Você está na página 1de 71

 

 
Cannabinoids and autoimmune diseases: A systematic review
Valeria Katchan, Paula David, Yehuda Shoenfeld
PII:
DOI:
Reference:

S1568-9972(16)30034-9
doi: 10.1016/j.autrev.2016.02.008
AUTREV 1826

To appear in:

Autoimmunity Reviews

Received date:
Accepted date:

27 January 2016
3 February 2016

Please cite this article as: Katchan Valeria, David Paula, Shoenfeld Yehuda, Cannabinoids and autoimmune diseases: A systematic review, Autoimmunity Reviews (2016), doi:
10.1016/j.autrev.2016.02.008

This is a PDF le of an unedited manuscript that has been accepted for publication.
As a service to our customers we are providing this early version of the manuscript.
The manuscript will undergo copyediting, typesetting, and review of the resulting proof
before it is published in its nal form. Please note that during the production process
errors may be discovered which could aect the content, and all legal disclaimers that
apply to the journal pertain.

ACCEPTED MANUSCRIPT
CANNABINOIDS AND AUTOIMMUNE DISEASES: A SYSTEMATIC REVIEW

IP

Valeria Katchan MD 1,3 ; Paula David 2,3, ; Yehuda Shoenfeld MD, FRCP, MaACR 3, 4

CR

1 Departamento de Medicina Interna, Hospital La Paz, Madrid, Spain


2 Faculdade de Cincias Mdicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro,

NU
S

Brasil

3 Zabludowicz Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer,

MA

Israel

PT

ED

4 Sackler Faculty of Medicine, Tel-Aviv University, Israel;

Corresponding author: Yehuda Shoenfeld MD, FRCP (Hon.), MaCR, Head of Zabludowicz

CE

Center for Autoimmune Diseases, Chaim Sheba Medical Center, Tel-Hashomer 52621, Israel. E-

AC

mail address: shoenfel@post.tau.ac.il


Tel: 972-3-5308070, 972-52-6669020

Key Words: Cannabinoids, Endocannabinoid system, Autoimmune Diseases, Immunosuppression, Tetrahydrocannabinol (THC), Type 1 Diabetes, Neuropathic Pain, Fibromyalgia, Inflammatory Bowel Disease, Multiple Sclerosis, Scleroderma, Rheumatoid Arthritis
ABSTRACT

ACCEPTED MANUSCRIPT
Cannabinoids have shown to have a variety effects on body systems. Through CB1 and CB2 receptors, amongst other, they exert an effect by modulating neurotransmitter and cytokine release.
Current research in the role of cannabinoids in the immune system shows that they possess im-

munosuppressive properties. They can inhibit proliferation of leucocytes, induce apoptosis of T

IP

cells and macrophages and reduce secretion of pro-inflammatory cytokines. In mice models,

CR

they are effective in reducing inflammation in arthritis, multiple sclerosis, have a positive effect

NU
S

on neuropathic pain and in type 1 diabetes mellitus. They are effective as treatment for fibromyalgia and have shown to have anti-fibrotic effect in scleroderma. Studies in human models
are scarce and not conclusive and more research is required in this field. Cannabinoids can be

MA

therefore promising immunosuppressive and anti-fibrotic agents in the therapy of autoimmune

AC

CE

PT

ED

disorders.

ACCEPTED MANUSCRIPT
1. Introduction
Cannabinoids are diverse chemical compounds that can bind to receptors of our endocannabinoid

system. Cannabinoids can be endocannabinoids, when produced by our own body, phytocanna-

IP

binoids, present naturally in the plant Cannabis sativa [1,2], and synthetic cannabinoids. The

CR

most well-known cannabinoid is 9-tetrahydrocannabinol (9-THC), the main constituent of


cannabis. Nabilone, a synthetic analogue of 9-THC has been licensed for medical use [2]. En-

NU
S

docannabinoid system and the physiological effects of cannabinoids have been widely studied in
the past decades. Cannabinoids are believed to have immunomodulatory effects and therefore
their potential role as an autoimmune/inflammatory treatment has been extensively investigated.

MA

Herewith, we revise the properties and the effects of cannabinoids on autoimmune disorders

ED

known so far.

PT

2. The Endocannabinoid System

CE

2.1 Cannabinoid Receptor Agonists


Cannabinoids receptor agonists are very heterogeneous and can be divided into four groups ac-

AC

cording to the difference in their chemical structure. These are classical, non-classical, aminoalkylindole and eicosanoid compounds groups. [1,2]
Briefly, the classical group consists of phytocannabinoids (9-THC, cannabinol) and their synthetic analogues. The eicosanoid group are mostly endocannabinoids, produced by our own cells.
These are arachidonoylethanolamide (anandamide),O-arachidonoylethanolamine (virodhamine),
2-arachidonoyl glycerol and 2-arachidonyl glyceryl ) and several synthetic analogues of anandamide. The two other groups, non-classical and aminoalkylindole consist of synthetic cannabinoids. (See Table 1)
Cannabinoids act on the two main receptors of the endocannabinoid system, CB1 and CB2 receptors. Every endocannabinoid agonist has a different affinity for the receptors. For example,

ACCEPTED MANUSCRIPT
9-THC has a higher affinity for CB1 receptor and the phytocannabinoid cannabinol is an agonist without marked CB1/CB2 selectivity. (Fig 1)

2.2 Cannabinoid RECEPTORS

IP

2.21 CB1 and CB2 receptors

CR

There are two types of cannabinoid receptor that have been mostly identified and studied. These
are CB1 and CB2 receptors. They belong to a class of cell membrane receptors under the G pro-

NU
S

tein-coupled receptor superfamily and consist of seven transmembrane spanning domains. The
effect of binding is variable from partial agonism, functional selectivity to inverse agonism and

MA

all play important roles in determining the cellular response to specific cannabinoid receptor li-

ED

gands.

2.22 Mechanism of action of CB1 and CB2 receptors

PT

Both, CB1 and CB2 receptors produce their effects through heterotrimeric Gi/o proteins coupled

CE

to them. CB1 receptor activated exhibits its effects mainly through activation of G alpha i/o. Biding of CB1 to its agonist leads to inhibition of the production of adenylate cyclase enzyme. Con-

AC

sequently, the binding between CB1 and its ligands decrease the intracellular cAMP levels and
an increase the mitogen-activated protein kinase (MAP kinase). In some cases CB1 receptor activation may be coupled to Gs proteins, which stimulate adenylate cyclase. cAMP. [3,4]
CB1 and CB2 receptors are also coupled to a variety of ion channels in the cell membrane,
which are the positively influenced inwardly rectifying potassium channels (=Kir or IRK)(-) and
the calcium channels. These channels are activated when there is a cAMP dependent interaction
between the receptor and the molecules such as protein kinase A (PKA), protein kinase C(PKC),
Raf-1, ERK, JNK, p38, c-fos, c-jun, and others. [5]
In case of CB1, activation can produce a subsequent decrease of Ca2+ entry into the cell, without
involvement of cAMP, which is the requirement for neurotransmitter release. The overall effect

ACCEPTED MANUSCRIPT
is a decrease in the neurotransmitter release. CB1 receptor is therefore a pre-synaptic receptor
that modulates neurotransmitter release when activated in a dose-dependent manner [6].
CB1 and CB2 cannabinoid receptors also act to regulate the phosphorylation and activation of

IP

different members of the family of mitogen- activated- protein kinases (MAPKs), including
extracellular signal regulated kinase-1 and -2 (ERK1/2), p38 MAPK and c-Jun N-terminal kinase

NU
S

and apoptosis as well as glucose metabolism [6].

CR

(JNK). MAPK, in turn controls gene expression related to cell proliferation, motility, adhesion

CB1 and CB 2 receptors produce their effects through the stimulation by their agonists.

MA

(endogenous/ exogenous/ synthetic). After release, agonist molecules are rapidly deactivated by
uptake into cells and metabolized. Metabolism of anandamide and 2-AG occurs by either enzymatic hydrolysis with a help of fatty acid amide hydrolase enzyme (FAAH) [7,8] or other meta-

ED

bolic processes, including hydrolysis of 2-AG by monoglyceride lipase [9].

PT

The mechanism is summarized in Figure 2.


2.23 Distribution of cannabinoid receptors:

CE

CB1 receptors are expressed by central and peripheral neurons and also by some non-neuronal

AC

cells [1,2, 10]. Within the central nervous system, the distribution pattern of CB1 receptors is
heterogeneous and can be related to their function. CB1 receptors are densely distributed in cerebral cortex, hippocampus, caudate-putamen, substantia nigra pars reticulata, globus pallidus,
entopeduncular nucleus and cerebellum, as well as in other areas of the brain and spinal cord.
They process or modulate nociceptive information.

Distribution within the central nervous

system might be related to CB1 receptor agonists' ability to impair cognition and memory, to alter the control of motor function and to produce anti nociception [1, 10, 11,12 ].
Some CB1 receptors are located on central and peripheral nerve terminal, where they modulate
the release of excitatory and inhibitory neurotransmitters when activated [1, 2].CB2 receptors are
expressed mainly on immune cells [ 2, 10] and have a role in immunomodulation.

ACCEPTED MANUSCRIPT
Both, CB1 and CB2 receptors share the ability to modulate the release of chemical messengers.
By acting on CB1 receptors, cannabinoids interact with a multitude neurotransmitters at the CNS
and can modulate their release. [13]. CB2 control the release of inflammatory cytokines, regulat-

ing the immune system.

IP

2.3 Other cannabinoid receptors of the endocannabinoid system. Cannabinoid Non- CB1 Non-

CR

CB2 receptors

NU
S

2.3a Vanilloid receptors:

One of the non CB1/CB2 receptors described, with binding capacity to cannabinoids is TRVP1
receptor, also called capsaicin receptor. It is a non-selective cation channel that is present on

MA

sensory neurons in tissues such as skin, heart, blood vessels and lung. TRPV1 receptors are
found mainly in the nociceptive neurons of the peripheral nervous system, but they have also

ED

been described in many other tissues, including the central nervous system. TRPV1 is involved

PT

in the transmission and modulation of pain (nociception) [35,14,15] through sensory primary afferent and perivascular neurons. It has been shown that activation of TRPV1 by the endogenous

CE

cannabinoid anandamide leads to a release of substance P (SP) and of calcitonin gene-related


(CGRP), which have allogenic and local vasodilatory and pro-inflammatory effects, as well as

AC

beneficial actions such as cardio protection and anti-hypertensive effects. [16-22]


2.3b Non-CB1, Non-CB2, non- Vanilloid receptors.
It has been shown by successive experiments that many biological effects of the cannabinoids
are not reversed by CB1 / CB2 antagonists. Additional receptor pathways, including the peroxisome proliferator-activated receptors (PPARs), G-protein receptor 55 (GPR55) as well as nicotine, 5-HT3 and adenosine A2A receptors, have been involved in cannabinoid signal transduction. [23,24]
2.3c Allosteric sites for cannabinoids
Apart from different receptors, there is an evidence for the presence of allosteric sites for anandamide and other cannabinoids on several non-cannabinoid receptors [1,26 ]. These are 5-HT2

ACCEPTED MANUSCRIPT
receptors [27], 5-HT3 receptors [28,29,30,31], 1-adrenoceptors, M1 and M4 muscarinic receptors [32] and -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) GLUA1 and
GLUA3 glutamate receptors [33,34]The biological consequences of occupation of the proposed

allosteric sites on 5-HT2 receptors (by HU-210) and on M1 and M4 receptors (by anandamide,

CR

IP

methanandamide and SR141716A) have not been determined yet.[1]

NU
S

2.4 Effects of cannabinoids on body systems. (See Table 2)

The effects of cannabinoids include analgesia, muscle relaxation, immunosuppression, antiinflammation, anti-allergic effects, sedation, improvement of mood, stimulation of appetite, anti-

MA

emesis, lowering of intraocular pressure, bronchodilation, neuroprotection and antineoplastic effects. [36]

ED

2.4a Cannabinoids and the Central Nervous System (CNS):

PT

Cannabinoids interact with different neurotransmitters and neuromodulators [36- 39], among
them acetylcholine, dopamine, -aminobutyric acid (GABA), histamine, serotonin, glutamate,

CE

norepinephrine, prostaglandins and opioid peptides. Some effects of THC of the body can be
explained by these interactions. For example, tachycardia and hypo salivation with dry mouth

AC

[40,41] are mediated by the effects of THC on the release and turn-over of acetylcholine [40].
The antiemetic properties of cannabinoid are explained by interactions with serotonin [42]. Therapeutic effects in movement and spastic disorders could be ascribed in part to interactions with
GABAergic, glutamergic and dopaminergic transmitters systems [43,44].
Neuroprotective cannabinoid effects observed in animal studies are based on inhibition of excessive glutamate production, inhibition of calcium influx into cells and the anti-oxidant properties
which reduce damage caused by oxygen radicals and modulation of vascular tone [36, 45-47].
The use of cannabinoids as a treatment of stroke and brain injury is therefore being investigated.

2.4b Cannabinoids and the circulatory system:

ACCEPTED MANUSCRIPT
Stimulating the endocannabinoid system with THC leads to tachycardia [36,48] and increased
cardiac output, cardiac labor and oxygen demand [49]. It can also produce peripheral vasodilation and orthostatic hypotension [50,51] mediated by central inhibition of the sympathicus sys-

tem, by activation of CB1 receptors [54]. It has been shown that endocannabinoids can be syn-

IP

thesized by vascular endothelium, circulating macrophages and platelets [52] and the subsequent

CR

activation of vascular cannabinoid CB1 receptors can decrease vascular resistance in the coro-

NU
S

nary arteries and the brain [53]


2.4c Cannabinoids, appetite and eating

Endocannabinoids control appetite though leptin signaling pathway in the hypothalamus [55].

MA

Leptin downregulates food intake by upregulating appetite-reducing neuropeptides and downregulating appetite-stimulating factors. . In animal model, reduced levels of leptin were associated

ED

with elevated levels of endocannabinoids in the hypothalamus [55]. Cannabinoid antagonists


have been tried to treat obesity but a selective loss of positive emotional memory in healthy vo-

PT

lunteers was noted. [62]

CE

Rest of the effects on osteoarticular, digestive, reproductive, ocular and immune system are re-

AC

sumed in Table 2.

3. Endocannabinoid System and its potential role in medicine


Several studies about cannabinoids system and their possible actions in medical purposes have
been done in the last decades. Cannabinoids have multiple physiological effects. Hence, modulation of endocannabinoids system, with agonists and antagonists, have been broadly studied as
potential novel treatments for different diseases. For example, Anandamide could increase food
intake in rats [63] and has been approved to increase weight in cancer and HIV+ patients, while
the antagonist SR-141716A inhibits the intake of food [64, 65, 66], being a potential anti-obesity
treatment, acting probably in the CB1 receptor in the hypothalamus [67]. They were already
shown to be effective in many medical conditions listed in Table 3.

ACCEPTED MANUSCRIPT
4. How cannabinoids are consumed
Cannabinoids can be consumed in several ways. However, the pharmacokinetics of 9-THC is
different in each manner [68]. Possible forms of administration are listed in Table 4, with their

advantages and limitations to being used for medical purposes. The most well-known and used

CR

NU
S

5. Cannabinoids and the immune system

IP

way of administration of cannabinoids is inhaling.

Immune cells express CB1 and CB2 receptors and can synthesize, secrete, transport and catabolize cannabinoids [69-73]. The expression of cannabinoids receptors is different in each immune

MA

cell. They are expressed, from the most abundant to the less extensive, on B cells, natural killer
cells (NK), monocytes, neutrophils, CD8 leucocytes and CD4 leucocytes. The level of expres-

ED

sion also depends on the immune stimulation and on the activation state of the cell [74].

PT

In mice, deficiency in the Fatty Acid Amide Hydrolase (FAAH), anandamide-catabolizing enzyme, elevates the levels of anandanamide (an endocannabinoid) in the CNS and periphery. In-

CE

creased levels of anandamide attenuate inflammatory responses, suggesting that the endocanna-

AC

binoids are physiologically involved in dampening the immune system [75-77].


The evidences that endocannabinoid system has probably immunomodulatory effects generated
the theory that exogenous cannabinoids might also have immunosuppressive effects, being a potential novel therapy to autoimmune and inflammatory disease. Since then, several studies about
this association have been published. [71, 78-80].
9-THC can modulate Th1/Th2 balance by enhancing Th2 and suppressing Th1 immune responses [81-84]. Although, the psychoactive effects of 9-THC limit its potential as a therapy.
Cannabidiol, on the other hand, has low affinity for the CB1 and CB2 receptors, resulting in no
psychoactive effects [85,86]. Besides, Cannabidiol is well tolerated without side effects when
chronically administered to humans [87, 88].

ACCEPTED MANUSCRIPT
Cannabinoids were shown, in vivo and in vitro, to exert their immunosuppressive properties
through 4 main pathways: induction of apoptosis, inhibition of cell proliferation, inhibition of

cytokines and chemokines production and induction of regulatory T cells (T regs) [89].

IP

5.1 Cannabinoids induce apoptosis

CR

Anandamide was shown to induce apoptosis in mitogen-induced T and B human lymphocytes,


human neuroblastoma CHP100 cells and lymphoma U937 cells, depending on the dose [90, 91].

NU
S

It was also observed that 9-THC can trigger apoptosis in murine macrophages and T cells,
through regulation of BCl2 and caspase activity [92].

MA

However, there is no demonstration of cannabinoids inducing apoptosis in vivo, perhaps because

through phagocytes [89]

ED

apoptosis detection in vivo is difficult since the apoptotic cells are rapidly and efficiently cleared

PT

9-THC injection in C57BL/6 mice (10 mg/kg body weight) was demonstrated to decrease the

CE

cellularity in the spleen and thymus, affecting several cell populations such as T cells, B cells
and macrophages. [93]. When administered at a lower concentration (10 M), 9-THC only in-

AC

duced AnnexinV + cells, which is indicative of early apoptotic cells, but at higher concentrations
(20 M), the splenocytes had both AnnexinV and PI positive, which represent late apoptotic as
well as necrotic cells.
9-THC may affect more intensively naive lymphocyte than activated lymphocytes. Levels of
apoptosis were shown to be greater in cultures treated only with THC, when compared to the
cultures that contained 9-THC and mitogen [93]. In the same study, it was noticed that activated lymphocytes can down-regulate the expression of CB2, decreasing their sensitivity to 9THC.

ACCEPTED MANUSCRIPT
Administration of CB2 antagonists block 9-THC-induced apoptosis in lymphocytes and thymocytes, while CB1 antagonists failed to show a significant effect, what suggest that 9-THC

induces apoptosis via CB2 receptors [93].

IP

Cannabidiol was also demonstrated to induce apoptosis in a time-and-dose dependent manner on

CR

CD4+ and CD8+ T cell populations, on murine thymocytes and on EL-4 cells. Cannabidiol lead
to apoptosis by producing reactive oxygen species (ROS) and activating caspase 8 and caspase 3.

NU
S

[94]
5.2 Cannabinoids inhibit immune cell proliferation

MA

While low doses of 9-THC stimulate T cell, high doses of 9-THC inhibited responses to LPS,
T cell mitogens and anti-CD3 antibody in both human and mice in vitro studies [95]. This bi-

ED

phasic role of cannabinoids has also been shown on B cells: a study demonstrated increased B

PT

cell proliferation in response to 9-THC [96], and, on the other hand, another study showed de-

CE

creased B cells response to LPS after cannabinoid treatment [95].


9-THC could suppress immune functions and increase susceptibility to infections in experi-

AC

mental models. The precise mechanism by which 9-THC suppress the immunity is still unclear.
The inhibition of lymphocytes proliferation in those cultures might be induced through direct
effects on immune cells, and not in CB1 and CB2, once it still observed in presence of CB1 and
CB2 antagonists [97]. 9-THC can also decreased proliferation of thymocytes. This effect was
shown to happen by inhibiting the calcium stabilization within the cell [98]. Low doses of anandamide were also able to inhibited proliferation of T and B lymphocytes after mitogen stimulation [90].
5.3 Cannabinoids inhibit cytokine and chemokine production and inflammatory cells migration
Cannabidiol inhibits IL1, IL12, TNF alfa and interferon-gama (INF-g) cytokine release by peripheral blood mononuclear cells, and enhances production of the Th2-associated cytokines, IL-4

ACCEPTED MANUSCRIPT
and IL-10 [99]. Cannabidiol can also reduce prostaglandin E2 and tissue cyclooxygenase (COX)
activity [P61]. 9-THC also changes destructive TH1 immunity to protective TH2 immunity, but

less effectively than Cannabidiol [70, 100-104]

IP

Cannabinoids can regulate monocytes differentiation to M1 or M2 macrophage phenotypes, as

CR

well as their ability to produce cytokines, chemokines and other immune mediators [105-112].
The responses to cannabinoids in the immune system are different in people who usually use

NU
S

consume cannabis and in the ones that are not. Phytocannabinoids can inhibit monocyte migration in individuals exposed to cannabis more than twice a week (non-nave), while they do not

MA

affect subjects that never tried or that were not exposed in the past 3 years (nave) [113]. In this
study, endocannabinoids and synthetic cannabinoids had no effect in both groups. The authors

ED

also demonstrated that monocytes isolated from cannabis non-nave subjects expressed fourfold
higher amounts of CB1 receptor mRNA than cannabis nave subjects, whereas CB2 receptor

PT

mRNA remained unchanged between the two cohorts. These results show that the sensitivity of

CE

monocytes isolated from non-nave subjects to phytocannabinoids correlates with enhanced expression of CB1 receptors.

AC

9-THC was also shown to have immunosuppressive effects in Legionella pneumophila (Lp)
infected dendritic cells. When the dendritic cells were pretreated with 9-THC, the immunization potential of Lp-loaded cells in mice was suppressed. 9-THC suppressed IL-12p40 production by the dendritic cells and inhibited expression of maturation markers such as MHCII, CD86,
and CD40 [114].
5.4 Cannabinoids induct regulatory T cells
9-THC can increase the number of Foxp3+ Treg cells. In a ConA-induced hepatitis model, 9THC was able to alleviate liver damage, it was not only by increasing levels of apoptosis in activated T cells, but also by inducing Treg cells to inhibit the cytokines induced in vivo by ConA

ACCEPTED MANUSCRIPT
[89]. More studies are still needed, but it suggests that Treg cells, unlike other T cells, may be
resistant to apoptosis induced by 9-THC and may suppress the activation of T cells that even-

tually escape from apoptosis. [89, 115]

IP

5.5 Other cannabinoids immunomodulatory effects

CR

Cannabis and certain cannabinoid receptor ligands have the ability to suppress serum immunoglobulin (Ig) levels [69, 70, 116]. 9-THC reduces B cell numbers while decreases IgG and IgM

NU
S

levels [116]. Although, this effect is controversial: some authors believe that there are no
changes in B cell levels [117].

MA

Cannabinoid receptor ligands can suppress spreading and phagocytosis, cytolysis, cytokine production, and antigen presentation in mouse peritoneal macrophages [118-124]. They also sup-

ED

press, in vivo and in vitro, NK cells and cytotoxic effector functions. [125-127].

PT

5.6 Cannabinoids as pro-inflammatory molecules?

CE

Despite all these data, some studies suggest that cannabinoids could also have pro-inflammatory
effects. [128,129]: promoting allergic reactions [130]; releasing inflammatory mediators through

AC

CB1 receptors in mast cells [129], increasing B cell proliferation [95].


The key to understand the role of cannabinoids in immunomodulation is to stress that their effects depend on the type of cannabinoid, on the type of cells that they are acting on and on the
doses administrated. In optimal concentrations, cannabinoids can induce apoptosis in immune
cells, alleviating inflammatory responses and protecting the host from acute and chronic inflammation. Thus, they can be beneficial when an immune suppression is necessary. Additional research is still needed to validate these studies in humans through clinical trials.
The effects of the three main cannabinoids that could be beneficial to autoimmune diseases are
summarized in Fig 4.

ACCEPTED MANUSCRIPT
6. Effects of cannabinoids in autoimmune disorders
6.1 Cannabinoids in Multiple Sclerosis (MS)
MS is a chronic neuroinflammatory autoimmune demyelinating disease of the human central

IP

nervous system, mediated by T lymphocytes. The inflammatory process leads to BBB disruption, causing swelling, activation of macrophages, and further production of cytokines and cyto-

CR

toxic proteins such as metalloproteinases.

NU
S

In mice model of MS (Experimental Autoimmune Encephalomyelitis (EAE)) it has been shown


that CB1 receptors play an important role in the control of neuroinflammation. Mice deficient in
CB1 tolerate inflammatory and exitotoxic insults poorly and develop substantial neurodegenera-

MA

tion following immune attack [131].

THC has been reported to inhibit neurodegeneration in the EAE model by reducing inflammato-

ED

ry cytokines and to reduce the associated induced elevated level of glutamate in cerebrospinal

PT

fluid [132]. Glutamate, the major excitatory neurotransmitter in the cerebral cortex, has been implicated in neurodegenerative disease when present at high levels.

CE

Studies in the CB1 knockout mice showed greater susceptibility to neurofilament damage, caspase 3 activation as well as greater dephosphorylation of a neurofilament H epitope, considered

AC

as a marker of axonal damage during chronic relapsing EAE. [133] Collectively, the data suggested that signaling through CB1 conferred neuroprotection during EAE.
Studies have also shown that CB1 receptor activation exhibited down-regulatory responses in
the motor regions of CNS system where they were expressed and could explain the improvement in motor symptoms such as spasticity, tremor, and ataxia of mice and human models
treated with CB1 agonists.[134] Rolipram, an inhibitor of type IV phosphodiesterase that suppresses EAE in different species, has been found to attenuate clinical decline, reduce motor inhibition, and normalize CB1 gene expression in the basal ganglia of EAE rats [135]. EAE rats also
have been reported to exhibit changes in endocannabinoid levels presenting decreased levels of

ACCEPTED MANUSCRIPT
anandamide and 2-AG in motor related regions such as the striatum and midbrain as well as in
other brain regions. [135].
CB2-selective agonists such as RWJ400065 have not shown to inhibit spasticity.

IP

However , CB2-deficient T lymphocytes in the CNS during EAE exhibit reduced levels of apoptosis, a higher rate of proliferation, and increased production of inflammatory cytokines resulting

CR

in severe clinical disease. In addition, selective glial expression of both, CB1, CB2, and FAAH

NU
S

has been reported as associated with MS, supporting a role for the endocannabinoid system in
the pathogenesis and/ or evolution of this disease [136]

Endogenous cannabinoid, anandamide plays an important role in controlling the inflammatory

MA

process during the course of MS progression. It was demonstrated that treatment with the selective anandamide uptake inhibitor UCM707 during established disease resulted in a significant

ED

improvement in motor function. UCM707 was able to reduce microglial activation, diminish

PT

MHC class II antigen expression, decrease cellular infiltrates in the spinal cord, and decrease the
production of the pro-inflammatory cytokines TNF-, IL-1, and IL-6. The endocannabinoid

CE

system is highly activated during CNS inflammation and anandamide protects neurons from inflammatory damage through a CB1/CB2-mediated rapid induction of microglial mitogen-

AC

activated protein kinase phosphatase-1 (mkp-1) [137].


Studies in humans are scarce. Recently the results of a clinical trial have been published where
the effect of (9)-tetrahydrocannabinol was tried in 498 patients with primary or secondary progressive MS, 1-year evidence of disease progression and baseline EDSS 4.06.5. No differences
were found in the rate of progression of MS in both groups although lower than expected progression rates might have affected the ability to detect clinical change. [138]
Nevertheless, there is sufficient data to suggest symptomatic improvement when cannabinoid
treatment is used in MS patients. Studies done in patients with MS with the aim to see the effect
of THC on spasticity and pain proved long-term symptomatic improvement of spasticity and relief of pain. [139-141]

ACCEPTED MANUSCRIPT

6.2 Cannabinoids in Rheumatoid Arthritis.


Joint diseases, inflammatory and degenerative, all share a pathological feature which is the loss

IP

of articular cartilage. In osteoarthritis and rheumatoid arthritis (RA), there is an increased cartilage breakdown. It is induced by an increased production of inflammatory cytokines, particularly

CR

IL-1 and tumor necrosis factor (TNF) produced by the articular chondrocytes or cells of the syn-

NU
S

ovium [142]. The overall result is an increase in metalloproteinases (MMPs) particularly MMP3 and MMP-13, which are responsible for cartilage destruction.
Cannabinoids were shown to have anti-inflammatory effects and reduce to joint damage in ani-

MA

mal models of arthritis [143-145] According to in vitro studies, cannabinoids reduce cytokine
production by RA fibroblasts as well as the release of matrix metalloproteinases (MMPs) from

ED

fibroblast-like synovial cells [146-148]. Cannabinoids have also shown to reduce interleukin 1

PT

(IL-1) induced proteoglycan and collagen degradation in bovine cartilage, thus reducing cartilage
extracellular matrix (ECM) breakdown [149].

CE

Studies have been performed with synthetic cannabinoid WIN-55,212-2 mesylate (WIN-55). It is
an agonist of CB1, CB2 receptors and also has an ability to activate other receptors including

AC

peroxisome proliferator activated receptors alpha and gamma (PPARa and g) [150-152]. It was
proved that WIN-55 can reduce the gene and protein expression of MMP-3 and MMP-13 in the
presence of IL-1b, suggesting that cannabinoids may have potential in terms of arthritis therapy.
It also significantly reduces the gene expression of TIMP-1 and TIMP-2 tissue inhibitors of matrix metalloproteinases to below basal levels. [153]
Similar results were obtained with a selective CB2R agonist (HU-308) in vitro. Pre-treatment
with HU-308 inhibited IL-1-induced proliferation of cultured RA fibroblast-like synoviocytes.
The agonist also decreased the production of proinflammatory cytokines (IL-6 specifically) and
of matrix metalloproteinases that are thought to be involved in cartilage erosion (MMP3 and
MMP13.) In another study non-psychoactive cannabinoid ajulemic acid (AJA) reduced MMP-1,

ACCEPTED MANUSCRIPT
MMP-3 and MMP-9 release from fibroblast like synovial cells stimulated with IL-1a and TNFa
[148]. In vivo, AJA has also been shown to reduce the severity of adjuvant-induced arthritis
[144]. When a novel synthetic cannabinoid acid, Hebrew University 320 (HU-320) was admi-

nistered to a mouse model of arthritis, inhibition of mouse macrophages TNF production was

IP

observed. The authors also noticed reduction of reactive oxygen intermediates and suppression

CR

of the rise in serum TNF level following endotoxin challenge. [153]

NU
S

All these data suggests that cannabinoids have anti-inflammatory properties and should be considered as a treatment for inflammatory arthritis. There are no studies in humans up to date on
the use of cannabinoids reducing inflammation in RA and further studies are needed to prove

MA

that.

There are however, studies which have shown that cannabinoids are efficient in the treatment of

ED

pain associated with RA. There is evidence that endocannabinoid system plays an important role
in the peripheral regulation of nociception [154,155]. CB1R present on nociceptor terminals may

PT

mediate the anti-nociceptive and anti-inflammatory actions of locally produced N-arachidonoyl

CE

ethanolamine through its inhibitory influence on the release of excitatory neuropeptides [155]
CB1R and CB2R are also expressed in the dorsal root ganglia, and their stimulation at this level

AC

also decreases nociceptive transmission [156,157]. At the central level, the endocannabinoid
system controls nociception through CB1R located at spinal and supraspinal levels [158] Pharmacological studies have suggested a peripheral endocannabinoid basal activity that modulates
nociceptor activity in osteoarthritic joints [159]. CB1R and CB2R mRNA and protein, were
found in synovial biopsies deriving from total knee arthroplasty of advanced osteoarthritis and
rheumatoid arthritis patients [160] Both, anandamide (AEA) and 2-arachidonoylglycerol (2-AG)
were detected in the synovial fluid of these patients, but not in healthy volunteers, providing further evidence for a functional endocannabinoid system in osteoarthritic joints [160] Spinal cord
levels of AEA, 2-AG and their synthesizing enzymes were also increased in the rat model of os-

ACCEPTED MANUSCRIPT
teoarthritis [161] All these findings provide evidence for both peripheral and central adaptive
changes involving the endocannabinoid system during osteoarthritis.
Studies on human models are scarce and contradictory. A recent review article on the effect of

cannabinoid nabiximol on chronic pain in rheumatic diseases shows that there is a low-quality

IP

evidence suggesting that cannabinoids may be associated with improvements in pain and sleep

CR

quality in RA. Measures of morning pain at rest, sleep quality and a global disease activity score

NU
S

showed improvement, but measures of pain intensity were unchanged [162]. However, Savitex (
dronabiol) is sometimes used to reduce pain in inflammatory joint disease- [163]
6.3 Cannabinoids in Scleroderma:

MA

Cannabinoid receptors CB1 and CB2 were found to be over-expressed in scleroderma fibroblasts
compared with healthy fibroblasts. A study was carried out to see the effect of WIN55,212-2,

ED

CB1 CB2 agonist on the production of collagen. It was demonstrated that WIN55,212-2 induced

PT

inhibition of type I collagen as well as procollagen type I PIP supernatant levels, which was also
associated with changes in skin fibrosis [164]. The reduction in type 1 collagen production was

CE

accompanied by a decrease in levels of growth factors involved in the fibrotic process, TGF beta and its downstream mediator CTGF. There was also an inhibition of fibroblast trans diffe-

AC

rentiation and inhibition of the over-expressed IL-6 in scleroderma cultures, known to be involved in scleroderma pathogenesis [165].
The agonist WIN55,212-2 also significantly increased the number of scleroderma fibroblasts
undergoing apoptosis. These observations are consistent with the anti-proliferative effect of cannabinoids. [166]. No toxicity of WIN55,212-2 on cell cultures was found. However, the antifibrogenic effect of WIN55,212-2 did not seem to be mediated by the classic cannabinoid receptors CB1 and CB2 and it was proposed that ERK signaling pathways may be involved. [167]
Further studies have shown that additional receptor pathways, including the transient receptor
potential vannilloid type-1 (TRPV1), the peroxisome proliferator-activated receptors (PPARs),
G-protein receptor 55 (GPR55) as well as nicotine, 5-HT3 and adenosine A2A receptors are in-

ACCEPTED MANUSCRIPT
volved in cannabinoid signal transduction.[168,169] Oral administration of Ajulemic Acid
(AjA), a non-psychoactive synthetic analogue of tetrahydrocannabinol able to bind the peroxisome proliferator-activated receptor- (PPAR-) prevented the development of skin fibrosis, and

reduced skin thickness nearly to control levels. At the biological level it produced dose depen-

IP

dent reduction of procollagen and 5 TGF- [170]. According to these observations, the stimula-

CR

tion of peroxisome proliferator-activated receptor- (PPAR-) could have an anti-fibrotic effect

NU
S

and cannabinoids should be studied as a potential anti fibrotic treatment.


6.4 Cannabis in Type 1 Diabetes

MA

In type 1 diabetes mellitus, the insulin-pancreatic b-cells are destroyed, most probably, mediated
by CD4 Th1 and CD8 T lymphocytes [171,172]. During insulitis, the initial lesion of this dis-

ED

ease, leukocytes, especially lymphocytes, surround and infiltrate the islets [173].
Cannabidiol treatment of 6 to 12 week-old Non-Obese-Diabetes (NOD) mice was shown to sig-

PT

nificantly reduce the incidence of diabetes (from 86% in non-treated control mice to 30% in

CE

mice treated with cannabidiol). The plasma levels of INF-gama and TNF-alfa were reduced too,
as well as other TH1 cytokines. The production of IL4 and IL10 (TH2 cytokines), on the other

AC

hand, was increased. When histological examination of the pancreatic islets was done, cannabidiol treated mice revealed significantly reduced insulitis. Therefore, cannabinoids can inhibit or
at least delay destructive insulitis and TH1 associated cytokine in NOD mice. [99]
Despite the fact that cannabinoid could prevent diabetes type 1 in NOD young mice, it was not
demonstrated to ameliorate diabetes after onset or around onset time. Thus, the same authors repeated the experiment with cannabidiol treatment, this time with 11 to 14 week-old NOD mice.
They observed amelioration of diabetes manifestations: while the untreated and the emulsifiertreated group had 86% and 100%, respectively, of diabetes incidence at the end of the treatment;
the group treated with cannabidiol had only 32%. Histological examination of the pancreas of
cannabidiol treated mice revealed more intact islets than in the controls [174].

ACCEPTED MANUSCRIPT
It was suggested that cannabidiol could have a role in prevention of human type 1 diabetes.
However, it depends on the mechanism of the anti-autoimmune properties of this molecule. The
main issue is to determine if it is a non-specific immunosuppressive agent or if it deviates Th1 to

the response. If it is a non-specific immunosuppression mechanism, it requires lifelong treat-

IP

ment. Log-term therapy causes substantial potential risks, including possible enhanced suscepti-

CR

bility to opportunistic infection and increased risk of malignancy. Diabetes also has longstanding

NU
S

consequences, but they can be reduced with intensive insulin therapy aimed at normalizing glucose levels [175], what makes cannabinoid therapy need to be carefully evaluated before using in
humans. On the other hand, if cannabidiol is causing the deviation from Th1 to Th2, it could be

MA

used to prevent diabetes in early on-set patients, before complete beta-cells destruction, and
maybe in high risk individuals. Once the autoimmune response is deviated from a destructive

ED

Th1 response to a protective Th2 response, cannabidiol therapy is no longer required, eliminat-

PT

ing the concern regarding long-term safety issues. [99]


Cytokines released by macrophages have a critical immunomodulatory role in skewing the im-

CE

mune response to either Th1 or Th2 [176]. Administration of IL-12 to macrophage-depleted

AC

NOD mice is known to initiate the disease [177]. Cannabidiol was shown to significantly reduce
macrophage IL-12 production. [174] By reducing IL12, it induces TH2 differentiation and also
reduces INF-g production. INF-g, that potentiates IL12 effects, activates cytotoxic T cells and
stimulates macrophages to produce TNF-alfa, IL1 and IL6, besides nitrogen free radicals. Those
molecules are associated with beta cells destruction [178-180]. Therefore, cannabidiol suppression of macrophage IL-12 production could inhibit Th1-mediated autoimmunity [99].
At onset of diabetes most patients have approximately 10% functional beta cells, evidenced by
residual c-peptide secretion. Approximately 50% of patients diagnosed with type 1 diabetes will
enter a honeymoon remission phase of diabetes and remain insulin independent within the first
year of diagnosis [99]. These patients have sufficient residual beta cells to maintain normoglycemia and insulin independence, so they could be candidates for immunomodulation therapy.

ACCEPTED MANUSCRIPT
Studies are still needed to prove if a Th1/Th2 shift actually happens in humans, and if it occurs,
it really provides long-term protection from disease without other interventions.

6.5. Cannabis and Inflammatory Bowel Diseases

IP

Cannabis has been widely used to cure disturbances and inflammation of the bowel. The me-

CR

chanisms involved are still unclear, but probably include peripheral actions on CB1 and CB2 and

NU
S

may also include central actions [181].

Many animal experiments studies of cannabinoids possible therapy in colitis have been done in
the last decade. Most of them showed reduced inflammation with cannabinoid agonists, while

MA

cannabinoid antagonists or cannabinoid receptor knockout increased inflammation [182-189].

ED

Recently, studies with human colitis patients are underway.


Cannabinoids receptors are differentially expressed in human IBD, indicating a cannabinoid reg-

PT

ulatory role in the disease progress [104, 190, 192].

CE

Anandamide and its synthesizing enzyme are decreased in Ulcerative Colitis (UC), and the expression of CB2 receptors and of enzymes responsible for synthesis and degradation of 2-

AC

Arachidonoylglycerol are increased [193]. It suggests that cannabinoids can ameliorate the IBD.
There are CB1 and CB2 receptors located at the colonic epithelium, so cannabinoids might enhance epithelial wound closure in the colon [194]. The activation of CB2 also leads to apoptosis
and decreased proliferation of T cells in colitis, and diminishes the recruitment of neutrophils, T
cells and macrophages to the inflamed colon [195].
The endocannabinoid system can also control gut motility and secretion, by CB receptors located
in the enteric nervous system [192, 196]. These CB1 receptors can protect the bowels from the
hyper stimulation that happen during IBD. By this mechanism, THC may improve IBD symptoms, mainly diarrhea, besides controlling the inflammation [196-198].

ACCEPTED MANUSCRIPT
The possible central effect would be the reduction in pain sensation and relief of nausea, induced
by stimulation on CB1 present in the central nervous system. This theory is supported by the fact
that one study with peripherally restricted CB1 and CB2 agonist was too weak to improve colitis

IP

[199].

CR

In a prospective placebo-controlled study with 21 Crohn Disease patients [200], cannabis induced clinical remission in 50% of patients. 80% of the participants had nonresponse or intoler-

NU
S

ance to antiTNF-alfa. Although, the improvement was only symptomatic, with no induction of
remission comparing to placebo. When patients discontinued the cannabis therapy, relapses were
noticed in 2 weeks. Other cohort study of patients with IBD using cannabis [201] also reported

MA

that the patients perceive that it improved abdominal pain. Patients with UC, in particular, re-

ED

ported cannabis to improve diarrhea.

In another observational study [202], this time with in 30 patients with Crohns disease, medical

PT

cannabis was associated with improvement in disease activity and reduction in the use of other

CE

medications was reported.

Usually, 25% to 38% of operated Crohns disease patients require a second operation within 5

AC

years [203], but in a retrospective cohort study [204] only 2 of 15 patients (13%) who had surgery before cannabis consumption required surgery while consuming cannabis. Studies are still
needed, but this one suggested that the use of cannabis reduces the need for surgery.
Some patients with UC and CD also use cannabis to enhance appetite [205]. In a study of 13
patients using cannabis for 3 months, a statistically significant increase in the subjects weight
was observed [206]. They also presented improvement in the disease activity index, perception
of general health status, and ability to perform daily activities.

ACCEPTED MANUSCRIPT
There is evidence that cannabinoids treatment could have beneficial effects on IBD. However,
further research is required before it can be established which cannabinoid to use, in what dose

and in which administration mode [202].

IP

6.6. Cannabis in neuropathic pain

CR

Neuropathic pain is a disease of peripheral or central nervous system. It happens when peripheral
nerves, spinal cord, or brain are injured or the sensory system functions in the wrong way. There

NU
S

are many causes such as underlying pathological process (e.g., neuropathy) and catastrophic injury (e.g., stroke or spinal cord injury) [207]. The treatment for neuropathic pain is a difficult

MA

field, because in randomized clinical trials, less than half of patients report clinically meaningful
pain relief from pharmacotherapy, most of them, just partial relief [208]. Because of that, and

ED

due to the studied anti-inflammatory potential of cannabis, a lot of studies about cannabinoid

PT

therapy to neuropathic pain have been performed [209].


Chronic pain is associated with the worst quality of life as compared with other chronic diseases

CE

such as chronic heart, lung or kidney disease, and what's more, is associated with a higher risk of
suicide [210, 211]. A systematic review about cannabis in chronic non-cancer pain, especially in

AC

neuropathic pain, [212] selected 18 randomized control trials demonstrating a modest analgesic
effect of cannabinoids in chronic pain.
A recent qualitative systematic review [213] stated that new safe and effective agonists at the
cannabinoid receptors may dissociate therapeutic effects from psychotropic effects. For example,
1,1Dimethylheptyl-8-tetrahydrocannabinol-11-oic acid (CT-3) is a synthetic analog of THC11-oic acid, one of the endogenous transformation products of THC. It was shown to be a potent
anti-inflammatory, analgesic and antiallodynic agent with no psychoactive properties in preclinical studies [214].

ACCEPTED MANUSCRIPT
The exact neurobiological mechanism of action of cannabinoids is still unclear. Some studies
claim that they work by binding to CB1 and in the intracellular peroxisome proliferator
activated receptor (PPAR), which is directly associated to anti-inflammatory and antitumor

effects [213-215]. Other studies suggest that there is an inhibition of eicosanoid synthesis and

CR

IP

down-regulation of COX2 [216].

In order to examine the analgesic efficacy and safety of CT-3 in chronic neuropathic pain in

NU
S

humans, a randomized placebo-controlled double-blind crossover trial was preformed [217].


There were 28 patients with examination consistent with chronic neuropathic pain for at least 6
months with hyperalgesia and allodynia. They analyzed visual analog scale (VAS) and verbal

MA

rating scale scores for pain. It was observed that the VAS values in the CT-3 sequence, measured
3 hours after intake of study drug, differed significantly from those in the placeboCT-3 se-

ED

quence (11.54 [14.16] vs 9.86 [21.43]; P = .02). Although 8 hours after intake of the drug, the

PT

pain scale differences between groups were less marked.

CE

In two different trials, patients with HIV peripheral neuropathy in cannabinoid therapy showed
reduction in the pain (46-52%) compared to 18-24% in the placebo groups [218, 219]. This type

AC

of pain is notoriously resistant to other treatments normally used for neuropathic pain, which
emphasize the importance of these study. [220]
Another group [221] made a human experimental model of neuropathic pain using intradermal
injection of capsaicin in healthy volunteers and tested the efficacy of different doses of THC as a
therapy. Low dose cigarettes (2% THC) had no analgesic value, while high dose (8% THC) cigarettes were associated with reports of an increase in pain. On the other hand, the medium dose of
cannabis cigarettes (4% THC) produced significant analgesia.
A randomized double-blind placebo-controlled trial published in 2014, [207] compared medium
dose (3.53% THC) to low dose (1.29% THC) cannabis, to see if the analgesia was sufficient in
the low doses. The purpose was that if it as effective as the medium dose, it should be used pre-

ACCEPTED MANUSCRIPT
ferentially, to avoid side effects. Both provided statistically significant 30% reductions in pain
intensity when compared to placebo.

In a crossover trial with 23 patients with post-traumatic or post-surgical neuropathic pain [222],

IP

it was given 4 different doses of THC to the patients (0%, 2.5%, 6% and 9.4%) for four 14-days

CR

periods. It was inhale through a pipe three times a day for the first 5 days, followed by 9 days
washout period, in each cycle. The pain intensity was measured daily by a numeric rating scale.

NU
S

They also analyzed the effects on mood, sleep and quality of life. The average daily pain measured was lower on the pre-specified primary contrast of 9.4% to 0% (5.4 to 6.1 respectively). Individuals who received 9.4% THC demonstrated improved ability to fall asleep and quality of

MA

sleep, when compared to the 0% THC group. There was no differences in mood or in quality of

ED

life.

All those studies showed that modulating the cannabinoid system can help patients with neuro-

PT

pathic pain, not only by relieving their pain, but also by improving their sleeping skills. Further

CE

long-term safety and efficacy are still needed.

AC

6.7 Fibromyalgia:

FM is characterized by chronic widespread pain and elevated response to pressure which is perceived as pain. Other symptoms include tiredness, morning stiffness, sleep and emotional disturbances as well as cognitive dysfunction [223]. The pathophysiology of the disorder is not
known. Several mechanisms have been proposed including central sensitization, suppression of
descending inhibitory pathways, excessive activity of glial cells, abnormalities of neurotransmitter release as well as abnormal response to stress. [224]. Currently, the treatment is based on the
relief of symptoms but poor results are achieved. The participation of the endocannabinoid system in multiple physiological functions such as pain modulation, stress response system, neuroendocrine regulation and cognitive functions amongst others, is well known [225]... It was
suggested that clinical endocannabinoid deficiency may underlie the hyperalgesic tender muscle

ACCEPTED MANUSCRIPT
points of this condition and play an important part in etiopathology of myofascial pain syndrome
fibromyalgia [226,227].
Some studies provide data that cannabinoids can prove to be an effective treatment of fibromyal-

gia symptoms. One retrospective study of fibromyalgia affected patients who were chronic users

IP

of cannabis, analyzed the effect of smoked, oral and combined cannabis on symptoms and the

CR

quality of life. A significant reduction of pain and stiffness, enhancement of relaxation, and an

NU
S

increase in somnolence and feeling of wellbeing were observed in cannabis users. Furthermore,
mental health component summary score was significantly higher (p=0.05) in cannabis users

MA

than in non-users [228].

Some data suggests the beneficial effect on sleep in fibromyalgia patients. Nabilone had shown
to be effective in improving sleep in patients with FM and was well tolerated. Low-dose nabi-

ED

lone given once daily at bedtime may be considered as an alternative to amitriptyline treatment.

PT

[229]

CE

7. Adverse Effects of Cannabis

AC

Due to the extend distribution of its receptors, cannabis are known to have many side effects.
The most important ones are listed in Table 5.
In a systematic study [230], 8371 adverse events related to medical cannabinoid use were reported, 4779 of which were reported in 23 randomized controlled trials and 3592 in 8 observational studies.
Patients in cannabinoid therapy presented twice the incidence of severe adverse events than control groups. Respiratory (16.5%), gastrointestinal (16.5%) and nervous system disorders (15.2%)
were the most frequently reported categories of serious adverse events among those assigned to
cannabinoids, whereas nervous system disorders (30%) were the most frequently reported among

ACCEPTED MANUSCRIPT
controls. The events considered severe side effect were: dyspnea, pneumonia, pleural effusion,
lower respiratory tract infection and pulmonary embolism; vomiting, diarrhea, gastroenteritis,

abdominal pain, constipation, duodenal ulcer; relapse of multiple sclerosis, convulsions.

IP

15 deaths were also reported among cannabinoid users against 3 deaths in the control group, al-

CR

though these results did not prove to be statistically significant. [230]


The incidence rate of non-serious adverse events was significantly higher among subjects as-

NU
S

signed to cannabinoid therapy than among controls, dizziness being the most frequent nonserious adverse event among cannabinoid-exposed participants.

MA

8. Conclusion

ED

Cannabinoids are potent inflammatory modulators and in several animal studies, in vivo and in
vitro, they have shown to have an immunosuppressive effect.

PT

However, human studies are still few.

CE

Models of multiple sclerosis, rheumatoid arthritis, scleroderma and type 1 diabetes evidenced

AC

clinical improvement as well as biochemical and/or histological anti-inflammatory changes. Inflammatory bowel disease, neuropathic pain and fibromyalgia were studied in human subjects
and improvement of pain, positive effect on sleep and better quality of life were noted. In ulcerative colitis there were less diarrhea symptoms after cannabinoid administration.
Certain types of cannabinoids, like cannabidiol, have low affinity to CB1 and CB2. Others, such
as certain synthetic cannabinoids, have higher affinity for CB2 receptor. However, they do produce immunomodulatory effect without being psychoactive. Therefore, they have a potential in
research, as a possible therapy for autoimmune diseases
Nevertheless, cannabis are known to have many adverse effects, including memory impairment.
THC is related to a decrease in gray matter volume and lower IQ levels. It can also trigger some

ACCEPTED MANUSCRIPT
psychiatric disorders. Because of the THC stimulation of dopaminergic pathways involved in
drug addiction, it can lead to dependence and drug abuse.

Further studies are required. How cannabinoids should be consumed, their optimal doses and

IP

which type of cannabinoids would be more effective, with fewer side effects still remain as ques-

AC

CE

PT

ED

MA

NU
S

CR

tions to be answered.

ACCEPTED MANUSCRIPT
Table 1. Cannabinoids receptor agonists groups and its components. [1,2]
Classical

Non-Classical

Aminoalkylin-

Eicosanoid

noids:

cyclic analogues

512

9-THC that

THC

lack a pyran ring:

THC

Canna

binol

AC

PT

CE

noids:

HU210

Virodhamine

2-arachidonoyl glycerol( 2-AG)

Synthetic analogues of anandamide:

R-(+)methanandamide

CP55
244

Anandamide

Arachidonyl-2-

HU-

chloroethylamide

308

(ACEA)

Arachidonylcyclo-

JWH-

propylamide (AC-

133

PA)

L-

O-689

759633

O-1812

L759656

CP47
497

Synthetic cannabi-

Endocannabinoids:

P5594

ED

MA

9-

NU
S

of

WIN5

IP

Bicyclic and tri-

CR

Phytocannabi-

dole

L-

ACCEPTED MANUSCRIPT
nantra-

dol

Desace-

IP

tyl-l-

CR

nantra-

AC

CE

PT

ED

MA

NU
S

dol.

ACCEPTED MANUSCRIPT
Table 2: Effects of Cannabinoids in the Human Body

Effect

Reference

Body System

Anti-emetic; neuroprotective, nociception. Se-

CR

dating. Simpaticomimethic

IP

CNS

Tachycardia, increased cardiac output

NU
S

Circulatory

[40]

[49,50,51,53,54]

Vasodilation and orthostatic hypotension


Stimulation by leptin inhibition

Osteoarticular

Bone formation

Gastrointestinal

Inhibition of motility and gastric secretion

PT

ED

MA

Appetite and eating

AC

CE

Reproductive system

Minor changes in hormone levels, no effect when

[55]

[56]

[57,58]
[51,59]

tolerance is reached.
Decrease in sperm count without functional impairment

Ocular

Vasodilation. Descrease intraocular pressure

[60,61]

Immune system

Inmunosupression, immunomodulation

[69-73]

ACCEPTED MANUSCRIPT
Table 3: Medical conditions in which cannabinoids were shown to be effective.

Improve pain

Post-operative and trauma

Improve pain

Chronic pain in cancer

231, 232

NU
S

pain

References

IP

Effect

CR

Medical condition

Chemotherapy induced nausea Improve symptoms of nausea


and vomiting

Cancer cachexia and HIV

Improve appetite and

234-239

240-243

ED

MA

and vomiting

233

increasing weight in

PT

disease

Spasticity, movement

Improve symptoms

244-246

Improve symptoms, pain and

191, 247

AC

CE

disorders and dystonia

Ggastro-intestinal conditions

(gastric ulcers, irritable bowel increase life quality


syndrome, ulcerative colitis,
Crohns disease, secretory
diarrhea, paralytic ileus and
gastroesophageal reflux
disease)

ACCEPTED MANUSCRIPT
Table 4: How cannabinoids can be consumed: advantages and limitations.

Limitations

References

Cutaneous absorption

No need to eat or

Takes too long to act.

by adhesives

inhale

CR

NU
S

applications.

No need to inhale.

MA

Oral absorption

Onset of action after

ED

30-60 minutes.

Peak of action 2-3

PT

hours after ingestions

CE

248

No clinical

impregnated by herbs

AC

IP

Administration form Advantadges

Increased
bioavailability by
presence of food in
the stomach and
gastric acid partial
destruction.
Psychoactive effects
due to conversion of
9-THC to 11hydroxy-THC in the
liver (9-10 times more
psychoactive than 9THC)

249, 250

ACCEPTED MANUSCRIPT
Limitations

References

Intravenous

No need to eat or

Needs a vehicle to

248, 251

administration (bolus

inhale.

permits aqueous

IP

Fast absorption (goes

administration due to
cannabis low water

directly to the

solubility.

NU
S

systemic circulation)

CR

or infusion)

Administration form Advantadges

Rectal administration

No need to eat or

(suppositories)

inhale.

251

Peak of action in 20-

Needs combustion

252

administration -

30 minutes after

(can lead to lung

smoking (cigarettes,

smoking

lesions).

High intensity and

Plasma levels

short duration of

depending on volume

action (2-3 hours)

and respiratory rate.

MA

Irregular absorption

ED

Fast absorption (goes


directly to the

PT

systemic circulation)

AC

CE

Inhaling

pipes)

ACCEPTED MANUSCRIPT
Limitations

References

Inhaling

No need for

Plasma levels

248, 253

administration -

combustion

depending on volume

dispositive that boils


the cannabis herbs,

as intravenous

creating a vapor that


can be inhale without

high lipossolubility, it
crosses rapidly the

MA

combustion)

alveolar membrane
reaching the blood

ED

through pulmonary

CE

PT

capillaries)

AC

and respiratory rate.

NU
S

injection (due to its

IP

Peak of action as fast

CR

vaporizer (portable

Administration form Advantadges

ACCEPTED MANUSCRIPT
Table 5: Side effects of cannabinoids in each system.
Examples:

References

AC

CE

PT

ED

MA

NU
S

CR

IP

Effects

ACCEPTED MANUSCRIPT
Effects

Examples:

References

Neuropsychiatric effects

Dysphoric mood alterations

230, 254-262

IP

like anxiety, panic.

CR

Psychosis

NU
S

Sedation

Impaired global and

prospective memory, verbal

MA

immediate and delayed recall


and visual recognition.

ED

Convulsion

AC

CE

PT

Lower IQ (depending on age at


onset)
Decrease in gray matter
volume in the medial temporal
cortex, temporal pole,
parahippocampal gyrus, left
insula, and orbitofrontal cortex
Increased appetite.
Drug Addiction

ACCEPTED MANUSCRIPT
Effects

Examples:

References

Immune system effects

Increase incidences of

230, 259, 260, 263-270

IP

common infectious diseases

CR

Increase susceptibility to viral


infections such as herpes

NU
S

simplex virus, Listeria


monocytogenes,

Staphylococcus albus,

MA

Treponema pallidum and

ED

Legionella pneumophila
Allergies to cannabis (rare):

CE

PT

contact urticaria and rhino

AC

Vascular system

conjunctivitis

Syncope
Transient ischemic attack
Stroke

259,260

ACCEPTED MANUSCRIPT
Effects

Examples:

References

Respiratory system

Chronic respiratory irritation

230, 267

IP

Inflammatory changes in

CR

respiratory tract

Suppression in responsiveness

Dyspnea

NU
S

to infectious disease

MA

Pneumonia

PT

ED

Pleura effusion

AC

CE

Gastrointestinal system

Vomiting
Nausea
Diarrhea
Gastroenteritis
Abdominal pain
constipation
Duodenal ulcer

230

CE

PT

ED

MA

NU
S

CR

IP

ACCEPTED MANUSCRIPT

AC

Figure 1: Cannabinoids agonists and antagonists

The table resumes the most important cannabinoid agonists and antagonists. AEA and 2-AG are
endogenous agonists and D9-tetrahydrocannabinol (D9-THC), the main psychoactive cannabis
component is an exogenous agonist. Together with synthetic derivatives (HU 210, HU 308,CP
55,940, etc.), they are the compounds most frequently used as pharmacologic tools.

CE

PT

ED

MA

NU
S

CR

IP

ACCEPTED MANUSCRIPT

AC

Fig. 2. CB1 CB2 signaling.


Both CB1 and CB2 cannabinoid receptors are associated with G proteins. Activation of these
leads to inhibition of adenylyl cyclase activity and activation of the different MAPK cascades. In
addition, the CB1 cannabinoid receptor negatively regulates voltage-gated Ca2+ channels and
positively regulates inwardly rectifying K+ channels. CB1 receptor can also induce elevation of
intracellular free Ca2+ through activation of phospholipase C (PLC). Cannabinoid-mediated inhibition of protein kinase A ( PKA) leads to inhibition of gene expression through a decrease in
cAMP . It also results in activation of MAPK cascades related to cell survival/apoptosis. These
signaling pathways are directly related to the variety of functions regulated by cannabinoid receptors.

CE

PT

ED

MA

NU
S

CR

IP

ACCEPTED MANUSCRIPT

AC

Fig 3. Action of cannabinoids at the presynaptic terminal.


Cannabinoid agonists such as THC, 2- Ag and AEA bind to CB1 receptors, causing change
in intracellular Ca2+ and K+ level. This in turn leads to neurotransmitter release inhibition at the presynaptic neuron. Cannabinoids are destroyed at the postsynaptic neuron by
FAAH enzyme and the metabolites are recycled.

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA

NU
S

CR

IP

Figure 4: Benefit effects of cannabinoids in autoimmune diseases.

ACCEPTED MANUSCRIPT
9. References:

1. Pertwee RG. Pharmacological Actions of Cannabinoids School of Medical Sciences, Institute

of Medical Sciences, University of Aberdeen, HEP . (2005) 168:151

IP

2. Howlett C, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, Felder CC, Herkenham

CR

M, Mackie K, Martin BR, Mecholaum R, Pertewee RG; International Union of Pharmacology.

NU
S

XXVII.Classification of Cannabinoid Receptors. Pharmacological Reviews Pharmacol Rev 2002


54:161202.

3. Pertwee RG. Cannabinoid pharmacology: the first 66 years. British Journal of Pharmacology

MA

2006 147 (S1): S163S171

4. Di Marzo V. CB1 receptor antagonism: biological basis for metabolic effects. Drug Discovery

ED

Today 2008: 116

5. Pagotto U, Marsicano G, Cota D, Lutz B, Pasquali R. The emerging role of the endocannabi-

PT

noid system in endocrine regulation and energy balance. Endocrine Reviews 27 (Feb 2006) (1):

CE

73100.

6. Howlett AC. Cannabinoid receptor signaling. Handb Exp Pharmacol2005; 5379.

AC

7. Di Marzo V. Endocannabinoids and other fatty acid derivatives with cannabimimetic properties: biochemistry and possible physiopathological relevance. Biochim Biophys Acta 1998;
1392:15375.

8. Giuffrida A, Beltramo M, Piomelli D. Mechanisms of endocannabinoid inactivation: biochemistry and pharmacology. J Pharmacol Exp Ther 2001; 298:714.
9. Dinh TP, Freund TF, Piomelli D. A role for monoglyceride lipase in 2-arachidonoylglycerol
inactivation. Chem Phys Lipids 2002; 121:14958.
10. Pertwee RG (1997) Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther
74:129180

ACCEPTED MANUSCRIPT
11. Pertwee RG, Gibson TM, Stevenson LA, Ross RA, Banner WK, Saha B, Razdan RK, Martin
BR. O-1057, a potentwater-soluble cannabinoid receptor agonist with antinociceptive properties.
Br J Pharmacol 2000 129:15771584

12. Iversen L. Cannabis and the brain. Brain 2003; 126:12521270

IP

13. Marsicano G, Moosmann B, Hermann H, Lutz B, Behl C. Neuroprotective properties of can-

CR

nabinoids against oxidative stress: role of the cannabinoid receptor CB1. J Neurochem 2002

NU
S

80:448456

14. Devane WA, Hanus L, Breuer A, Pertwee RG, Stevenson LA, Griffin G, Gibson D, Mandelbaum A, Etinger A, Mechoulam R. Isolation and structure of a brain constituent that binds to the

MA

cannabinoid receptor, Science 258 (1992) 19461949.

15. Di Marzo V, Petrosino S. Endocannabinoids and the regulation of their levels in health and

ED

disease. Curr. Opin. Lipidol. 18 (2007) 129140.


16. Hwang SW, Cho H, Kwak J, Lee SY, Kang CJ, Jung J, Cho S, Min KH, Suh YG, Kim D, Oh

PT

U. Direct activation of capsaicin receptors by products of lipoxygenases: endogenous capsaicin-

CE

like substances. Proc. Natl. Acad. Sci. U.S.A. 97 (2000) 61556160.


17. Ahluwalia J, Urban L, Bevan S, Nagy I. Anandamide regulates neuropeptide release from

AC

capsaicin-sensitive primary sensory neurons by activating both the cannabinoid 1 receptor and
the vanilloid receptor 1 in vitro. Eur. J. Neurosci. 17 (2003) 26112618.
18. Price TJ, Patwardhan A, Akopian AN, Hargreaves KM, Flores CM. Modulation of trigeminal sensory neuron activity by the dual cannabinoidvanilloid agonists anandamide, Narachidonoyl-dopamine and arachidonyl- 2-chloroethylamide. Br. J. Pharmacol. 141 (2004)
11181130.
19. Zygmunt PM, Petersson J, Andersson DA, Chuang H, Srgrd M, Di Marzo V, Julius D,
Hgesttt ED. Vanilloid receptors on sensory nerves mediate the vasodilator action of Anandamide. Nature 400 (1999) 452457.

ACCEPTED MANUSCRIPT
20. Tognetto M, Amadesi S, Harrison S, Creminon S, Trevisani M, Carreras M, Matera M, Geppetti P, Bianchi A. Anandamide excites central terminals of dorsal root ganglion neurons via vanilloid receptor-1 activation. J. Neurosci. 21 (2001) 11041109.

21. Shin J, Cho H, Hwang SW, Jung J, Shin CY, Lee SY, Kim SH, Lee MG, Choi YH, Kim J,

IP

Haber NA, Reichling DB, Khasar S, Levine JD, Oh U. Bradykinin- 12-lipoxygenase-VR1 sig-

CR

naling pathway for inflammatory hyperalgesia. Proc. Natl. Acad. Sci. U.S.A. 99 (2002) 10150

NU
S

10155.

22. OSullivan SE, Kendall DA, Randall MD. Characterisation of the vasorelaxant properties of
the novel endocannabinoid N-arachidonoyl-dopamine (NADA). Br. J. Pharmacol. 141 (2004)

MA

803812

23. Klein TW. Cannabinoid-based drugs as anti-inflammatory therapeutics. Nat Rev Immunol

ED

2005;5:40011

24. Lazzerini PE, Natale M, Gianchecchi E et al. Adenosine A2A receptor activation stimulates

PT

collagen production in sclerodermic dermal fibroblasts either directly and through a cross-talk

CE

with the cannabinoid system. J Mol Med (Berl). 2012;90:331-42.


25. Pertwee RG. Inverse agonism at cannabinoid receptors. In: IJzerman AP (ed) Inverse

AC

agonism. Elsevier 2003, Amsterdam, pp 7586


26. Pertwee RG. Novel pharmacological targets for cannabinoids. Curr Neuropharmacol 2004
2:929

27. Cheer JF, Cadogan A-K, Marsden CA, Fone KCF, Kendall DA . Modification of 5HT2 receptor mediated behaviour in the rat by oleamide and the role of cannabinoid
receptors. Neuropharmacology 1999 38:533541
28. Barann M, Molderings G, Brss M, Bnisch H, Urban BW, Gthert M. Direct inhibition
by cannabinoids of human 5-HT3A receptors: probable involvement of an allosteric
modulatory site. Br J Pharmacol 2002 137:589596
29. Fan P. Cannabinoid agonists inhibit the activation of 5-HT3 receptors in rat nodose

ACCEPTED MANUSCRIPT
ganglion neurons. J Neurophysiol 1995: 73:907910
30. Godlewski G, Gthert M, Malinowska B. Cannabinoid receptor-independent inhibition by
cannabinoid agonists of the peripheral 5-HT3 receptor-mediated von Bezold- Jarisch reflex. Br J

Pharmacol 2003: 138:767774

IP

31. Oz M, Zhang L, Morales M. Endogenous cannabinoid, anandamide, acts as a noncompetitive

CR

inhibitor on 5-HT3 receptor-mediated responses in Xenopus oocytes. Synapse 2002 46:150156

NU
S

32. Christopoulos A, Wilson K. Interaction of anandamide with the M1 and M4 muscarinic acetylcholine receptors. Brain Res 2001: 915:7078

33. Akinshola BE, Chakrabarti A, Onaivi ES.In-vitro and in-vivo action of cannabinoids. Neuro-

MA

chem Res 1999: 24:12331240

34. Akinshola BE, Taylor RE, Ogunseitan AB, Onaivi ES. Anandamide inhibition of recombi-

ED

nant AMPA receptor subunits in Xenopus oocytes is increased by forskolin and 8-bromo-cyclic
AMP. Naunyn Schmiedebergs Arch Pharmacol 1999: 360:242248

PT

35. De Petrocellis L, Di Marzo V. Review Role of endocannabinoids and endovanilloids in Ca2+

CE

signaling Cell Calcium 45 (2009) 611624


36. Grotenhermen F. Pharmacology of Cannabinoids. Neuroendocrinology Letters Nos.1/2, Feb-

AC

Apr Vol.25,(1/2):1423 2004


37. Dewey WL. Cannabinoid pharmacology. Pharmacol Rev 1986; 38:15178.
38. Pertwee R. In vivo interactions between psychotropic cannabinoids and other drugs involving central and peripheral neurochemical mediators. Marijuana/Cannabinoids: neurobiology and
neurophysiology. CRC Press; 1992. p. 165218
39. Baker D, Pryce G, Giovannoni G, Thompson AJ. The therapeutic potential of cannabis. Lancet Neurol 2003; 2:2918.
40. Domino EF. Cannabinoids and the cholinergic system. Marihuana and medicine. Humana
Press; 1999. p. 2236.

ACCEPTED MANUSCRIPT
41. Mattes RD, Shaw LM, Engelman K. Effects of cannabinoids (marijuana) on taste intensity
and hedonic ratings and salivary flow of adults. Chem Senses 1994; 19:12540.
42. Fan P. Cannabinoid agonists inhibit the activation of 5-HT3 receptors in rat nodose ganglion

neurons. J Neurophysiol 1995; 73:90710.

IP

43. Mller-Vahl KR, Kolbe H, Schneider U, Emrich HM. Movement Disorders. Cannabis and

CR

cannabinoids. Pharmacology, toxicology, and therapeutic potential. Haworth Press; 2002. p.

NU
S

205214.

44. Musty RE, Paul Consroe P. Spastic disorders. Cannabis and cannabinoids. Pharmacology,
toxicology, and therapeutic potential. Haworth Press; 2002. p. 195204.

Investig Drugs 2002; 11:136574.

MA

45. Grundy RI. The therapeutic potential of the cannabinoids in neuroprotection. Expert Opin

ED

46. Hampson A. Cannabinoids as neuroprotectants against ischemia. Cannabis and cannabinoids.


Pharmacology, toxicology, and therapeutic potential. Haworth Press; 2002. p. 10110.

PT

47. Mechoulam R, Shohami E. HU-211: Cannabinoid Neuroprotective Agent. In: Grotenhermen

CE

F, Russo E, editors. Cannabis and cannabinoids. Pharmacology, toxicology, and therapeutic potential. Haworth Press; 2002. p. 389400.

AC

48. Perez-Reyes M. The psychologic and physiologic effects of active cannabinoids. Marihuana
and medicine. Humana Press; 1999. p. 24552.
49. Tashkin DP, Levisman JA, Abbasi AS, Shapiro BJ, Ellis NM. Shortterm effects of smoked
marihuana on left ventricular function in man. Chest 1977; 72:206.
50. Benowitz NL, Jones RT. Cardiovascular effects of prolonged delta-9-tetrahydrocannabinol
ingestion. Clin Pharmacol Ther 1975; 18:28797.
51. Hollister LE. Health aspects of cannabis. Pharmacological Reviews 1986; 38:120.
52. Wagner JA, Varga K, Kunos G. Cardiovascular actions of cannabinoids and their generation
during shock. J Mol Med 1998; 76: 82436.

ACCEPTED MANUSCRIPT
53. Wagner JA, Jarai Z, Batkai S, Kunos G. Hemodynamic effects of cannabinoids: coronary
and cerebral vasodilation mediated by cannabinoid CB1 receptors. Eur J Pharmacol 2001;
423:20310.

54. Lake KD, Compton DR, Varga K, Martin BR, Kunos G. Cannabinoid induced hypotension

IP

and bradycardia in rats is mediated by CB1- like cannabinoid receptors. J Pharmacol Exp Ther

CR

1997; 281: 10307.

NU
S

55. Di Marzo V, Goparaju SK, Wang L, Liu J, Batkai S, Jarai Z, et al. Leptin-regulated endocannabinoids are involved in maintaining food intake. Nature 2001; 410:8225.
56. Mechoulam R, Shohami E, Fride E, Bab I. The ubiquitous role of endocannabinoids in phy-

MA

siological processes: examples in neuroprotection, feeding and bone formation. First European
Workshop on Cannabinoid Research. Madrid (Spain); 2003.

ED

57. Shook JE, Burks TF. Psychoactive cannabinoids reduce gastrointestinal propulsion and motility in rodents. J Pharmacol Exp Ther 1989; 249:4449.

PT

58. McCallum RW, Soykan I, Sridhar KR, Ricci DA, Lange RC, Plankey MW. Delta-9-

CE

tetrahydrocannabinol delays the gastric emptying of solid food in humans: a double-blind, randomized study. AlimentPharmacol Ther 1999; 13:7780.

AC

59. Murphy L. Hormonal system and reproduction. Cannabis and cannabinoids. Pharmacology,
toxicology, and therapeutic potential. Haworth Press; 2002. p. 28998,
60. Pate D. Glaucoma and cannabinoids. Cannabis and cannabinoids. Pharmacology, toxicology,
and therapeutic potential. Haworth Press; 2002. p. 21524. 101
61. Laine K, Jarvinen K, Jarvinen T. Topically administered CB(2)- receptor agonist, JWH-133,
does not decrease intraocular pressure (IOP) in normotensive rabbits. Life Sci 2003; 72:83742.
62. Horder J, Cowen PJ, Di Simplicio M, Browning M, Harmer CJ. Acute administration of the
cannabinoid CB1 antagonist rimonabant impairs positive affective memory in healthy volunteers
Psychopharmacology (Berl)2009 Jul;205(1):85-91.

ACCEPTED MANUSCRIPT
63. Costa B, Vailati S, Colleoni M. SR 141716A, a cannabinoid receptor antagonist, reverses the
behavioural effects of anandamide-treated rats. Behav Pharmacol. 1999;10:32731.
64. Arnone M, Maruani J, Chaperon F, Thiebot MH, Poncelet M, Soubrie P, Le Fur G. Selective

IP

(CB1) receptors. Psychopharmacol Berl. 1997;132:1046

inhibition of sucrose and ethanol intake by SR 141716, an antagonist of central cannabinoid

CR

65. Colombo G, Agabio R, Diaz G, Lobina C, Reali R, Gessa GL. Appetite suppression and

NU
S

weight loss after the cannabinoid antagonist SR 141716. Life Sci. 1998;63:1137.
66. Simiand J, Keane M, Keane P-E, Soubrie P. SR 141716, a CB1 cannabinoid receptor antagonist, selectively reduces sweet food intake in marmoset. Behav Pharmacol. 1998;9:17981.

MA

67. Berry EM, Mechoulam R. Tetrahydrocannabinol and endocannabinoids in feeding and appetite. Pharmacol Ther. 2002;95:18590.

ED

68. Bonf L, Vinagre RCO, de Figueiredo NV. Cannabinoids in chronic pain and palliative care.
Revista Brasileira de Anestesiologia, 2008 58(3), 267-279.

PT

69. Klein TW, Newton C, Larsen K, Lu L, Perkins I, Nong L, et al. The cannabinoid system and

CE

immune modulation. J Leukoc Biol. 2003;74:48696.


70. Berdyshev EV. Cannabinoid receptors and the regulation of immune response. Chem Phys

AC

Lipids. 2000;108:16990.

71. Cabral GA, Staab A. Effects on the immune system. Handb Exp Pharmacol. 2005:385423.
72. Pestonjamasp VK, Burstein SH. Anandamide synthesis is induced by arachidonate mobilizing agonists in cells of the immune system. Biochim Biophys Acta. 1998;1394:24960.
73. Bisogno T, Maurelli S, Melck D, De Petrocellis L, Di Marzo V. Biosynthesis, uptake, and
degradation of anandamide and palmitoylethanolamide in leukocytes. J Biol Chem.
1997;272:331523.
74. Lee SF, Newton C, Widen R, Friedman H, Klein TW. Differential expression of cannabinoid
cb(2) receptor mrna in mouse immune cell subpopulations and following b cell stimulation. Eur
J Pharmacol. 2001;423(23):235241.

ACCEPTED MANUSCRIPT
75. Massa F, Monory K. Endocannabinoids and the gastrointestinal tract. J Endocrinol Invest.
2006;29:4757.
76. Karsak M, Gaffal E, Date R, Wang-Eckhardt L, Rehnelt J, Petrosino S, et al. Attenuation of

allergic contact dermatitis through the endocannabinoid system. Science. 2007;316:14947.

IP

77. Massa F, Storr M, Lutz B. The endocannabinoid system in the physiology and pathophysiol-

CR

ogy of the gastrointestinal tract. J Mol Med. 2005;83:94454.

NU
S

78. Kumar RN, Chambers WA, Pertwee RG. Pharmacological actions and therapeutic uses of
cannabis and cannabinoids. Anaesthesia. 2001;56:105968.
79. Klein TW, Newton C, Larsen K, Lu L, Perkins I, Nong L, et al. The cannabinoid system and

MA

immune modulation. J Leukoc Biol. 2003;74:48696.

80. Berdyshev EV. Cannabinoid receptors and the regulation of immune response. Chem Phys

ED

Lipids. 2000;108:16990.

81. Newton CA, Klein TW, Friedman H. Secondary immunity to Legionella pneumophila and

PT

Th1 activity are suppressed by delta-9-tetrahydrocannabinol injection. Infect Immun 1994; 62:

CE

4015 4020.

82. Klein TW, Newton CA, Nakachi N, Friedman H. Delta 9- tetrahydrocannabinol treatment

AC

suppresses immunity and early IFN-g, IL-12, and IL-12 receptor beta 2 responses to Legionella
pneumophila infection. J Immunol 2000; 164: 6461 6466.
83. Zhu LX, Sharma S, Stolina M, et al. Delta-9-tetrahydrocan- nabinol inhibits antitumor immunity by a CB2 receptor- mediated, cytokine-dependent pathway. J Immunol 2000; 165:373
380.
84. Yuan M, Kiertscher SM, Cheng Q, Zoumalan R, Tashkin DP, Roth MD. Delta 9Tetrahydrocannabinol regulates Th1/Th2 cytokine balance in activated human T-cells. J Neuroimmunol 2002;133:124 131.
85. Munro S, Thomas KL, Abu-Shaar M. Molecular character- ization of a peripheral receptor
for cannabinoids. Nature 1993;365:61 65.

ACCEPTED MANUSCRIPT
86. Thomas BF, Gilliam AF, Burch DF, Roche MJ, Seltzman HH. Comparative receptor binding
analyses of cannabinoid agonists and antagonists. J Pharmacol Exp Ther 1998; 285:285 292.
87. Cunha JM, Carlini EA, Pereira AE, et al. Chronic administration of cannabidiol to healthy

volunteers and epileptic patients. Pharmacology 1980;21:175185.

IP

88. Consroe P, Laguna J, Allender J, et al. Controlled clinical trial of cannabidiol in Hunting-

CR

tons disease. Pharmacol Biochem Behav 1991;40:701708.

NU
S

89. Rieder SA, Chauhan A, Singh U, Nagarkatti M, Nagarkatti P. Cannabinoid-induced apoptosis in immune cells as a pathway to immunosuppression. Immunobiology. 2010 August ; 215(8):
598605.

MA

90. Schwarz H, Blanco FJ, Lotz M. Anandamide, an endogenous cannabinoid receptor agonist
inhibits lymphocyte proliferation and induces apoptosis. J Neuroimmunol. 1994;55(1):107115.

ED

91. Maccarrone M, Lorenzon T, Bari M, Melino G, Finazzi-Agro A. Anandamide induces apoptosis in human cells via vanilloid receptors. Evidence for a protective role of cannabinoid recep-

PT

tors. J. Biol. Chem. (2000) 275(41):3193831945.

CE

92. Zhu W, Friedman H, Klein TW. Delta9-tetrahydrocannabinol induces apoptosis in macrophages and lymphocytes: involvement of bcl-2 and caspase-1. J Pharmacol Exp Ther.

AC

1998;286(2):11031109.

93. McKallip RJ, Lombard C, Martin BR, Nagarkatti M, Nagarkatti PS. Delta(9)tetrahydrocannabinol-induced apoptosis in the thymus and spleen as a mechanism of immunosuppression in vitro and in vivo. J Pharmacol Exp Ther. 2002b;302(2):451465.
94. Lee CY, Wey SP, Liao MH, Hsu WL, Wu HY, Jan TR. A comparative study on cannabidiolinduced apoptosis in murine thymocytes and el-4 thymoma cells. Int Immunopharmacol.
2008;8(5):732740.
95. Klein TW, Newton C, Zhu W, Daaka Y, Friedman H. Delta 9-tetrahydrocannabinol, cytokines, and immunity to Legionella pneumophila. Proc Soc Exp Biol Med. 1995;209(3):205212.

ACCEPTED MANUSCRIPT
96. Derocq JM, Segui M, Marchand J, Le Fur G, Casellas P. Cannabinoids enhance human b-cell
growth at low nanomolar concentrations. FEBS Lett. 1995;369(23):177182.
97. Derocq JM, Segui M, Marchand J, Le Fur G, Casellas P. Cannabinoids enhance human b-cell

growth at low nanomolar concentrations. FEBS Lett. 1995;369(23):177182

IP

98. Yebra M, Klein TW, Friedman H. Delta 9-tetrahydrocannabinol suppresses concanavalin A

CR

induced increase in cytoplasmic free calcium in mouse thymocytes. Life Sci. 1992;51(2):151

NU
S

160.

99. Weiss L, Zeira M, Reich S, Har-Noy M, Mechoulam R, Slavin S, Gallily R. Cannabidiol


lowers incidence of diabetes in non-obese diabetic mice, Autoimmunity 2006: 39:2, 143-151.

MA

100. Watzl B, Scuderi P, Watson RR. Influence of marijuana components (THC and CBD) on
human mononuclear cell cytokine secretion in vitro. Adv Exp Med Biol 1991;288:63 70.

ED

101. Hollister LE. Marijuana and immunity. J Psychoactive Drugs. 1992;24:15964.

2003;88:4626.

PT

102. Zurier RB. Prospects for cannabinoids as anti-inflammatory agents. J Cell Biochem.

CE

103. Cabral GA, Dove Pettit DA. Drugs and immunity: cannabinoids and their role in decreased
resistance to infectious disease. J Neuroimmunol. 1998;83:11623.

AC

104. De Filippis D, D'Amico A, Iuvone T. Cannabinomimetic control of mast cell mediator release: new perspective in chronic inflammation. J Neuroendocrinol. 2008;20(1):205.
105. Montecucco F, Burger F, Mach F, Steffens S. CB2 cannabinoid receptor agonist JWH-015
modulates human monocyte migration through defined intracellular signaling pathways. Am J
Physiol Heart Circ Physiol. 2008; 294:H1145H1155.
106. Roth MD, Baldwin GC, Tashkin DP. Effects of delta-9-tetrahydrocannabinol on human
immune function and host defense. Chem Phys Lipids. 2002; 121:229239.
107. Klein TW, Cabral GA. Cannabinoid-induced immune suppression and modulation of antigen- presenting cells. J Neuroimmune Pharmacol. 2006; 1:5064.

ACCEPTED MANUSCRIPT
108. Raborn ES, Cabral GA. Cannabinoid inhibition of macrophage migration to the transactivating (Tat) protein of HIV-1 is linked to the CB(2) cannabinoid receptor. J Pharmacol Exp
Ther. 2010; 333:319327.

109. Patinkin D, Milman G, Breuer A, Fride E, Mechoulam R. Endocannabinoids as positive or

IP

negative factors in hematopoietic cell migration and differentiation. Eur J Pharmacol. 2008;

CR

595:16.

NU
S

110. Raborn ES, Marciano-Cabral F, Buckley NE, Martin BR, Cabral GA. The cannabinoid delta-9- tetrahydrocannabinol mediates inhibition of macrophage chemotaxis to RANTES/CCL5:
linkage to the CB2 receptor. J Neuroimmune Pharmacol. 2008; 3:117129.

MA

111. Miller AM, Stella N. CB2 receptor-mediated migration of immune cells: it can go either
way. Br J Pharmacol. 2008; 153:299308.

ED

112. Walter L, Franklin A, Witting A, Wade C, Xie Y, Kunos G, Mackie K, Stella N.

23:13981405.

PT

Nonpsychotropic cannabinoid receptors regulate microglial cell migration. J Neurosci. 2003;

CE

113. Sexton M, Silvestroni A, Moller T, Stella N. Differential migratory properties of


monocytes isolated from human subjects nave and non-nave to Cannabis.

AC

Inflammopharmacology. 2013 June ; 21(3): 253259.


114. Lu T, Newton C, Perkins I, Friedman H, Klein TW. Cannabinoid treatment suppresses the
t-helper cell-polarizing function of mouse dendritic cells stimulated with Legionella
pneumophila infection. J Pharmacol Exp Ther. 2006;319(1):269276.
115. Hegde VL, Hegde S, Cravatt BF, Hofseth LJ, Nagarkatti M, Nagarkatti PS. Attenuation of
experimental autoimmune hepatitis by exogenous and endogenous cannabinoids: Involvement of
regulatory t cells. Mol Pharmacol. 2008;74(1):2033.
116. El-Gohary M, Eid MA. Effect of cannabinoid ingestion (in the form of bhang) on the
immune system of high school and university students. Hum Exp Toxicol. 2004;23:14956.

ACCEPTED MANUSCRIPT
117. Rachelefsky GS, Opelz G, Mickey MR, Lessin P, Kiuchi M, Silverstein MJ, et al. Intact
humoral and cell-mediated immunity in chronic marijuana smoking. J Allergy Clin Immunol.
1976;58(4):483490.

118. Carlisle SJ, Marciano-Cabral F, Staab A, Ludwick C, Cabral GA. Differential expression of

CR

cell activation. Int Immunopharmacol. 2002;2:6982.

IP

the CB2 cannabinoid receptor by rodent macrophages and macrophage-like cells in relation to

NU
S

119. Correa F, Mestre L, Docagne F, Guaza C. Activation of cannabinoid CB2 receptor


negatively regulates IL-12p40 production in murine macrophages: role of IL-10 and ERK1/2
kinase signaling. Br J Pharmacol. 2005;145:4418

MA

120. Gokoh M, Kishimoto S, Oka S, Metani Y, Sugiura T. 2-Arachidonoylglycerol, an


endogenous cannabinoid receptor ligand, enhances the adhesion of HL-60 cells differentiated

ED

into macrophage-like cells and human peripheral blood monocytes. FEBS Lett. 2005;579:6473
8.

PT

121. Maresz K, Carrier EJ, Ponomarev ED, Hillard CJ, Dittel BN. Modulation of the

CE

cannabinoid CB2 receptor in microglial cells in response to inflammatory stimuli. J Neurochem.


2005;95:43745.

AC

122. Cabral GA, Mishkin EM. Delta-9-tetrahydrocannabinol inhibits macrophage protein


expression in response to bacterial immunomodulators. J Toxicol Environ Health.
1989;26(2):175182.
123. Lopez-Cepero M, Friedman M, Klein T, Friedman H. Tetrahydrocannabinol-induced
suppression of macrophage spreading and phagocytic activity in vitro. J Leukoc Biol.
1986;39(6):679686.
124. McCoy KL, Gainey D, Cabral GA. Delta 9-tetrahydrocannabinol modulates antigen
processing by macrophages. J Pharmacol Exp Ther. 1995;273(3):12161223.

ACCEPTED MANUSCRIPT
125. Wang M, Richards AL, Friedman H, Djeu JY. Selective inhibition of natural killer but not
natural cytotoxic activity in a cloned cell line by delta-9-tetrahydrocannabinol. J Leukoc Biol.
1991;50:1927.

126. Pross S, Nakano Y, Smith J, Widen R, Rodriguez A, Newton C, et al. Suppressive effect of

IP

tetrahydrocannabinol on specific T cell subpopulations in the thymus. Thymus. 1992;19:97104.

CR

127. Fischer-Stenger K, Updegrove AW, Cabral GA. Delta 9-tetrahydrocannabinol decreases

NU
S

cytotoxic T lymphocyte activity to herpes simplex virus type 1-infected cells. Proc Soc Exp Biol
Med. 1992;200:42230.

128. Samson MT, Small-Howard A, Shimoda LM, Koblan-Huberson M, Stokes AJ, Turner H.

MA

Differential Roles of CB1 and CB2 Cannabinoid Receptors in Mast Cells. J Immunol.
2003;170:495362.

ED

129. Small-Howard AL, Shimoda LM, Adra CN, Turner H. Anti-inflammatory potential of CB1mediated cAMP elevation in mast cells. Biochem J. 2005; 25:25.

PT

130. Ueda Y, Miyagawa N, Wakitani K. Involvement of cannabinoid CB2 receptors in the IgE-

CE

mediated triphasic cutaneous reaction in mice. Life Sci. 2007;80:4149


131. Pryce G, Ahmed Z, Hankey DJ, Jackson SJ, Croxford JL, Pocock JM, Ledent C, Petzold A,

AC

Thompson AJ, Giovannoni G, Cuzner ML, Baker D. Cannabinoids inhibit neurodegeneration in


models of multiple sclerosis. Brain 2003;126(Pt10):21912202.

132. Fujiwara M, Egashira N. New perspectives in the studies on endocannabinoid and cannabis: abnormal behaviors associate with CB1 cannabinoid receptor and development of therapeutic application. J Pharmacol Sci 2004;96(4):362366.
133. Jackson SJ, Pryce G, Diemel LT, Cuzner ML, Baker D. Cannabinoid-receptor 1 null mice
are susceptible to neurofilament damage and caspase 3 activation. Neuroscience
2005;134(1):261268.
134. Pryce G, Baker D. Control of spasticity in a multiple sclerosis model is mediated by CB1,
not CB2, cannabinoid receptors. Br. J Pharmacol 2007;150(4):519525.

ACCEPTED MANUSCRIPT
135. Cabranes A, Venderova K, de Lago E, Fezza F, Sanchez A, Mestre L, Valenti M, GarciaMerino A, Ramos JA, Di Marzo V, Ruiz. Decreased endocannabinoid levels in the brain and
beneficial effects of agents activating cannabinoid and/or vanilloid receptors in a rat model of

multiple sclerosis. J. Neurobiol Dis 2005;20(2):207217.

IP

136. Arevalo-Martin A, Vela JM, Molina-Holgado E, Borrell J, Guaza C. Therapeutic action of

CR

cannabinoids in a murine model of multiple sclerosis. J Neurosci 2003;23(7):2511 2516.

NU
S

137.Eljaschewitsch E, Witting A, Mawrin C, Lee T, Schmidt PM, Wolf S, Hoertnagl H, Raine


CS, Schneider-Stock R, Nitsch R, Ullrich O. The endocannabinoid anandamide protects neurons
during CNS inflammation by induction of MKP-1 in microglial cells. Neuron 2006;49(1):6779.

MA

138. Ball S, Vickery J, Hobart J, Wright D, Green C, Shearer J, Nunn A, Cano MG, MacManus
D, Miller D, Mallik S, Zajicek J. The Cannabinoid Use in Progressive Inflammatory brain Dis-

ED

ease (CUPID) trial: a randomised double-blindplacebo-controlled parallel-group multicentre trial


and economic evaluation of cannabinoids to slow progression in multiple sclerosis; Health Tech-

PT

nology Assessment. Vol 19 2015 ISSN 1366-5278

CE

139. Notcutt LW, Langford R, Davies P, Ratcliffe S, Potts R. A placebo-controlled, parallelgroup, randomized withdrawal study of subjects with symptoms of spasticity due to multiple

AC

sclerosis who are receiving long-term Sativex (nabiximols). Eur J Pain. 2009 May Mult Scler.
2012 Feb;18(2):219-28

140 .Conte A, Bettolo CM, Onesti E, Frasca V, Iacovelli E, Gilio F, Giacomelli E, Gabriele M,
Aragona M, Tomassini V, Pantano P, Pozzilli C, Inghilleri M.); Cannabinoid-induced effects on
the nociceptive system: a neurophysiological study in patients with secondary progressive multiple sclerosis Eur J Pain. 2009 May;13(5):472-7.
141. Collin C, Davies P, Mutiboko IK, Ratcliffe S; Sativex Spasticity in MS Study Group. Randomized controlled trial of cannabis-based medicine in spasticity caused by multiple sclerosis;
Eur J Neurol. 2007 Mar;14(3):290-6

ACCEPTED MANUSCRIPT
142. Goldring MB, Otero M. Inflammation in osteoarthritis. Curr Rheumatol 2011;23:471-8,
143.Sumariwalla PF, Gallily R, Tchilibon S, Fride E, Mechoulam R, Feldmann M. A novel synthetic, nonpsychoactive cannabinoid acid (HU-320) with antiinflammatory properties in murine

collagen-induced arthritis. Arthritis Rheum 2004;50:985-98

IP

144. Zurier RB, Rossetti RG, Lane JH, Goldberg JM, Hunter SA, Burstein SH. Dimethylheptyl-

CR

THC-11 oic acid: a nonpsychoactive antiinflammatory agent with a cannabinoid template struc-

NU
S

ture. Arthritis Rheum 1998;41:163-70.

145. Malfait AM, Gallily R, Sumariwalla PF, Malik AS, Andreakos E, Mechoulam R, et al. The
nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in murine

MA

collagen-induced arthritis. Proc Natl Acad Sci U S A 2000;97: 9561-6,


146. Zurier RB, Rossetti RG, Burstein SH, Bidinger B. Suppression of human monocyte inter-

ED

leukin-1beta production by ajulemic acid, a nonpsychoactive cannabinoid. Biochem Pharmacol


2003;65:649-55.

PT

147. Selvi E, Lorenzini S, Garcia-Gonzalez E, Maggio R, Lazzerini PE, Capecchi PL, et al. In-

CE

hibitory effect of synthetic cannabinoids on cytokine production in rheumatoid fibroblast-like


synoviocytes. Clin Exp Rheumatol 2008;26:574-81.

AC

148. Johnson DR, Stebulis JA, Rossetti RG, Burstein SH, Zurier RB. Suppression of fibroblast
metalloproteinases by ajulemic acid, a nonpsychoactive cannabinoid acid. J Cell Biochem
2007;100: 184-90.

149. Mbvundula EC, Bunning RA, Rainsford KD. Arthritis and cannabinoids: HU-210 and Win55,212-2 prevent IL-1alphainduced matrix degradation in bovine articular chondrocytes in-vitro.
J Pharm Pharmacol 2006;58:351-8.
150. Pertwee RG, Howlett AC, Abood ME, Alexander SP, Di Marzo V, Elphick MR, et al. Cannabinoid receptors and their ligands: beyond CB and CB. Pharmacol Rev 2010;62:588-631,
151. Sun Y, Alexander SP, Kendall DA, Bennett AJ. Cannabinoids and PPARalpha signalling.
Biochem Soc Trans 2006; 34:1095-7,

ACCEPTED MANUSCRIPT
152. OSullivan SE, Kendall DA. Cannabinoid activation of peroxisome proliferator-activated
receptors: potential for modulation of inflammatory disease. Immunobiology 2010;215:611e6
153. Percy F. Sumariwalla,1 Ruth Gallily,2 Susanna Tchilibon,2 Ester ; A Novel Synthetic,

Nonpsychoactive Cannabinoid Acid (HU-320) With Antiinflammatory Properties in Murine

IP

Collagen-Induced Arthritis. Arth and Rheum Vol. 50, No. 3, March 2004, pp 985998.

CR

154. Agarwal N, Pacher P, Tegeder I, Amaya F, Constantin CE, Brenner GJ, Rubino T, Mi-

NU
S

chalski CW, Marsicano G, Monory K, Mackie K, Marian C, Batkai S, Parolaro D, Fischer MJ,
Reeh P, Kunos G, Kress M, Lutz B, Woolf CJ, Kuner R. Cannabinoids mediate analgesia largely
via peripheral type 1 cannabinoid receptors in nociceptors. Nat. Neurosci 2007 10, 870879.

MA

155. Chappell AS, Desaiah D, Liu-Seifert H, Zhang S, Skljarevski V, Belenkov Y, Brown JP. A
double-blind, randomized, placebocontrolled study of the efficacy and safety of duloxetine for

ED

the treatment of chronic pain due to osteoarthritis of the knee. Pain Pract. 2001 11, 3341.
156. Millns PJ, Chapman V, Kendall DA. Cannabinoid inhibition of the capsaicin-induced cal-

PT

cium response in rat dorsal root ganglion neurones. Brit. J. Pharmacol. 2001, 132, 969971.

CE

157. Anand U, Otto WR, Sanchez-Herrera D, Facer P, Yiangou Y, Korchev Y, Birch R, Benham
C, Bountra C, Chessell IP, Anand P. Cannabinoid receptor CB2 localisation and agonist-

AC

mediated inhibition of capsaicin responses in human sensory neurons. Pain2008: 138, 667680.
158. Meng ID, Manning BH, Martin WJ, Fields HL. An analgesia circuit activated by cannabinoids. Nature 1998: 395, 381383.
159. Schuelert N, McDougall JJ. Cannabinoid-mediated antinociception is enhanced in rat osteoarthritic knees. Arthritis Rheum. 2008: 58, 145153.
160. Richardson D, Pearson RG, Kurian N, Latif ML, Garle MJ, Barrett DA, Kendall DA,
Scammell BE, Reeve AJ, Chapman V. Characterisation of the cannabinoid receptor system in
synovial tissue and fluid in patients with osteoarthritis and rheumatoid arthritis. Arthritis Res.
Ther. 2008, 10, R43.

ACCEPTED MANUSCRIPT
161. Sagar DR, Staniaszek LE, Okine BN, Woodhams S, Norris LM, Pearson RG, Garle MJ,
Alexander SP, Bennett AJ, Barrett DA, Kendall DA, Scammell BE, Chapman V. Tonic modulation of spinal hyperexcitability by the endocannabinoid receptor system in a rat model of os-

teoarthritis pain. Arthritis Rheum.2010, 62, 36663676

IP

162. Blake DR, et al. Preliminary assessment of the efficacy, tolerability and safety of a canna-

CR

bisbased medicine (Sativex) in the treatment of pain caused by rheumatoid arthritis. Rheumatol-

NU
S

ogy, 2006. 45(1): p. 50-2.

163. Lynch ME; CampbellF Cannabinoids for treatment of chronic non-cancer pain; a systematic

MA

review of randomized trials. Br J Clin Pharmacol. 2011 Nov;72(5):735-44.

164. Dziadzio M, Smith E, Abraham DJ et al. Serological assessment of type I collagen burden

ED

in scleroderma spectrum disorders: a systematic review. Clin Exp Rheumatol 2004;22:35667.


165. Duncan MR, Berman B. Stimulation of collagen and glycosaminoglycan production in cul-

CE

1991;97:68692.

PT

tured human adult dermal fibroblasts by recombinant human interleukin 6. J Invest Dermatol

166. Ellert-Miklaszewska A, Kaminska B, Konarska L. Cannabinoids down-regulate PI3K/ akt

AC

and Erk signalling pathways and activate proapoptotic function of bad protein. Cell Signal
2005;17:2537.

167. Garcia-Gonzalez E, Galeazzi M; Selvi E. Are Cannabinoids able to modulate fibrotic progression in Systemic Sclerosis? Journal.
168. Klein TW. Cannabinoid-based drugs as anti-inflammatory therapeutics. Nat Rev Immunol
2005;5:40011.
169. Lazzerini PE, Natale M, Gianchecchi E et al. Adenosine A2A receptor activation stimulates
collagen production in sclerodermic dermal fibroblasts either directly and through a cross-talk
with the cannabinoid system. J Mol Med (Berl). 2012;90:331-42.

ACCEPTED MANUSCRIPT
170. Gonzalez EG, Selvi E, Balistreri E et al. Synthetic cannabinoid ajulemic acid exerts potent
antifibrotic effects in experimental models of systemic sclerosis. Ann Rheum Dis. 2012;71:154551.

171. Atkinson MA, Leiter EH . The NOD mouse model of type 1 diabetes: as good as it gets?

IP

Nat Med 1999 5:601604.

CR

172. Mandrup-Poulsen T. Beta cell death and protection. Ann NY Acad Sci 2003 1005:3242.

NU
S

173. Di Marzo V, Piscitelli F, Mechoulam R.Cannabinoids and endocannabinoids in metabolic


disorders with focus on diabetes. Handb Exp Pharmacol. 2011;(203):75-104.
174. Weiss L, Zeira M, Reich S et al. Cannabidiol arrests onset of autoimmune diabetes in NOD

MA

mice. Neuropharmacol 2008: 54:244249.

175. Satoh J, Takahashi K, Nakazawa T, Sakata Y, Toyota T. Current concept of the pathogene-

ED

sis of autoimmune type 1 diabetes mellitus. Nippon Rinsho 1999;57:528533.


176. Swain SL. T-cell subsets. Who does the polarizing? Curr Biol 1995;5:849 851.

PT

177. Jun HS, Santamaria P, Lim HW, Zhang ML, Yoon JW. Absolute requirement of macro-

CE

phages for the development and activation of beta-cell cytotoxic CD8+T-cells in T-cell receptor
transgenic NOD mice. Diabetes 1999; 48:34-42.

AC

178. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity.
Nat Rev Immunol 2003; 3:133 146.
179. Skeen MJ, Miller MA, Shinnick TM, Ziegler HK. Regulation of murine macrophage IL-12
production. Activation of macrophages in vivo, restimulation in vitro, and modulation by other
cytokines. J Immunol 1996; 156:1196 1206.
180. Ohno Y, Aoki N, Nishimura A. In vitro production of interleukin-1, interleukin-6, and tumor necrosis factor-alpha in insulin-dependent diabetes mellitus. J Clin Endocrinol Metab 1993;
77:1072 1077.
181. Schicho R, Storr M. Cannabis finds its way into treatment of Crohns disease. Pharmacology. 2014; 93 (0): 1-3

ACCEPTED MANUSCRIPT
182. Izzo AA, Capasso F, Costagliola A, Bisogno T, Marsicano G, Ligresti A, Matias I, Capasso
R, Pinto L, Borrelli F, Cecio A, Lutz B, Mascolo N, DiMarzo V. An endogenous cannabinoid
tone attenuates cholera toxin-induced fluid accumulation in mice. Gastroenterology 2003;

125:765774.

IP

183. Massa F, Marsicano G, Hermann H, Cannich A, et al. The endogenous cannabinoid system

CR

protects against colonic inflammation. J Clin Invest 2004; 113:12021209

NU
S

184. Engel MA, Kellermann CA, Burnat G, Hahn EG, et al: Mice lacking cannabinoid CB1-,
CB2-receptors or both receptors show in- creased susceptibility to trinitrobenzene sulfonic acid
(TNBS)-induced colitis. J Physiol Pharmacol 2010; 61:8997.

MA

185. Borrelli F, Aviello G, Romano B, Orlando P, et al: Cannabidiol, a safe and nonpsychotropic ingredient of the marijuana plant Canna- bis sativa, is protective in a murine model

ED

of colitis. J Mol Med (Berl) 2009; 87:11111121.


186. Schicho R, Storr, M: Topical and systemic cannabidiol improves trinitrobenzene sulfonic

PT

acid colitis in mice. Pharmacology 2012; 89: 149155.

CE

187. Jamontt JM, Molleman A, Pertwee RG, Par- sons ME: The effects of delta-tetrahydrocannabinol and cannabidiol alone and in combination on damage, inflammation and in vitro motility

AC

disturbances in rat colitis. Br J Phar- macol 2010; 160:712723.


188. Kimball ES, Schneider CR, Wallace NH, Hornby PJ: Agonists of cannabinoid receptor 1
and 2 inhibit experimental colitis induced by oil of mustard and by dextran sulfate sodium. Am J
Physiol Gastrointest Liver Physi- ol 2006; 291:G364G371.
189. Alhouayek M, Lambert DM, Delzenne NM, Cani PD, et al: Increasing endogenous 2-arachidonoylglycerol levels counteracts colitis and related systemic inflammation. FASEB J 2011;
25: 27112721.
190. Di Sabatino A, Battista N, Biancheri P, Rapino C, Rovedatti L, Astarita G, Vanoli A, Dainese E, Guerci M, Piomelli D, Pender SL, MacDonald TT, Maccarrone M, Corazza GR. The en-

ACCEPTED MANUSCRIPT
dogenous cannabinoid system in the gut of patients with inflammatory bowel disease. Mucosal
Immunol. 2011; 4: 574583.
191. Di Carlo G, Izzo AA. Cannabinoids for gastrointestinal diseases: potential therapeutic ap-

plications. Expert Opin Investig Drugs. 2003; 12(1):3949.

IP

192. Zalesky A, Solowij N, Yucel M, Lubman DI, Takagi M, Harding IH, Lorenzetti V, Wang

CR

R, Searle K, Pantelis C, Seal M: Effect of long-term cannabis use on axonal fiber connectivity.

NU
S

Brain 2012; 135:22452255.

193. Marquz L, Surez J, Iglesias M, Bermudez-Silva FJ, Rodrguez de Fonseca F, Andreu M.


Ulcerative colitis induces changes on the expression of the endocannabinoid system in the hu-

MA

man colonic tissue. PLoS One. 2009; 4: 6893.

194. Wright K, Rooney N, Feeney M, Tate J, Robertson D, Welham M, Ward S. Differential ex-

ED

pression of cannabinoid receptors in the human colon: cannabinoids promote epithelial wound
healing. Gastroenterology. 2005; 129:437453.

PT

195. Singh UP, Singh NP, Singh B, Price RL, Nagarkatti M, Nagarkatti PS. Cannabinoid

CE

196. Izzo AA, Sharkey KA. Cannabinoids and the gut: new developments and emerging concepts. Pharmacol Ther. 2010; 126:2138.

AC

197. Alhouayek M, Muccioli GG. The endocannabinoid system in inflammatory bowel diseases:
from pathophysiology to therapeutic opportunity. Trends Mol Med. 2012; 18:615625.
198. Izzo AA, Camilleri M. Emerging role of cannabinoids in gastrointestinal and liver diseases:
basic and clinical aspects. Gut 2008; 57:11401155.
199. Cluny NL, Keenan CM, Duncan M, Fox A, Lutz B, Sharkey KA. Naphthalen-1-yl-(4- pentyloxynaphthalen-1-yl) methanone (SAB378), a peripherally restricted cannabinoid CB1/CB2
receptors
agonist, inhibits gastrointestinal motility but has no effect on experimental colitis in

mice. J Pharmacol Exp Ther. 2010; 334: 973980.

ACCEPTED MANUSCRIPT
200. Naftali T, Schleider LBL, Dotan I, Lansky EP, Benjaminov FS, Konikoff FM. Cannabis
Induces a Clinical Response in Patients With Crohns Disease: A Prospective PlaceboControlled Study. Clin. Gastroenterol. Hepatol. 11; 1276-1280.

201. Lal S, Prasad N, Ryan M, Tangri S, Silverberg MS, Gordon A, Steinhart H. Cannabis use

IP

amongst patients with inflammatory bowel disease. Eur J Gastroenterol Hepatol 2011 23:891

CR

896.

NU
S

202. Naftali T, Mechulam R, Lev LB, Konikoff FM. Cannabis for Inflammatory Bowel Disease.
Dig Dis 2014;32: 468474.

203. Whelan G, Farmer RG, Fazio VW, et al. Recurrence after surgery in Crohn's disease. Rela-

MA

tionship to location of disease (clinical pattern) and surgical indication. Gastroenterology 1985;
88: 1826-33.

ED

204. Naftali T, Lev LB, Yablekovitz D, Half E, Konikoff FM. Treatment of crohns disease with
cannabis: an Observational study. IMAJ 2011 (13) 455-458.

PT

205. Baker D, Pryce G, Giovannoni G, Thompson AJ. The therapeutic potential of cannabis.

CE

Lancet Neurol 2003; 2:291298.


206. Lahat A, Lang A, Ben-Horin S: Impact of can- nabis treatment on the quality of life, weight

AC

and clinical disease activity in inflammatory bowel disease patients: a pilot prospective study.
Digestion 2012; 85:18.
207. Wilsey B, Marcotte TD, Deutsch R, Gouaux B, Sakai S, Donaghe H. Low Dose Vaporized
Cannabis Significantly Improves Neuropathic Pain. J Pain. 2013 February; 14(2): 136148.
208. Dworkin RH, Turk DC, Walco GA, Wells CD, OConnor AB, Audette J, Baron R, Gourlay
GK, Haanpaa ML, Baron R, Haanp ML, Kent JL, Krane EJ, Lebel AA, Levy RM, Mackey
SC, Mayer J, Miaskowski C, Raja SN, Rice AS, Schmader KE, Stacey B, Stanos S, Treede RD.
Recommendations for the pharmacological management of neuropathic pain: an overview and
literature update. Mayo Clinic Proceedings. 2010; 85:S314.

ACCEPTED MANUSCRIPT
209. Rahn EJ, Hohmann AG. Cannabinoids as pharmacotherapies for neuropathic pain: from the
bench to the bedside. Neurotherapeutics. 2009; 6:71337.
210. Lynch ME. The need for a Canadian pain strategy. Pain Res Manage 2011; 16: 7780.

211. Tang N, Crane C. Suicidality in chronic pain: review of the prevalence, risk factors and

IP

psychological links. Psychol Med 2006; 36: 57586.

CR

212. Lynch M, Campbell F. Cannabinoids for treatment of chronic non-cancer pain; a systematic

NU
S

review of randomized trials. Br J Clin Pharmacol 72:5: 735744.


213. Campbell FA, Tramr MR, Carroll D. et al. Are cannabinoids an effective and safe treatment option in the management of pain? BMJ.2001; 323:1-6.

Pharm Des.2000;6:1339-1345.

MA

214. Burstein SH. Ajulemic acid (CT3): a potent analog of the acid metabolites of THC. Curr

ED

215. Liu J, Li H, Burstein SH. et al. Activation and binding of peroxisome proliferator-activated
receptor by synthetic cannabinoid ajulemic acid. Mol Pharmacol.2003; 63:983-992.

PT

216. Burstein SH, Friderichs E, Kgel B. et al. Analgesic effects of 1,1dimethylheptyl-delta8-

CE

THC-11-oic acid (CT3) in mice. Life Sci.1998; 63:161-168.


217. Karst M, Salim K, Burstein S, Conrad I, Hoy L, Schneider U. Analgesic Effect of the Syn-

AC

thetic Cannabinoid CT-3 on Chronic Neuropathic Pain. A Randomized Controlled Trial. JAMA
2003; 290(13):1757-1762.
218. Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, Kelly ME, Rowbotham MC,
Petersen KL. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebocontrolled trial. Neurology. 2007; 68: 51521.
219. Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, Bentley H,
Atkinson JH. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover
clinical trial. Neuropsychopharmacology. 2009; 34: 67280.
220. Phillips TJC, Cherry CL, Moss PJ, Rice ASC. Painful HIV-associated sensory neuropathy.
Pain Clin Updates 2010; XVIII: 18.

ACCEPTED MANUSCRIPT
221. Wallace M, Schulteis G, Atkinson JH, Wolfson T, Lazzaretto D, Bentley H, Gouaux B, Abramson I. Dose-dependent effects of smoked cannabis on capsaicin-induced pain and hyperalgesia in healthy volunteers. Anesthesiology. 2007; 107: 78596.

222. Ware MA, Wang T, Shapiro S, Robinson A, Ducruet T, Huynh T, Gamsa A, Bennett GJ,

IP

Collet JP. Smoked cannabis for chronic neuropathic pain: a randomized controlled trial. CMAJ

CR

2010 182(14).

NU
S

223 Wolfe F (February 1989). "Fibromyalgia: the clinical syndrome". Rheum Dis Clin North
Am 15 (1): 118.

MA

224: McBeth J, Tomenson B, Chew-Graham CA, Macfarlane GJ, Jackson J, Littlewood A,


Creed FH.Common and unique associated factors for medically unexplained chronic widespread

ED

pain and chronic fatigue. J Psychosom Res. 2015 Dec; 79(6):484-91.


225. 5Z: Basu S, Dittel BH. Unraveling the complexities of cannabinoid receptor 2 (CB2) im-

PT

mune regulation in health and disease. Immunologic Research. 2011 51(1): 2638.

CE

226. Bohr T. Problems with myofascial pain syndrome and fibromyalgia syndrome. Neurology
1996. 46(3): 593597.

AC

227. Richardson JD, Aanonsen L, Hargreaves KM. SR 141716A, a cannabinoid receptor antagonist, produces hyperalgesia in untreated mice. Eur J Pharmacol 1997 319(23): R34.
228. Fiz VJ, Duran M, Capella D, Carbonell J, Farre M. Cannabis Use in Patients with Fibromyalgia: Effect on Symptoms Relief and Health-Related Quality of Life. PLoS ONE. Volume 6.
Issue 4.18440
229. Ware MA, Fitzcharles MA, Joseph L, Shir Y. The effects of nabilone on sleep in
fibromyalgia: results of a randomized controlled trial . Anesth Analg. 2010 Feb 1;110(2):604-10
230. Wang T, Collet J-P, Shapiro S, Ware MA. Adverse effects of medical cannabinoids: a systematic review. CMAJ: Canadian Medical Association Journal. 2008; 178(13):1669-1678.

ACCEPTED MANUSCRIPT
231. Noyes R, Jr, Brunk SF, Baram DA, Canter A. Analgesic effect of delta-9tetrahydrocannabinol. J Clin Pharmacol. 1975; 15(23): 13943.
232. Noyes R, Brunk SF, Avery DH, Canter A. The analgesic properties of delta-9-

IP

tetrahydrocannabinol and codeine. Clin Pharmacol Ther. 1975; 18:849.


233. Jain AK, Ryan JR, McMahon FG, Smith G. Evaluation of intramuscular levonantradol and

CR

plecebo in acute postoperative pain. J Clin Pharmacol. 1981; 21: 320S36.

NU
S

234. Chang AE, Shiling DJ, Stillman RC, et al. A prospective evaluation of delta-9tetrahydrocannabinol as an antiemetic in patients receiving adriamycin and cytoxan chemotherapy. Cancer. 1981; 47: 174651.

MA

235. Vinciguerra V, Moore T, Brennan E. Inhalation marijuana as an antiemetic for cancer chemotherapy. N Y State J Med. 1988; 88(10): 5257.

ED

236. Orr LE, McKernan JF, Bloome B. Antiemetic effect of tetrahydrocannabinol. Compared

PT

with placebo and prochlorperazine in chemotherapy-associated nausea and emesis. Arch of Internal Medicine. 1980; 140: 14313.

CE

237. Sallan SE, Cronin C, Zelen M, Zinberg NE. Antiemetics in patients receiving chemotherapy

AC

for cancer: a randomized comparison of delta-9-tetrahydrocannabinol and prochlorperazine. N


Engl J Med. 1980; 302(3):1358.
238. Steele N, Gralla RJ, Braun DW, Jr, Young CW. Double-blind comparison of the antiemetic
effects of nabilone and prochlorperazine on chemotherapy-induced emesis. Cancer Treat Rep.
1980; 64(23): 21924.
239. Tyson LB, Gralla RJ, Clark RA, Kris MG, Bordin LA, Bosl GJ. Phase 1 trial of levonantradol in chemotherapy-induced emesis. Am J Clin Oncol. 1985; 8(6): 52832.
240. Gorter R. Management of anorexia-cachexia associated with cancer and HIV infection. Oncology Suppl. 1991; 5: 1317.
241. Struwe M, Kaempfer SH, Geiger CJ, et al. Effect of dronabinol on nutritional status in HIV
infection. Ann Pharmacother. 1993; 27(78): 82731.

ACCEPTED MANUSCRIPT
242. Beal JE, Olson RLL, Morales JO, et al. Dronabinol as a treatment for anorexia associated
with weight loss in patients with AIDS. J Pain Symptom Manage. 1995; 10: 8997.
243. Beal JE, Olson R, Lefkowitz L, et al. Long-term efficacy and safety of dronabinol for ac-

quired immunodeficiency syndrome-associated anorexia. J Pain Symptom Manage. 1997; 14: 7

IP

14.

CR

244. National Academy of Science, Institute of Medicine. The medical value of marijuana and

NU
S

related substances. Marijuna and Medicine, Assessing the Science Base. National Academy
Press; 1999.

245. Ungerleider JT, Andyrsiak T, Fairbanks L, Ellison GW, Myers LW. Delta-9-THC in the

MA

treatment of spasticity associated with multiple sclerosis. Adv Alcohol Subst Abuse. 1987;
7(1):3950.

ED

246. Brady CM, DasGupta R, Dalton C, Wiseman OJ, Berkley KJ, Fowler CJ. An open-label

PT

pilot study of cannabis-based extracts for bladder dysfunction in advanced multiple sclerosis.
Mult Scler. 2004; 10(4):42533.

CE

247. Kunos G, Pacher P. Cannabinoids cool the intestine. Nat Med. 2004;10(7):6789.

AC

248. Bonf, L. Vinagre RCO, de Figueiredo NV. Cannabinoids in chronic pain and palliative
care. Revista Brasileira de Anestesiologia, 2008 58(3), 267-279.
249. Siegling A, Hofmann HA, Denzer D Cannabinoid CB1 receptor upregulation in a rat
model of chronic neuropathic pain. Eur J Pharmacol, 2001; 415:5-7.
250. Julien RM. A Primer of Drug Action: a Concise, Nontechnical Guide to the Actions, Uses,
and Side Effects of Psychoactive Drugs, 8th Ed., New York, Henry Holt, 1997; 548.
251. Kalant H. Medicinal use of cannabis: history and current status. Pain Res Manag, 2001; 6:
80-91.
252. Cerro JC. Drogodependencias. Cannabis. Madri, Panamericana, 1998; 191-214.
253. MacRae EJBN- Reduo de Danos para o Uso da Cannabis: Panorama Atual de Drogas e
Dependncias. 1 Ed., So Paulo, Atheneu, 2006; 361-370.

ACCEPTED MANUSCRIPT
254. Moreira FA, Grieb M, Lutz B. Central side-effects of therapies based on CB1 cannabinoid
receptor agonists and antagonists: focus on anxiety and depression. Best Pract Res Clin Endocrinol Metab. 2009; 23: 13344.

IP

255. Ghodse AH. Cannabis psychosis. Br J Addict. 1986; 81: 4738.

256. Hall W, Degenhardt L. Cannabis use and the risk of developing a psychotic disorder. World

CR

Psychiatry. 2008; 7: 6871.

NU
S

257. Shukla PC, Moore UB. Marijuana-induced transient global amnesia. South Med J. 2004;
97: 7824.

258. Battistella G, Fornari E, Annoni J-M, et al. Long-Term Effects of Cannabis on Brain Struc-

MA

ture. Neuropsychopharmacology. 2014; 39(9): 2041-2048


259. Mayoral M, Calderon H, Cano R, Lombardero M. Allergic rhinoconjunctivitis caused by

ED

Cannabis sativa pollen. J Investig Allergol Clin Immunol. 2008; 18: 734.

PT

260. Williams C, Thompstone J, Wilkinson M. Work-related contact urticaria to Cannabis sativa.


Contact Dermatitis. 2008; 58: 623.

CE

261. Buchy L, Seidman LJ, Cadenhead KS, Cannon TD, Cornblatt BA, McGlashan TH, Perkins

AC

DO, Stone W, Tsuang MT, Walker EF, Woods SW, Bearden CE, Mathalon DH, Addington J.
Evaluating the relationship between cannabis use and IQ in youth and young adults at clinical
high risk of psychosis. Psychiatry Res. 2015 Dec 30; 230(3): 878-84
262. Grant I, Cahn BR. Cannabis and endocannabinoid modulators: Therapeutic promises and
challenges. Clin Neurosci Res. 2005; 5(2-4): 185199.
263. El-Gohary M, Eid MA. Effect of cannabinoid ingestion (in the form of bhang) on the immune system of high school and university students. Hum Exp Toxicol. 2004; 23: 14956.
264. Degenhardt L, Hall WD. The adverse effects of cannabinoids: implications for use of medical marijuana. Cmaj. 2008; 178: 16856.
265. Hill S, Thomas SH. Recreational drug toxicity. Clin Med. 2008; 8: 99103.

ACCEPTED MANUSCRIPT
266. Rubino T, Parolaro D. Long lasting consequences of cannabis exposure in adolescence. Mol
Cell Endocrinol. 2008; 286: S10813.
267. Tashkin DP. Smoked marijuana as a cause of lung injury. Monaldi Arch Chest Dis. 2005;

IP

63:93100.

268. Kaminski NE. Regulation of the cAMP cascade, gene expression and immune function by

CR

cannabinoid receptors. J Neuroimmunol. 1998; 83: 12432.

NU
S

269. Maykut MO. Health consequences of acute and chronic marihuana use. Prog Neuropsychopharmacol Biol Psychiatry. 1985; 9: 20938.

270. Klein TW, Newton C, Friedman H. Cannabinoid receptors and immunity. Immunol Today.

AC

CE

PT

ED

MA

1998; 19; 37381.

Você também pode gostar