tumors, ΔCitation9-THC (15 mg/kg/day, 15 days) caused a significant reduction in tumor growth.Citation82

Both ΔCitation9-THC and AEA (or the stable equivalent Met-AEA) inhibited the growth of lymphoma cell lines, although this effect was related to the serum content of the cell media used. In serum-free media, ΔCitation9-THC showed significant inhibition of growth in EL-4, LSA, and P815 cells at concentrations above 3 μM. However, with the inclusion of 5% calf serum in the media, no significant effect was observed until concentrations of 10 μM were reached.Citation83 Herrera et al reported a decrease in the viability of Jurkat cells at concentrations of 1.5 μM and above when using heat-inactivated fetal bovine serum, though this study did not assess cell viability in a completely media-free system.Citation84 However, several other studies have reported a decrease in the cell viability of Jurket cells following exposure to ΔCitation9-THC.Citation83,Citation85Citation88 In mantle-cell lymphoma, two studies reported an effect of AEA or the stable equivalent, Met-AEA, with the effective concentration ranging from 5 to 10 μM for each compound.Citation89,Citation90

In contrast to the inhibitory effects documented above, the natural cannabinoids have also shown to increase cell proliferation, although normally these results have been documented in studies investigating smoke mixtures that include natural cannabinoid compounds. One study using pure ΔCitation9-THC at concentrations between 0.1 and 0.3 μM showed increased proliferation of NCI-H292 lung carcinoma cells.Citation91 In the lungs of habitual marijuana smokers, significant increases in the proliferation marker Ki67 were observed along with changes in the expression of the epidermal growth-factor receptor, the human epidermal growth-factor receptor 2/neu receptor, p53, and DNA polyploidy.Citation92 In hepatoma cells, ΔCitation9-THC (2 μg/mL) induced the drug metabolizing enzyme cytochrome P450 1 A1, which is linked to the development of tobacco-related cancers.Citation93 This induction effect was seen with both a marijuana-derived tar mixture and pure extracts of ΔCitation9-THC. No effects on cellular proliferation were reported.Citation93 Epidemiological studies around the development of cancer in cannabis smokers have been similarly inconclusive as to whether natural cannabinoids are pro- or anticarcinogenic.Citation94Citation96

Synthetic cannabinoids (WIN-55,212-2, and JWH-133)

WIN-55,212-2 is a nonselective agonist of CB1 and CB2 that exhibits anticancer effects in prostate, glioblastoma, glioma, breast, lymphoma, and melanoma cell lines.Citation53,Citation79,Citation97,Citation98 In the LNCaP prostate cancer cell line, WIN-55,212-2 dose-dependently decreased cell viability, with an overall IC50 value of 6 μM.Citation53 Similar to malignant glioma cells, the levels of both CB1 and CB2 were elevated in this cell line, as well as in DU145, PC3, and CWR22Rv1 lines.Citation53 The inhibition of prostate cancer growth can be mediated by both the CB1-and CB2-receptor isoforms. When WIN-55,212-2 (7.5 μM) was coadministered with either SR141716 or SR144528 (2 μM), the cell growth recovered.Citation53 In a study of cell invasion, WIN-55,212-2 at 1 nM caused a 40% reduction in the invasion measure of PC3 cells.Citation97 In contrast, in DU-145 cells no reduction was seen until 100 nM (causing a 20% reduction), and no effect was observed in LNCaP cells. In breast cancer cells (MDA-MB-231 and MDA-MB-468) and an MDA-MB-231 tumor-implant model, WIN-55,212-2 inhibited cell and tumor growth (in vitro IC50≤10 μM, in vivo 5 mg/kg/day, intratumorally for 4 weeks).Citation54 One factor of critical importance in all these studies is the dose of agonist administered. The cannabinoid receptor shows ligand-binding affinity for WIN-55,212-2 within the nanomolar range (3 and 16 nM for human CB1 and CB2, respectively).Citation18 Therefore, it can be suggested that at doses above 4 μM, any observed effects may not be cannabinoid receptor-mediated. This warrants further investigation, particularly in the prostate and breast cancer cell lines previously discussed.

WIN-55,212-2 (1.25 μM) inhibited cell growth by greater than 50% in the SF126, U87-MG, U251, U373-MG, and SF-188 glioblastoma cell lines, while in C6 glioma cell lines 15 μM was required to generate a similar effect.Citation79,Citation98 In vivo, regression of C6 cell tumors was observed following 8 days of WIN-55,212-2 administration (50 μg/day intratumorally).Citation61

In vivo, the growth of melanoma xenografts was decreased in WIN-55,212-2-treated mice. Blázquez et al demonstrated decreased cell viability in the melanoma-derived B16 and A375 cell lines and a significant reduction in tumor volume in B16-implanted mice following 8 days of WIN-55,212-2 treatment (50 μg/day, peritumorally).Citation56 Similar results were seen in a PDV.C57 implantation model of melanoma, with tumor shrinkage observed following 11 days of WIN-55,212-2 treatment (1.5 μg/day, peritumorally by continuous-flow pump).Citation99

Similarly, WIN-55,212-2 has been shown to have efficacy in mantle-cell lymphoma tumors and cell lines. In Rec-1, JeKo, and JVM-2 cell lines, WIN-55,212-2 induced cell death and cleavage of caspase 3 at doses above 5 μM.Citation89,Citation100 This effect was blocked by pre-treatment with either a CB1 or CB2 inhibitor (SR141716A or S144528).Citation100 In tumors derived from mantle-cell lymphoma patient samples, similar cellular toxicity was observed, with IC50 values ranging between 1.47 and 4.81 μM, depending on the individual patient.Citation60 Interestingly, this decrease in cell viability did not correlate with the cleavage of caspase 3, suggesting that the mechanism of action is not via apoptosis.Citation60

JWH-133 is a selective agonist for the CB2 receptor; therefore, it has been investigated as a possible cancer therapeutic that may lack psychoactive side effects. The first study to demonstrate the cytotoxicity of JWH-133 in vitro was conducted in glioma cells.Citation61 CB2 is expressed at low levels in microglial cells under normal conditions; however, overexpression is correlated with the development of malignancy.Citation101,Citation102 In glioma cells, JWH-133 reduced cell viability by 50% in vitro, while in vivo studies showed a 71% decrease in tumor growth after 8 days (Rag−/− mice, 40 μg/day intratumorally). This effect on tumor growth was inhibited by the CB2 antagonist SR144528, but not the CB1-specific antagonist SR141716.Citation61 These results are supported by studies in glioma (C6 cells) and astrocytoma xenografts, where the overall vascularization of tumors was reduced by 88% and 21% respectively (50 μg/day intratumorally, 8 or 25 days for gliomas and astrocytes).Citation103

In a skin-tumor model, JWH-133 (83 μg/day for 11 days via continuous-flow pump) caused a 60% decrease in tumor size in PDV.C57 cell xenografts.Citation99 In melanoma xenografts, JWH-133 decreased tumor volume by 75% over an 8-day period (50 μg/day), with tumors displaying decreased vascularity and increased numbers of apoptotic cells.Citation56

In breast cancer cell lines, JWH-133 decreases cell proliferation and induces apoptosis. In the breast MDA-MB-231 cell line, a 58% reduction in growth and migration was observed at 10 μM.Citation54 A similar effect was seen in the MDA-MB-468 breast cancer cell line. These results were confirmed in vivo, with a 46% reduction in MDA-MB-231-implanted tumor growth over an 8-week period (5 mg/kg/day intraperitoneally), an effect that was blocked by the simultaneous administration of SR144528.Citation54 In MMTV-neu mice, a model of Erb2-driven metastatic breast cancer, JWH-133 at 50 μg/day (peritumorally twice a week for 90 days) exhibited a range of anticancer effects, including decreased tumor size, inhibition of new tumor development, decreased tumor cell proliferation, inhibition of angiogenesis, and decreased lung metastasis.Citation73 These results were comparable to findings with the mixed agonist ΔCitation9-THC, suggesting an important role for the CB2 receptor in the ΔCitation9-THC mediated effect.

Overview of potential mechanisms

Effects on tumor growth and development

Cannabinoids affect a range of pathways that regulate cell division and viability; however, the knowledge in this area remains incomplete. For example, it is still difficult to explain the myriad of results around cell survival that have been reported in the literature. This is confounded by a lack of understanding of the possible receptors involved and ongoing doubt over their definitive localization. In addition, the actual mechanisms of cell death remain controversial, with some authors maintaining that autophagy precedes apoptosis and others suggesting that apoptosis is stimulated directly. Current evidence suggests that the type and stage of the cancer is likely to be important, with hormone-dependent cancers possibly reacting differently to cannabinoid exposure than gliomas (the most studied cancer type in terms of cannabinoid action).

Endogenous cannabinoids regulate the de novo synthesis of ceramides, lipid-based components of the cell membrane that perform both structural and signaling functions. It is becoming increasingly obvious that ceramide functions as a physiological signaling molecule, particularly with regard to the control of apoptosis, but also growth arrest, differentiation, cell migration, and adhesion.Citation104 As such, the role and regulation of ceramide signaling is attracting increasing attention, and ceramide now has an accepted role in the development of some cancers.Citation105 Activation of either CB1 or CB2 in glioma cells is associated with an increase in ceramide levels leading to the activation of the extracellular signal-regulated kinase (ERK) pathway via Raf-1 activation and p38 MAPK activation.Citation14,Citation106 Both these pathways ultimately cause apoptosis through caspase activation and/or cell-cycle arrest.Citation14 In breast cancer cells, the CB1 antagonist SR141716 inhibited cell proliferation through the effects of ERK1/2 colocalized inside membrane lipid rafts/caveloae.Citation59 Such rafts play a critical role in the growth and metastasis of breast tumors.Citation107,Citation108 A final component of the ERK pathway, p53, plays a crucial role in switching between cell-cycle arrest and apoptosis.Citation109 In cultured cortical neurons, ΔCitation9-THC activated p53 via the CB1 receptor, thereby activating the apoptotic cascade involving B-cell lymphoma (Bcl)-2 and Bcl-2-associated X protein, suggesting that the cannabinoid pathway ultimately causes cellular death via apoptosis.Citation110

It is also likely, at least in some cell types, that autophagy precedes the apoptotic cascade. Autophagy has been reported in glioma, pancreatic, breast, and hepatocellular carcinoma cells, with additional reports that WIN-55,212-2 causes autophagy in mantle-cell lymphoma.Citation111,Citation112 As yet, there is no evidence for autophagy in rhabdomyosarcoma, leukemia, prostate, or melanoma cell lines.Citation111 Autophagy is linked to endoplasmic reticulum stress, and follows similar pathways to the aforementioned ERK-mediated mechanism. The upregulation of ceramide appears to be the likely link between cannabinoid exposure and these endoplasmic reticulum effects. CB1 and CB2 activation induces serine palmitoyltransferase, the rate-limiting step of de novo ceramide synthesis.Citation113 Ceramide activates nuclear protein 1 (previously p8) through the action of ER-associated eIF2α which triggers a signaling cascade through tribbles homologue 3, AKT, and mammalian target of rapamycin complex 1 to cause autophagy.Citation114 This process involves the encapsulation of key organelles in doublemembrane vesicles for breakdown and recycling. There has been debate as to whether this process is cytoprotective or cytotoxic, but it appears it can be both. In the case of cannabinoid treatment, apoptosis through mitochondrial disruption is observed.Citation114,Citation115 Alternatively, signaling via the putative GPR55 receptor in ovarian and prostate cells activates ERK and AKT signaling pathways, which alters intracellular calcium signaling, thereby affecting cellular proliferation.Citation116

In addition to regulation through the ceramide pathway, cannabinoids exhibit a direct effect on cAMP levels through the regulation of adenylate cyclase, downregulation of protein kinase A, and a decrease in gene transcription.Citation14,Citation68,Citation117 In hormone-responsive cancer cells, this leads to decreases in the expression of breast cancer-associated antigen 1, prostate-specific antigen, and the androgen receptor in breast and prostate cells, respectively.Citation25,Citation53 The downregulation of protein synthesis also results in a decrease in the expression of the high-affinity nerve-growth factor tyrosine-kinase receptor A and the prolactin receptor, thereby decreasing cell sensitivity to key growth promoters.Citation69 In addition to cell-level effects through protein kinase A, cannabinoids regulate the action of hormones through the hypothalamic–pituitary–gonadal axis. For example, exposing rats to ΔCitation9-THC (1 mg/kg, intravenously) or WIN-55,212-2 (0.5 mg/kg, intravenously) resulted in a decrease in the secretion of luteinizing hormone within 30 minutes.Citation118 This effect was blocked by the preadministration of the CB1 antagonist SR141716.Citation118 Similar results have been reported for prolactin release, while levels of adrenocorticotropic hormone have been shown to increase following ΔCitation9-THC.Citation119,Citation120 Normal anterior pituitary tissue expresses cannabinoid receptors, which indicates that the cannabinoids may be able to exert modulatory actions directly at the level of the pituitary gland.Citation121 This further complicates the results that may occur in in vivo investigations of hormone-responsive tumors. However, the decrease in luteinizing hormone may suggest a concomitant decrease in steroid-hormone production that may be protective, especially in breast cancer.Citation122

In glioma cells, there is evidence that cannabinoids, specifically cannabidiol, activate apoptosis independently of cannabinoid-receptor binding. The mechanism for this effect is likely to involve the induction of oxidative stress through the generation of reactive oxygen species. This concept is supported by the observation that the antiproliferative effect was not blocked by a CB2 antagonist, but was inhibited by tocopherol, a potent antioxidant. This effect was not observed in noncancerous primary glial cells.Citation123

Effects on invasion and metastasis

Cannabinoids affect a wide range of markers associated with the invasion and metastasis of cancers, including markers of migration, adhesion, invasion, and metastasis itself.Citation124 For example, studies on migration have shown that AEA, 2-AG, cannabidiol, HU-210, JWH-133, Met-fluoro-AEA (Met-F-AEA), ΔCitation9-THC, and WIN-55,212-2 all decreased migration, or markers of migration, in a wide range of cell lines. Joseph et al reported that the adrenalin-induced migration of SW480 and MDA-MB-468 cells was inhibited following exposure to AEA or JWH-133 at 40 and 10 nM, respectively.Citation125 A similar effect was reportedly observed following treatment with HU-210, although detail was not provided.Citation125 In T lymphocytes, only JWH-133 (10 nM) was able to reduce stromal cell-derived factor 1-induced migration, with AEA (10 nM) showing no effect.Citation125 These results have been corroborated in cervical cells exposed to 2-AG or WIN-55,212-2, with both compounds reducing scratch closure in monolayers of SW756 cells.Citation126 Cannabidiol (≥3 μM) inhibited the migration of U87 glioma cells in a Boyden chamber assay, but interestingly this effect was not blocked by the cannabinoid receptor antagonists SR141716 (CB1) or SR144528 (CB2).Citation127 This suggests that some, if not all, the migration effects observed in cell systems may not be mediated by the cannabinoid receptor.

The adhesion of cells within the extracellular matrix is an important component of maintaining correct multicellular structure, with dysfunction of this process associated with metastasis. In a model of metastatic spreading using MDA-MB-231 cells, Met-F-AEA (0.5 mg/kg every 72 hours for 21 days) significantly reduced the number and size of metastatic nodes, an effect antagonized by SR141716.Citation128 The authors were able to demonstrate that the effect on metastasis was linked to the phosphorylation of two tyrosine-kinase proteins involved in migration and adhesion: focal adhesion kinase (FAK) and Src.Citation128 This contradicts previous results in neuroblastoma cells, where HU-210 at 10 nM caused phosphorylation of the FAK-related nonkinase but not FAK itself.Citation129 However, WIN-55,212-2 (20 μM) also modulated a range of adhesion proteins, including intercellular adhesion molecule 1, IL-1, vascular cell-adhesion protein 1, and IL-8.Citation130 The authors conclude that this inhibition of the IL-1 pathway is mediated by effects of WIN-55,212-2 on nuclear factor κB transactivation. This inhibition is likely to mediate a range of downstream effects, including anti-inflammatory and anticancerous actions.Citation130

Tissue inhibitors of matrix metalloproteinases (TIMPs) may be a key mechanism by which cannabinoids inhibit tissue invasion. Decreased TIMP expression is highly correlated with cancer invasiveness, and the expression of TIMP-1 is a potent suppressor of tumor growth and angiogenesis.Citation131,Citation132 However, the effect of cannabinoids on TIMP expression levels is controversial. Several studies have documented an increase in TIMP-1 levels following cannabinoid administration. In lung and cervical cell lines (A549 and HeLa, C33A, respectively), ΔCitation9-THC at 0.01 μM increased the expression of TIMP-1, which correlated with a decrease in cell invasion.Citation133 This effect was blocked by the preadministration of the specific inverse agonists AM-251 (CB1) or AM-630 (CB2).Citation133 However, ΔCitation9-THC decreased TIMP-1 expression in C6.9 and C6.4 glioma cell lines, a result that was confirmed in biopsies from patients with recurrent glioblastoma multiforme tumors undergoing a clinical trial of THC efficacy.Citation134 JWH-133 also downregulated TIMP-1 expression in glioma cells.Citation134 This suggests that the effects of cannabinoids on invasion may prove to be cancer- and cannabinoid-specific.

In terms of migration, cannabidiol, Met-F-AEA, ΔCitation9-THC, and WIN-55,212-2 have all proven to have direct effects on migration markers. In breast cancer cells, cannabidiol decreased lung metastasis of MDA-MB-231 cells,Citation135 while Met-F-AEA inhibited the migration of this cell line on type IV collagen.Citation128 This effect was also seen in the TSA-E1 murine breast cancer cell line and was antagonized by the administration of SR141716.Citation128 Similarly, ΔCitation9-THC (1–20 μM) inhibited the epidermal growth factor-induced growth, chemotaxis, and chemoinvasion of the lung cancer cell lines A549 and SW1573.Citation136 Finally, WIN-55,212-2 (50 μg/day, daily for 8 days) reduced the metastasis of B16 melanoma cells to the lung and liver in a nude mouse implantation model.Citation56

All these results suggest that overall the cannabinoids affect multiple cellular signaling pathways, which means they have the potential to decrease cancer development, growth, and metastasis. However, there are likely to be both cancer- and cannabinoid-specific elements to these effects. The final role of the cannabinoid receptors (CB1 and CB2) versus novel receptors (eg, GPR55) is also likely to be of ongoing importance.

Effects on angiogenesis

Angiogenesis is critical for tumor development, and many anticancer agents are selected for their antiangiogenic properties. In vivo models show tumors from cannabinoid-treated animals have a decreased number of sprouting blood vessels, reduced vascular networks, and small, undifferentiated intratumoral blood vessels.Citation103,Citation137,Citation138 Cannabinoids may produce a dual attack on the development of tumor blood vessels, through the inhibition of proangiogenic regulators, such as vascular endothelial growth factor (VEGF), and through a direct effect on endothelial cells.Citation103

A number of experiments have determined that the levels of the major vascularization factors, including VEGF, are downregulated following administration of ΔCitation9-THC, Met-F-AEA, WIN-55,212-2, and JWH-133.Citation103 ΔCitation9-THC decreased VEGF levels in lung cancer cell lines (A549 and SW1573), and this effect correlated with a decrease in vascularization of A549 xenoplantation tumors in severe combined immunodeficient mice.Citation136 Met-F-AEA decreased the production of VEGF and the expression of its receptor, VEGFR-1, in K-ras-transformed thyroid cells.Citation138 WIN-55,212-2 and JWH-133 both showed inhibitory effects on VEGF and related markers of angiogenesis in skin carcinoma tumors (implanted PDV.C57 cells).Citation99 JWH-133 downregulated VEGF in subcutaneously implanted glioma cells, and caused a concomitant decrease in the associated compounds connective tissue growth factor, heme oxygenase 1, Id-3, midkine, and Tie-1.Citation139 Overall, cannabinoids appear to have consistent effects on the vascularization pathway, causing a decrease in tumor vascularization in in vivo models.

The endothelial cell lines human umbilical vein endothelial cells (HUVEC) and ECV304 showed direct susceptibility to WIN-55,212-2, with exposure to concentrations above 25 nM inducing cell death.Citation103 This effect was repeatable on exposure to other cannabinoids, including HU-210 (25 nM), ΔCitation9-THC (1 μM), or JWH-133 (25 nM).Citation103 Similarly, the administration of synthetic cannabinoid analogues (LYR-7 or LYR-8) both decreased HUVEC viability at concentrations of 5 μM and above.Citation140 However, this effect was not blocked by the preincubation of cells with the specific inhibitors AM281 (CB1) or AM630 (CB2).Citation140 Therefore, the cannabinoids show significant potential as antiangiogenic agents, and this may prove key to their success as a clinical therapy, but the role of the cannabinoid receptors in this response is still to be fully elucidated.

The future of cannabinoid compounds in cancer treatment

Overall, the cannabinoids may show future promise in the treatment of cancer, but there are many significant hurdles to be overcome. There is much still to be learned about the action of the cannabinoids and the endocannabinoid system. The current disagreements in the literature suggest gaps remain in the knowledge base around the normal signaling pathways used by endocannabinoids, the physiological systems that are involved, and the range of effects that these compounds cause. Future research will help clarify the actions of the cannabinoids, and particularly the endocannabinoid signaling pathway, which will be critical in the ongoing development of these compounds.

It is a distinct possibility that the cannabinoids may have a place in the future treatment of cancer. Several reports have shown that the synthetic cannabinoids in particular have the potential to show sufficient specificity and efficacy to be precursors to clinical treatments. However, at this point in time, the results from studies are lacking sufficient depth of understanding to allow this transition to occur. The contradictory nature of reports around the efficacy of compounds highlights our lack of detailed understanding of mechanisms of action. The resolution of the conflicting evidence around cannabinoid action will continue to be a research priority in the near future, and it is expected that developing a more robust understanding of the mechanisms of action underlying cannabinoid action will facilitate the acceptance of cannabinoid use in a clinical setting.

Disclosure

The authors declare they have no conflict of interest, financial or otherwise, regarding this work.

Unknown widget #61484a86-c382-427f-bdec-2a5847160d2f

of type scholix-links

References

  • AlexanderASmithPFRosengrenRJCannabinoids in the treatment of cancerCancer Lett2009285161219442435
  • CameronLWilliamsJCannabis, alcohol and cigarettes: substitutes or complements?Econ Rec2001772361934
  • GaoniYMechoulamRIsolation, structure, and partial synthesis of an active constituent of hashishJ Am Chem Soc196486816461647
  • DevaneWHanusLBreuerAIsolation and structure of a brain constituent that binds to the cannabinoid receptorScience19922585090194619491470919
  • Console-BramLMarcuJAboodMECannabinoid receptors: nomenclature and pharmacological principlesProg Neuropsychopharmacology Biol Psychiatry2012381415
  • PiomelliDArachidonic acid in cell signalingCurr Opin Cell Biol1993522742807685181
  • MechoulamRFrideEDi MarzoVEndocannabinoidsEur J Pharmacol199835911189831287
  • MaccarroneMFinazzi-AgròAEndocannabinoids and their actionsVitam Horm20026522525512481549
  • DevaneWADysarzFAJohnsonMRMelvinLSHowlettACDetermination and characterization of a cannabinoid receptor in rat brainMol Pharmacol19883456056132848184
  • MatsudaLALolaitSJBrownsteinMJYoungACBonnerTIStructure of a cannabinoid receptor and functional expression of the cloned cDNANature199034662845615642165569
  • MunroSThomasKLAbu-ShaarMMolecular characterization of a peripheral receptor for cannabinoidsNature1993365644161657689702
  • PertweeRGCannabinoid pharmacology: the first 66 yearsBr J Pharmacol2006147Suppl 1S163S17116402100
  • GuzmánMCannabinoids: potential anticancer agentsNat Rev Cancer200331074575514570037
  • SarfarazSAdhamiVMSyedDNAfaqFMukhtarHCannabinoids for cancer treatment: progress and promiseCancer Res200868233934218199524
  • LangeJHKruseCGRecent advances in CB1 cannabinoid receptor antagonistsCurr Opin Drug Discov Devel200474498506
  • BergmanJDelatteMSParonisCAVemuriKThakurGAMakriyannisASome effects of CB1 antagonists with inverse agonist and neutral biochemical propertiesPhysiol Behav2008934–566667018076956
  • RossRABrockieHCStevensonLAAgonist-inverse agonist characterization at CB1 and CB2 cannabinoid receptors of L759633, L759656 and AM630Br J Pharmacol1999126366567210188977
  • McPartlandJMGlassMPertweeRGMeta-analysis of cannabinoid ligand binding affinity and receptor distribution: interspecies differencesBr J Pharmacol2007152558359317641667
  • BrownAJNovel cannabinoid receptorsBr J Pharmacol2007152556757517906678
  • PertweeRGHowlettACAboodMEInternational Union of Basic and Clinical Pharmacology. LXXIX. Cannabinoid receptors and their ligands: beyond CB1 and CB2Pharmacol Rev201062458863121079038
  • GodlewskiGKunosGOverview of nonclassical cannabinoid receptorsAboodMESorensenRGStellaNEndocannabinoids: Actions at Non-CB1/CB2 Cannabinoid ReceptorsNew YorkSpringer2013327
  • MarzoVPetrocellisLTemperature-sensitive transient receptor potential channels as ionotropic cannabinoid receptorsAboodMESorensenRGStellaNEndocannabinoids: Actions at Non-CB1/CB2 Cannabinoid ReceptorsNew YorkSpringer2013175197
  • OddiSFezzaFPasquarielloNEvidence for the intracellular accumulation of anandamide in adiposomesCell Mol Life Sci200865584085018213445
  • Rodríguez De FonsecaFDel ArcoIBermudez-SilvaFJBilbaoACippitelliANavarroMThe endocannabinoid system: physiology and pharmacologyAlcohol Alcohol200540121415550444
  • De PetrocellisLMelckDPalmisanoAThe endogenous cannabinoid anandamide inhibits human breast cancer cell proliferationProc Natl Acad Sci U S A19989514837583809653194
  • FonsecaBMCostaMAAlmadaMCorreia-da-SilvaGTeixeiraNAEndogenous cannabinoids revisited: a biochemistry perspectiveProstaglandins Other Lipid Mediat20131021031330
  • UedaNPuffenbargerRAYamamotoSDeutschDGThe fatty acid amide hydrolase (FAAH)Chem Phys Lipids20001081–210712111106785
  • GulyasAICravattBFBraceyMHSegregation of two endocannabinoid-hydrolyzing enzymes into pre- and postsynaptic compartments in the rat hippocampus, cerebellum and amygdalaEur J Neurosci200420244145815233753
  • WooAYWangTBZengXStereochemistry of an agonist determines coupling preference of β2-adrenoceptor to different G proteins in cardiomyocytesMol Pharmacol200975115816518838481
  • GlassMNorthupJKAgonist selective regulation of G proteins by cannabinoid CB1 and CB2 receptorsMol Pharmacol19995661362136910570066
  • TuruGHunyadyLSignal transduction of the CB1 cannabinoid receptorJ Mol Endocrinol2010442758519620237
  • HowlettACBreivogelCSChildersSRDeadwylerSAHampsonREPorrinoLJCannabinoid physiology and pharmacology: 30 years of progressNeuropharmacology200447Suppl 134535815464149
  • AboodMEMartinBRNeurobiology of marijuana abuseTrends Pharmacol Sci19921352012061604713
  • WalkerJMHuangSMCannabinoid analgesiaPharmacol Ther200295212713512182960
  • FreundTFKatonaIPiomelliDRole of endogenous cannabinoids in synaptic signalingPhysiol Rev20038331017106612843414
  • BreivogelCSChildersSRThe functional neuroanatomy of brain cannabinoid receptorsNeurobiol Dis199856 Pt B4174319974175
  • PatelSHillardCJCannabinoid CB1 receptor agonists produce cerebellar dysfunction in miceJ Pharmacol Exp Ther2001297262963711303052
  • PertweeRGWickensAPEnhancement by chlordiazepoxide of catalepsy induced in rats by intravenous or intrapallidal injections of enantiomeric cannabinoidsNeuropharmacology19913032372441649415
  • MonoryKBlaudzunHMassaFGenetic dissection of behavioural and autonomic effects of Δ9-tetrahydrocannabinol in micePLoS Biol2007510e26917927447
  • TurcotteDLe DorzeJAEsfahaniFFrostEGomoriANamakaMExamining the roles of cannabinoids in pain and other therapeutic indications: a reviewExpert Opin Pharmacother2010111173120001426
  • DrewLJHarrisJMillnsPJKendallDAChapmanVActivation of spinal cannabinoid 1 receptors inhibits C-fibre driven hyperexcitable neuronal responses and increases [35S]GTPγS binding in the dorsal horn of the spinal cord of noninflamed and inflamed ratsEur J Neurosci20001262079208610886347
  • HohmannAGBrileyEMHerkenhamMPre- and postsynaptic distribution of cannabinoid and mu opioid receptors in rat spinal cordBrain Res19998221–2172510082879
  • KleinTWNewtonCLarsenKThe cannabinoid system and immune modulationJ Leukoc Biol200374448649612960289
  • AshtonJCThe use of knockout mice to test the specificity of antibodies for cannabinoid receptorsHippocampus201222364364421538668
  • BornerCHolltVKrausJActivation of human T cells induces upregulation of cannabinoid receptor type 1 transcriptionNeuroimmunomodulation200714628128618287809
  • GaliègueSMarySMarchandJExpression of central and peripheral cannabinoid receptors in human immune tissues and leukocyte subpopulationsEur J Biochem1995232154617556170
  • KleinTWLaneBNewtonCAFriedmanHThe cannabinoid system and cytokine networkProc Soc Exp Biol Med200022511810998193
  • TanasescuRConstantinescuCSCannabinoids and the immune system: an overviewImmunobiology2010215858859720153077
  • PisantiSMalfitanoAMGrimaldiCUse of cannabinoid receptor agonists in cancer therapy as palliative and curative agentsBest Pract Res Clin Endocrinol Metab200923111713119285265
  • BowlesDWO’BryantCLCamidgeDRJimenoAThe intersection between cannabis and cancer in the United StatesCrit Rev Oncol Hematol201283111022019199
  • TodaroBCannabinoids in the treatment of chemotherapy-induced nausea and vomitingJ Natl Comp Canc Netw2012104487492
  • MunsonAEHarrisLSFriedmanMADeweyWLCarchmanRAAntineoplastic activity of cannabinoidsJ Natl Cancer Inst19755535976021159836
  • SarfarazSAfaqFAdhamiVMMukhtarHCannabinoid receptor as a novel target for the treatment of prostate cancerCancer Res20056551635164115753356
  • QamriZPreetANasserMWSynthetic cannabinoid receptor agonists inhibit tumor growth and metastasis of breast cancerMol Cancer Ther20098113117312919887554
  • BouaboulaMRinaldiMCarayonPCannabinoid-receptor expression in human leukocytesEur J Biochem199321411731808508790
  • BlázquezCCarracedoABarradoLCannabinoid receptors as novel targets for the treatment of melanomaFASEB J200620142633263517065222
  • WangDWangHNingWBacklundMGDeySKDuBoisRNLoss of cannabinoid receptor 1 accelerates intestinal tumor growthCancer Res200868156468647618676872
  • XuXLiuYHuangSOverexpression of cannabinoid receptors CB1 and CB2 correlates with improved prognosis of patients with hepatocellular carcinomaCancer Genet Cytogenet20061711313817074588
  • SarnataroDPisantiSSantoroAThe cannabinoid CB1 receptor antagonist rimonabant (SR141716) inhibits human breast cancer cell proliferation through a lipid raft-mediated mechanismMol Pharmacol20067041298130616822929
  • WasikAMChristenssonBSanderBThe role of cannabinoid receptors and the endocannabinoid system in mantle cell lymphoma and other non-Hodgkin lymphomasSemin Cancer Biol201121531332122024769
  • SánchezCde CeballosMLGomez del PulgarTInhibition of glioma growth in vivo by selective activation of the CB2 cannabinoid receptorCancer Res200161155784578911479216
  • McKallipRJNagarkattiMNagarkattiPSΔ-9-Tetrahydrocannabinol enhances breast cancer growth and metastasis by suppression of the antitumor immune responseJ Immunol200517463281328915749859
  • Rinaldi-CarmonaMLe DuigouAOustricDModulation of CB1 cannabinoid receptor functions after a long-term exposure to agonist or inverse agonist in the Chinese hamster ovary cell expression systemJ Pharmacol Exp Ther19982873103810479864290
  • LeterrierCBonnardDCarrelDRossierJLenkeiZConstitutive endocytic cycle of the CB1 cannabinoid receptorJ Biol Chem200427934360133602115210689
  • CastanedaJHaruiAKiertscherSRothJRothMDifferential expression of intracellular and extracellular CB2 cannabinoid receptor protein by human peripheral blood leukocytesJ Neuroimmune Pharmacol20138132333223299999
  • BifulcoMLaezzaCPisantiSGazzerroPCannabinoids and cancer: pros and cons of an antitumour strategyBr J Pharmacol2006148212313516501583
  • MassiPSolinasMCinquinaVParolaroDCannabidiol as potential anticancer drugBr J Clin Pharmacol201375230331222506672
  • GuindonJHohmannAGThe endocannabinoid system and cancer: therapeutic implicationBr J Pharmacol201116371447146321410463
  • CaffarelMMAndradasCPérez-GómezEGuzmánMSánchezCCannabinoids: a new hope for breast cancer therapy?Cancer Treat Rev201238791191822776349
  • TakedaSYamamotoIWatanabeKModulation of Δ9-tetrahydrocannabinol-induced MCF-7 breast cancer cell growth by cyclooxygenase and aromataseToxicology20092591–2253219428940
  • McAllisterSDChristianRTHorowitzMPGarciaADesprezPYCannabidiol as a novel inhibitor of Id-1 gene expression in aggressive breast cancer cellsMol Cancer Ther20076112921292718025276
  • Von BuerenAOSchlumpfMLichtensteigerWDelta(9)-tetrahydrocannabinol inhibits 17β-estradiol-induced proliferation and fails to activate androgen and estrogen receptors in MCF7 human breast cancer cellsAnticancer Res2008281A858918383828
  • CaffarelMMAndradasCMiraECannabinoids reduce ErbB2-driven breast cancer progression through Akt inhibitionMol Cancer2010919620649976
  • SánchezMGRuiz-LlorenteLSánchezAMDíaz-LaviadaIActivation of phosphoinositide 3-kinase/PKB pathway by CB1 and CB2 cannabinoid receptors expressed in prostate PC-3 cells. Involvement in Raf- 1 stimulation and NGF inductionCell Signal200315985185912834810
  • SánchezMGSánchezAMRuiz-LlorenteLDíaz-LaviadaIEnhancement of androgen receptor expression induced by (R)-methanandamide in prostate LNCaP cellsFEBS Lett2003555356156614675774
  • MimeaultMPommeryNWattezNBaillyCHénichartJPAntiproliferative and apoptotic effects of anandamide in human prostatic cancer cell lines: implication of epidermal growth factor receptor downregulation and ceramide productionProstate200356111212746841
  • NithipatikomKIsbellMAEndsleyMPWoodliffJECampbellWBAnti-proliferative effect of a putative endocannabinoid, 2- arachidonylglyceryl ether in prostate carcinoma cellsProstaglandins Other Lipid Mediat2011941–2344321167293
  • JacobssonSOPRongårdEStridhMTigerGFowlerCJSerum-dependent effects of tamoxifen and cannabinoids upon C6 glioma cell viabilityBiochem Pharmacol200060121807181311108795
  • McAllisterSChanCTaftRCannabinoids selectively inhibit proliferation and induce death of cultured human glioblastoma multiforme cellsJ Neurooncol2005741314016078104
  • MassiPVaccaniACerutiSColomboAAbbracchioMPParolaroDAntitumor effects of cannabidiol, a nonpsychoactive cannabinoid, on human glioma cell linesJ Pharmacol Exp Ther2004308383884514617682
  • JacobssonSOPWallinTFowlerCJInhibition of rat C6 glioma cell proliferation by endogenous and synthetic cannabinoids. Relative involvement of cannabinoid and vanilloid receptorsJ Pharmacol Exp Ther2001299395195911714882
  • CarracedoAGironellaMLorenteMCannabinoids induce apoptosis of pancreatic tumor cells via endoplasmic reticulum stress-related genesCancer Res200666136748675516818650
  • McKallipRJLombardCFisherMTargeting CB2 cannabinoid receptors as a novel therapy to treat malignant lymphoblastic diseaseBlood2002100262763412091357
  • HerreraBCarracedoADiez-ZaeraMGómez del PulgarTGuzmánMVelascoGThe CB2 cannabinoid receptor signals apoptosis via ceramide-dependent activation of the mitochondrial intrinsic pathwayExp Cell Res2006312112121213116624285
  • JiaWHegdeVLSinghNPDelta9-tetrahydrocannabinol-induced apoptosis in Jurkat leukemia T cells is regulated by translocation of Bad to mitochondriaMol Cancer Res20064854956216908594
  • McKallipRJJiaWSchlomerJWarrenJWNagarkattiPSNagarkattiMCannabidiol-induced apoptosis in human leukemia cells: a novel role of cannabidiol in the regulation of p22phox and Nox4 expressionMol Pharmacol200670389790816754784
  • LombardCNagarkattiMNagarkattiPSTargeting cannabinoid receptors to treat leukemia: role of cross-talk between extrinsic and intrinsic pathways in delta9-tetrahydrocannabinol (THC)-induced apoptosis of Jurkat cellsLeuk Res200529891592215978942
  • HerreraBCarracedoADiez-ZaeraMGuzmánMVelascoGp38 MAPK is involved in CB2 receptor induced apoptosis of human leukaemia cellsFEBS Lett2005579225084508816139274
  • FlygareJGustafssonKKimbyEChristenssonBSanderBCannabinoid receptor ligands mediate growth inhibition and cell death in mantle cell lymphomaFEBS Lett2005579306885688916337199
  • GustafssonKWangXSeveraDExpression of cannabinoid receptors type 1 and type 2 in non-Hodgkin lymphoma: growth inhibition by receptor activationInt J Cancer200812351025103318546271
  • HartSFischerOMUllrichACannabinoids induce cancer cell proliferation via tumor necrosis factor alpha-converting enzyme (TACE/ADAM17)-mediated transactivation of the epidermal growth factor receptorCancer Res20046461943195015026328
  • BarskySHRothMDKleerupECSimmonsMTashkinDPHistopathologic and molecular alterations in bronchial epithelium in habitual smokers of marijuana, cocaine, and/or tobaccoJ Natl Cancer Inst19989016119812059719080
  • RothMDMarques-MagallanesJAYuanMSunWTashkinDPHankinsonOInduction and regulation of the carcinogen-metabolizing enzyme CYP1A1 by marijuana smoke and 9-tetrahydrocannabinolAm J Respir Cell Mol Biol200124333934411245634
  • MarcusDMJaniABRossiPJPopulation-based case-control study of recreational drug use and testis cancer risk confirms an association between marijuana use and nonseminoma riskCancer20131196128423233231
  • D’SouzaGPalefskyJMZhongYMarijuana use is not associated with cervical human papillomavirus natural history or cervical neoplasia in HIV-seropositive or HIV-seronegative womenCancer Epidemiol Biomarkers Prev201019386987220160270
  • LiangCMcCleanMDMarsitCA population-based case-control study of marijuana use and head and neck squamous cell carcinomaCancer Prev Res (Phila)20092875976819638490
  • NithipatikomKEndsleyMPIsbellMA2-Arachidonoylglycerol: a novel inhibitor of androgen-independent prostate cancer cell invasionCancer Res200464248826883015604240
  • Ellert-MiklaszewskaAKaminskaBKonarskaLCannabinoids downregulate PI3K/Akt and Erk signalling pathways and activate proapoptotic function of Bad proteinCell Signal2005171253715451022
  • CasanovaMLBlázquezCMartínez-PalacioJInhibition of skin tumor growth and angiogenesis in vivo by activation of cannabinoid receptorsJ Clin Invest20031111435012511587
  • GustafssonKChristenssonBSanderBFlygareJCannabinoid receptor-mediated apoptosis induced by R(+)-methanandamide and Win55,212-2 is associated with ceramide accumulation and p38 activation in mantle cell lymphomaMol Pharmacol20067051612162016936228
  • Fernández-RuizJRomeroJVelascoGTolónRMRamosJAGuzmánMCannabinoid CB2 receptor: a new target for controlling neural cell survival?Trends Pharmacol Sci2007281394517141334
  • Ellert-MiklaszewskaAGrajkowskaWGabrusiewiczKKaminskaBKonarskaLDistinctive pattern of cannabinoid receptor type II (CB2) expression in adult and pediatric brain tumorsBrain Res20071137116116917239827
  • BlázquezCCasanovaMLPlanasAInhibition of tumor angiogenesis by cannabinoidsFASEB J200317352953112514108
  • HannunYAObeidLMPrinciples of bioactive lipid signalling: lessons from sphingolipidsNat Rev Mol Cell Biol20089213915018216770
  • MoradSAFCabotMCCeramide-orchestrated signalling in cancer cellsNat Rev Cancer2013131516523235911
  • Galve-RoperhISánchezCCortésMLGómez del PulgarTIzquierdoMGuzmánMAnti-tumoral action of cannabinoids: Involvement of sustained ceramide accumulation and extracellular signal-regulated kinase activationNat Med20006331331910700234
  • WilliamsTMMedinaFBadanoICaveolin-1 gene disruption promotes mammary tumorigenesis and dramatically enhances lung metastasis in vivo: role of Cav-1 in cell invasiveness and matrix metalloproteinase (MMP-2/9) secretionJ Biol Chem200427949516305164615355971
  • SloanEKStanleyKLAndersonRLCaveolin-1 inhibits breast cancer growth and metastasisOncogene200423477893789715334058
  • VousdenKHLuXLive or let die: the cell’s response to p53Nat Rev Cancer20022859460412154352
  • DownerEJGowranAMurphyACCampbellVAThe tumour suppressor protein, p53, is involved in the activation of the apoptotic cascade by Δ9-tetrahydrocannabinol in cultured cortical neuronsEur J Pharmacol20075641–3576517379209
  • VelascoGSánchezCGuzmánMTowards the use of cannabinoids as antitumour agentsNat Rev Cancer201212643644422555283
  • ShrivastavaAKuzontkoskiPMGroopmanJEPrasadACannabidiol induces programmed cell death in breast cancer cells by coordinating the cross-talk between apoptosis and autophagyMol Cancer Ther20111071161117221566064
  • Gómez del PulgarTVelascoGSánchezCHaroAGuzmánMDe novo-synthesized ceramide is involved in cannabinoid-induced apoptosisBiochem J2002363Pt 118318811903061
  • SalazarMLorenteMGarcía-TaboadaEThe pseudokinase tribbles homologue-3 plays a crucial role in cannabinoid anticancer actionBiochim Biophys Acta Epub462013
  • SalazarMCarracedoASalanuevaIJTRB3 links ER stress to autophagy in cannabinoid antitumoral actionAutophagy2009571048104919652543
  • PineiroRMaffucciTFalascaMThe putative cannabinoid receptor GPR55 defines a novel autocrine loop in cancer cell proliferationOncogene201130214215220838378
  • BifulcoMMalfitanoAMPisantiSLaezzaCEndocannabinoids in endocrine and related tumoursEndocr Relat Cancer200815239140818508995
  • MurphyLLMuñozRMAdrianBAFunction of cannabinoid receptors in the neuroendocrine regulation of hormone secretionNeurobiol Dis1998564324469974176
  • de MiguelRRomeroJMuñozRMEffects of cannabinoids on prolactin and gonadotrophin secretion: involvement of changes in hypothalamic γ-aminobutyric acid (GABA) inputsBiochem Pharmacol19985610133113389825732
  • DeweyWLPengTCHarrisLSThe effect of 1-trans-Δ9-tetrahydrocannabinol on the hypothalamo-hypophyseal-adrenal axis of ratsEur J Pharmacol19701233823844322071
  • PagottoUMarsicanoGFezzaFNormal human pituitary gland and pituitary adenomas express cannabinoid receptor type 1 and synthesize endogenous cannabinoids: first evidence for a direct role of cannabinoids on hormone modulation at the human pituitary levelJ Clin Endocrinol Metab20018662687269611397872
  • BernsteinLRossRKEndogenous hormones and breast cancer riskEpidemiol Rev199315148658405212
  • MassiPVaccaniABianchessiSCostaBMacchiPParolaroDThe non-psychoactive cannabidiol triggers caspase activation and oxidative stress in human glioma cellsCell Mol Life Sci200663172057206616909207
  • HermansonDMarnettLCannabinoids, endocannabinoids, and cancerCancer Metastasis Rev2011303–459961222038019
  • JosephJNiggemannBZaenkerKEntschladenFAnandamide is an endogenous inhibitor for the migration of tumor cells and T lymphocytesCancer Immunol Immunother200453872372815034673
  • RudolphMIBozaYYefiRThe influence of mast cell mediators on migration of SW756 cervical carcinoma cellsJ Pharmacol Sci2008106220821818296861
  • VaccaniAMassiPColomboARubinoTParolaroDCannabidiol inhibits human glioma cell migration through a cannabinoid receptor-independent mechanismBr J Pharmacol200514481032103615700028
  • GrimaldiCPisantiSLaezzaCAnandamide inhibits adhesion and migration of breast cancer cellsExp Cell Res2006312436337316343481
  • ZhouDSongZHCB1 cannabinoid receptor-mediated tyrosine phosphorylation of focal adhesion kinase-related non-kinaseFEBS Lett20025251–316416812163181
  • CurranNMGriffinBDO’TooleDBradyKJFitzgeraldSNMoynaghPNThe synthetic cannabinoid R(+)WIN 55,212-2 inhibits the interleukin-1 signaling pathway in human astrocytes in a cannabinoid receptor-independent mannerJ Biol Chem200528043357973580616105834
  • KhokhaRWaterhousePYagelSAntisense RNA-induced reduction in murine TIMP levels confers oncogenicity on Swiss 3T3 cellsScience198924348939479502465572
  • ZacchignaSZentilinLMoriniMAAV-mediated gene transfer of tissue inhibitor of metalloproteinases-1 inhibits vascular tumor growth and angiogenesis in vivoCancer Gene Ther2004111738014681728
  • RamerRHinzBInhibition of cancer cell invasion by cannabinoids via increased expression of tissue inhibitor of matrix metalloproteinases-1J Natl Cancer Inst20081001596918159069
  • BlázquezCCarracedoASalazarMDown-regulation of tissue inhibitor of metalloproteinases-1 in gliomas: a new marker of cannabinoid antitumoral activity?Neuropharmacology200854123524317675107
  • LigrestiAMorielloASStarowiczKAntitumor activity of plant cannabinoids with emphasis on the effect of cannabidiol on human breast carcinomaJ Pharmacol Exp Ther200631831375138716728591
  • PreetAGanjuRKGroopmanJE[Delta]9-tetrahydrocannabinol inhibits epithelial growth factor-induced lung cancer cell migration in vitro as well as its growth and metastasis in vivoOncogene200727333934617621270
  • PisantiSBorselliCOlivieroOLaezzaCGazzerroPBifulcoMAntiangiogenic activity of the endocannabinoid anandamide: correlation to its tumor-suppressor efficacyJ Cell Physiol2007211249550317192847
  • PortellaGLaezzaCLaccettiPDe PetrocellisLDi MarzoVBifulcoMInhibitory effects of cannabinoid CB1 receptor stimulation on tumor growth and metastatic spreading: actions on signals involved in angiogenesis and metastasisFASEB J200317121771177312958205
  • BlázquezCGonzález-FeriaLÁlvarezLHaroACasanovaMLGuzmánMCannabinoids inhibit the vascular endothelial growth factor pathway in gliomasCancer Res200464165617562315313899
  • ThapaDLeeJSHeoSWNovel hexahydrocannabinol analogs as potential anti-cancer agents inhibit cell proliferation and tumor angiogenesisEur J Pharmacol20116501647120950604