Cannabinoids and inflammation: implications for people living with HIV : AIDS

Secondary Logo

Journal Logo

EDITORIAL REVIEW

Cannabinoids and inflammation

implications for people living with HIV

Costiniuk, Cecilia T.a,b,c,d; Jenabian, Mohammad-Alid,e,f

Author Information
AIDS 33(15):p 2273-2288, December 1, 2019. | DOI: 10.1097/QAD.0000000000002345
  • Free

Abstract

 

Thanks to the success of modern antiretroviral therapy (ART), people living with HIV (PLWH) have life expectancies which approach that of persons in the general population. However, despite the ability of ART to suppress viral replication, PLWH have high levels of chronic systemic inflammation which drives the development of comorbidities such as cardiovascular disease, diabetes and non-AIDS associated malignancies. Historically, cannabis has played an important role in alleviating many symptoms experienced by persons with advanced HIV infection in the pre-ART era and continues to be used by many PLWH in the ART era, though for different reasons. Δ9-Tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD) are the phytocannabinoids, which have received most attention for their medicinal properties. Due to their ability to suppress lymphocyte proliferation and inflammatory cytokine production, there is interest in examining their therapeutic potential as immunomodulators. CB2 receptor activation has been shown in vitro to reduce CD4+ T-cell infection by CXCR4-tropic HIV and to reduce HIV replication. Studies involving SIV-infected macaques have shown that Δ9-THC can reduce morbidity and mortality and has favourable effects on gut mucosal immunity. Furthermore, Δ9-THC administration was associated with reduced lymph node fibrosis and diminished levels of SIV proviral DNA in spleens of rhesus macaques compared with placebo-treated macaques. In humans, cannabis use does not induce a reduction in peripheral CD4+ T-cell count or loss of HIV virological control in cross-sectional studies. Rather, cannabis use in ART-treated PLWH was associated with decreased levels of T-cell activation, inflammatory monocytes and pro-inflammatory cytokine secretion, all of which are related to HIV disease progression and comorbidities. Randomized clinical trials should provide further insights into the ability of cannabis and cannabinoid-based medicines to attenuate HIV-associated inflammation. In turn, these findings may provide a novel means to reduce morbidity and mortality in PLWH as adjunctive agents to ART.

Introduction

Historically, cannabis was used by people living with HIV (PLWH) to alleviate nausea, vomiting, pain, wasting and anorexia [1,2]. Since the advent of combined antiretroviral therapy (ART), PLWH no longer suffer from anorexia and wasting syndrome, and prolonged nausea and vomiting because of ART is rare. In the modern ART era, preventing and managing common comorbidities seen in the general population, such as cardiovascular disease, diabetes and non-AIDS associated cancers, is a key priority [3,4]. HIV itself appears to be a risk factor for these comorbidities and appears to interact with traditional risk factors [5]. Chronic HIV infection is associated with a state of persistent systemic immune activation, which is thought to be a primary driver for many comorbidities such as atherosclerosis and liver fibrosis, in PLWH [6].

Although cannabis has a long history of being used for therapeutic purposes, its anti-inflammatory and immunomodulatory properties make it especially fascinating in the context of chronic HIV infection. Although cannabis use remains common among PLWH in the modern ART era for alleviating symptoms of anxiety and depression and for recreational reasons [2,7], its effects on the immune system in the context of chronic HIV infection merit close attention. Herein, we review the data pertaining to the anti-inflammatory and immunomodulatory effect of cannabis and cannabinoid-based medicines in PLWH.

HIV infection as a chronic inflammatory disease

For the past 25 years, the majority of research on HIV has focused on inhibiting HIV replication. Most people living with HIV (PLWH) can achieve plasma viral load levels below the level of detection within 3 months of ART initiation [8]. Modern antiretrovirals are now very well tolerated by the majority of PLWH and many individuals can now expect to live near-normal life expectancies [9,10]. However, PLWH suffer from a burden of chronic diseases that exceeds the burden found in the general population. Leading comorbidities of PLWH on suppressive ART including cardiovascular disease, metabolic disorders, such as dyslipidemia in addition to malignancies, notably lung and colon cancers [3,4,11–13]. It is thought that low-level HIV replication (below that detected by conventional clinical viral load assays) and chronic systemic inflammation are contributing to the high prevalence of comorbidities [6,14].

The gut is a key player in HIV pathogenesis, as 60–80% of the body's CD4+ T cells are found in the gut, and there is cross-talk between intestinal microbiome, epithelial cells and immune cells [15–17]. During HIV infection, there is massive loss of CD4+ T cells leading to enteropathy and increased gut mucosal permeability, which results in translocation of microbial products from the gut into the periphery and consequently systemic immune activation [6,14,18] Microbial translocation results in stimulation of immune target cells and the release of pro-inflammatory cytokines and consequently systemic immune activation, lymphoid tissue fibrosis and T-cell and B-cell dysfunction [6,18,19]. Following ART initiation, HIV RNA significantly decreases and CD4+ T cells gradually recover, although in some persons this CD4+ T-cell recovery is incomplete. In contrast, recovery of CD4+ T cells in the gastrointestinal tract and lymphoid tissues lags behind the recovery in peripheral blood [15]. Immune activation decreases after ART but in most patients remains significantly increased compared with healthy controls [8]. Chronic immune activation and systemic inflammation lead, in turn, to non-AIDS morbidity and mortality in PLWH [6,18,19].

The endocannabinoid system and cannabinoids

The endocannabinoid system is constituted of endogenous cannabinoids (’endocannabinoids’), cannabinoid receptors, which induce or inhibit signal transduction pathways upon activation, in addition to enzymes, which synthesize or degrade cannabinoids (Table 1) [20–34]. CB1 and CB2 are G-protein-coupled receptors. CB1 receptors are expressed predominantly in the central nervous system but are also found in the lung, gastrointestinal tract, liver and kidneys [35,36]. Their presence in the central nervous system accounts for the psychoactive effects of cannabis [37,38]. High densities of CB1 receptors are located in the basal ganglia, substantia nigra, globus pallidus, cerebellum and hippocampus, but not in the brainstem where breathing is controlled, accounting for the low risk of overdose and toxicity [39]. Within the nervous system, CB1 regulates release of both excitatory and inhibitor neurotransmitters. As CB1 activation reduces excessive glutamate release, this prevents damage from overexcitation [40]. Within the gastrointestinal tract, CB1 activation on enteric neurons is the main mechanism by which cannabinoids affect gut motility, through inhibition of acetylcholine release which in turn reduces smooth muscle contractility and peristalsis [41,42]. In contrast, CB2 receptors are abundant on immune cells including T and B lymphocytes, natural killer cells, monocytes and neutrophils as well as the liver [35,37]. CB2 receptor activation induces apoptosis, inhibits pro-inflammatory cytokine expression, and induces a shift from a Th1 to Th2 immune response and induced myeloid-derived suppressor and T-regulatory cells [43].

T1-1
Table 1:
The endocannabinoid system.

There are many endogenous cannabinoids, or endocannabinoids, with the most studied endocannabinoids outlined in Table 1. Other endocannabinoids including N-palmitoyl ethanolamide (PEA) and oleoylethanolamine (OAE) have also been investigated [44]. Endocannabinoids exert many physiological effects similar to corticosteroids, such as reducing inflammation and inducing euphoria. CB1 receptors are the main targets for the endogenous cannabinoid, anandamide. Anandamide is attributed with inducing a sense of euphoria, as found in the ‘runner's high’ [35,36]. The other principal endocannabinoid is 2-arachidonoylglycerol (2-AG), which functions at both CB1 and CB2 receptors and is found in higher concentrations in the central nervous system than anadamide [35,36]. Neurons can produce endocannabinoids, but unlike most neurotransmitters they are released from cell bodies and not axons [45]. Furthermore, endocannabinoids can impede the release of neurotransmitters into the synapse [40].

The endocannabinoid system plays a role in regulation of a wide spectrum of processes including cognition, pain/sensation, mood, memory, locomotor activity, motivation and others [35,36]. Moreover, the endocannabinoid system is important for normal physiological functions of the gastrointestinal track, such as motility, hunger signaling, gut permeability and inflammation [25]. Of special relevance to PLWH, the endocannabinoid system has been shown to regulate epithelial barrier permeability through its interactions with the gut microbiota [25]. Activation of CB1 receptors in mice increased levels of lipopolysaccharide (LPS), the endotoxin produced by Gram-negative bacteria, via a pathway that decreases expression of tight junction proteins, occludin and zona occudens-1 [46]. Therefore, manipulation of the CB1 receptors receptor can affect gut permeability [25,46]. Furthermore, CB1 and CB2 receptor activation has been shown to ameliorate intestinal inflammation in multiple models of murine colitis [47].

With regards to varieties of cannabis plants, numerous chemovars or chemical varieties of cannabis plants exist [48]. The best described phytocannabinoids with medicinal properties include Δ9-tetrahydrocannabinol (Δ9-THC) and cannabidiol (CBD). Δ9-THC is a partial agonist at both CB1 and CB2 receptors and is usually the cause for the psychotropic effects of cannabis [37,38]. CBD is thought to mediate its effects through a variety of signalling mechanisms and act as a negative allosteric modulator at CB1[37,38]. CBD has the ability to enhance adenosine signalling (A2A receptor) through inhibition of uptake and provide a noncannabinoid receptor mechanism by which CBD can decrease inflammation [37,38]. An ‘entourage effect’ has been described, whereby a variety of metabolites thought to be inactive significantly increase the activity of the primary endogenous endocannabinoids, anandamide and 2-arachidonoylglycerol [24,49]. The entourage effect has been put forth as an explanation for the more robust performance of botanicals than their components in isolation [49,50]. Examples of phytocannabinoids which have exhibited therapeutic benefits include tetrahydrocannabivarin, cannabigerol and cannabichromene [51]. Terpenoids, which give cannabis plants their flavour and smell, also contribute to the entourage effect. Therefore, ‘minor cannabinoids’ other than Δ9-THC and CBD and cannabis terpenoids are likely to contribute to the overall medicinal effect of plants [49]. Several examples of cannabinoid synergy have previously been described [49]. Synthetic cannabinoids also exist, namely dronabinol (Marinol), which is most often used to control symptoms of chemotherapy-induced nausea and vomiting in cancer [49].

Cannabinoids and inflammation

A major underlying reason for the interest in cannabinoids as medicinal agents relate to their immunomodulatory properties (Table 2) [52–76]. As reviewed by Chiurchiu et al., endocannabinoids 2-arachidonyl glycerol (2-AG) and N-arachidonoylethanolamine or (AEA) have an effect on cells involved in both the innate and adaptive immune responses [33]. Early in-vitro studies demonstrated that Δ9-THC inhibits murine T-cell proliferation and macrophage propagation [77–80]. Stimulated cultured murine splenocytes exposed to varying concentrations of Δ9-THC demonstrated a dose-responsive suppressive effect of Δ9-THC on CD8+ T-cell proliferation and cytolytic activity [77–80]. Furthermore, Δ9-THC decreases T-cell proliferation by inhibition of interleukin (IL)-2 production and stimulation of cAMP immunosuppressive pathway [81,82]. Turning to cytokine-mediated polarization of T cells, several laboratories have shown that Δ9-THC polarizes the immune response towards a Th2 phenotype [83], facilitating immune responses against helminths and other extracellular parasites. Indeed, switch from Th1 to Th2 by Δ9-THC treatment is associated with histone modification signals to Th2 cytokine-related genes and suppressive modification signals to Th1 cytokine genes [84]. Δ9-THC can potently suppress the antitumor immune response, leading to enhanced tumor growth in vivo[85]. Nonnecrotic tumors and spleens were isolated from 3LL tumor-bearing C57BL/6 mice and evaluated for IL-10, transforming growth factor (TGF)-β, and interferon (IFN)-γ production in tumor homogenates and splenocyte culture supernatants. Tumor homogenates and splenocyte supernatants from Δ9-THC-treated mice contained significantly more IL-10 and TGF-β, but less IFN-γ than controls treated with diluent [85,86].

T2-1
Table 2:
Cannabinoid effects on human immune responses.

With regards to CBD, numerous studies have confirmed its anti-inflammatory effects [87,88]. In a murine model of LPS-induced acute lung injury, CBD was shown to reduce inflammation in acute lung injury [89,90]. It was also shown to reduce TNF-α and IFN-γ levels in murine collagen-induced arthritis [91]. In in-vitro cultured astrocytes, CBD diminished β-amyloid-induced neuroinflammation whereas in a mouse model of Alzheimer's disease, it reduced inducible nitric oxide synthase and IL-1β expression [92,93]. In murine models of experimental colitis, CBD reduced colon injury inducible nitric oxide synthase expression, interleukin-1β and interleukin-10 production [94]. When combined together, Δ9-THC and CBD display synergistic anti-inflammatory effects. In a mouse model of Alzheimer's disease, reduced astrogliosis, microgliosis and inflammatory-related molecules in treated AβPP/PS1 mice were more marked after treatment with Δ9-THC and CBD combined than with either Δ9-THC or CBD independently [95]. However, the interactions between cannabinoids are complex and have not been fully elucidated [96].

In human studies, discrepancies have been found compared to animal studies because of usage of much higher doses of cannabinoids in animal vs. human studies and extensive first-pass metabolism for both Δ9-THC and CBD following oral administration [97,98]. Zgair et al.[99,100] showed that oral co-administration of CBD and Δ9-THC with long-chain triglycerides (as would be contained in a high-fat meal) can augment intestinal lymphatic transport and markedly increase systemic bioavailability of cannabinoids in rats [97]. In addition, high concentrations of cannabinoids within the intestinal lymphatic system have previously been reported for compounds, which are absorbed following oral administration into systemic circulation primarily through the intestinal lymphatic system [97,99]. Consequently, Zgair et al.[97] orally administered CBD and Δ9-THC with lipids to rats and found that they could obtain concentrations sufficiently high in the intestinal lymphatic system to induce immunomodulatory effects. Interestingly, the distribution of CBD and Δ9-THC to lymphoid tissues in the intestinal lymphatic system and mesenteric lymph nodes was much higher than the distribution to the spleen, which is the largest lymphatic tissue [97]. Using human peripheral blood mononuclear cells (PBMCs), they showed that cells from multiple sclerosis patients were more susceptible to the immunosuppressive effects of cannabinoids [97]. These investigators also demonstrated that oral administration of CBD and Δ9-THC in humans with lipids can result in significant increases in intestinal lymphatic transport similar to what has been obtained for rats in this study [97,101].

Anti-inflammatory effects of cannabis in the context of HIV/simian immunodeficiency virus infection

Given that chronic HIV infection is associated with a state of persistent immune activation, the effects of cannabis in the context of HIV and SIV infection have been explored in vitro, as well as during animal and human studies (Table 3) [102–130].

T3-1
Table 3:
Role of cannabinoids in HIV infection.
T4-1
Table 3:
(Continued) Role of cannabinoids in HIV infection.

In-vitro studies assessing the impact of cannabinoids during HIV infection

CB1 and CB2 and HIV-1 co-receptors CCR5 and CXCR4 all signal via the Gαi-coupled pathways, sparking interest in exploring the effect of drugs targeting cannabinoid receptors on chemokine co-receptor function and HIV-1 infectivity [102]. In primary CD4+ T cells, agonism at CB2, but not CB1, decreased infection with CXCR4-tropic virus after cell-free and cell-to-cell viral transmission [102]. Furthermore, agonism at CB2 reduced F-actin levels, altering cytoskeleton architecture of resting CD4+ T cells [102]. In another study, change in CB2 expression in relation to HIV-1 infection was evaluated during the process of in-vitro monocyte differentiation into macrophage [103]. Gene and protein expression profiles of CB2 in primary human monocytes were studied. PBMCs from six healthy donors were used within 24 h or allowed to differentiate into macrophages for 7 days by adhesion and exposure to monocyte-granulocyte stimulating factor (M-GSF). Differentiated monocyte-derived macrophages (MDMs) were either examined uninfected or infected with macrophage tropic HIV-1ADA for 7 days. There was a step-wise increase in CB2 expression from monocytes to MDMs to HIV-1-infected MDMs [103]. The team also exposed the HIV-infected MDMs to CB2 receptor agonist JWH133 and found a dose-dependent decrease of reverse transcriptase activity [103]. Administration of the CB2 inverse-agonist/antagonist SR144528 had no significant effect on reverse transcriptase activity; however, when co-administered with JWH133, it counteracted agonist effects completely [103].

Given the proposed association between HIV-associated dementia and a reduced blood–brain barrier (BBB) function, the ability of cannabinoid agonists to restore integrity of human brain microvascular endothelial cells and the BBB following damage because of HIV-1 Gp120 has been examined [104]. Cannabinoid agonists inhibited HIV-1 Gp120-induced calcium influx mediated by substance P and greatly reduced permeability of human brain microvascular endothelial cells [104]. Cannabinoid agonists also prevented tight junction protein down-regulation of zona occludins protein-1, claudin-5, and junctional adhesion molecule-1 in human brain microvascular endothelial cells [104]. Furthermore, cannabinoid agonists inhibited the transmigration of human monocytes across the BBB and blocked the BBB permeability in vivo[104]. In another study involving CD4+ T cells and microglial cell cultures, WIN 55,212-2, a synthetic cannabinoid, was found to inhibit HIV-1 expression in a concentration-dependent and time-dependent manner [105]. This was in contrast to morphine, whereby HIV-1 expression was influenced by the timing of morphine exposure the time of drug exposure [105].

Animal model studies on the role of cannabinoids during HIV/simian immunodeficiency virus infection

Building on data from their in-vitro studies, Chen et al.[106] used a murine model to determine whether Δ9-THC produced similar effects in vivo as had been observed during in-vitro studies. A gp120-expressing plasmid, pVRCgp120, or a vector plasmid, pVRC2000, was injected intramuscularly into mice which were also dosed with Δ9-THC orally [106]. The gp120-specific IFN-γ and IL-2 responses were detected when splenocytes were re-stimulated with a gp120-derived peptide [106]. pVRCgp120 stimulation followed by peptide re-stimulation led to a rise in activation markers CD69+, CD80+, and major histocompatibility complex II [106]. Furthermore, Δ9-THC augmented the IFN-γ response and cellular activation C57Bl/6 wild type mice, but Δ9-THC either suppressed or did not have any effect on CB1 and CB2 knockout (CB1−/−CB2−/−) mice [106]. Therefore, Δ9-THC enhanced HIV antigen-specific immune responses, which occurred through CB1/CB2-dependent and independent mechanisms [106]. In another study, murine perinatal exposure to Δ9-THC on immune function was examined [107]. Murine fetal thymuses express high levels of CB1 and CB2 receptors [107]. Perinatal Δ9-THC exposure resulted in reduced thymic cellularity on gestational days 16, 17, and 18 and postgestational day 1, in addition to significant changes in T-cell subpopulations [107]. These changes were reversed by CB1 and CB2 antagonists [107]. Of note, Δ9-THC perinatal exposure resulted in impact on immune responses in murine life, with peripheral T cells from such mice having subdued proliferative responses to T-cell mitogens and lowered T-cell and antibody responses to HIV-1p17/p24/gp120 [107].

In order to examine the effect of chronic Δ9-THC administration on SIV disease progression, Molina et al.[108] administered Δ9-THC (0.32 mg/kg intramuscularly twice daily) to Indian-derived male rhesus macaques, beginning 28 days prior to infection with SIVmac251. They demonstrated that Δ9-THC administration decreased mortality because of SIV over 11 months and reduced plasma and cerebrospinal fluid SIV viral load [108]. Interestingly, a subsequent study by the same group did not find these protective effects in Indian-derived female rhesus macaques, suggesting a possible drug–sex steroid interaction and highlighting the need for further studies focusing on sex-specific cannabinoid effects on SIV disease [131]. It has been postulated that attenuation of localized tissue inflammatory responses and viral replication in male macaques could have accounted for the protective effects [108].

In another study, which involved Chinese rhesus male macaques, the effects of chronic Δ9-THC administration was examined in four groups of macaques (Δ9-THC+SIV+, Δ9-THC+SIV−, placebo/SIV+ and placebo/SIV− (n = 4/group) [109]. Twice daily Δ9-THC injections for 428 days did not change viral loads, CD4+ counts, CD4+/CD8+ ratios, T-cell proliferation, survival rate and clinical signs of disease progression over time compared with those macaques, which received placebo injections [109]. Moreover, regardless of Δ9-THC administration, all SIV-infected macaques displayed significant decline in CD4+/CD8+ T-cell ratios, loss of CD4+ T cells and increased levels of persistent Ki67+CD8+ T-cell proliferation vs. uninfected animals [109]. In addition, chronic Δ9-THC administration increased B-cell frequency, especially after 3 months, regardless of SIV infection status [109]. The main biological effect of Δ9-THC seen in this study was down-regulation of circulating IgE+ B cells in vivo, which the authors postulate could be mediated via a CB2-related mechanism [109].

The gut is known to harbor the majority of the body's total CD4+ T cells, which are HIV's primary target cells [15–17]. In another study by Kumar et al. involving Indian-origin rhesus macaques, animals were treated for 17 months with Δ9-THC (0.18–0.25 mg/kg intramuscularly twice daily) vs. vehicle administration for 17 months [110]. Duodenal tissues were examined from the macaques about 5 months after infection with SIVmac251. There were lower SIV RNA viral loads in plasma, PBMCs and duodenal tissue in the Δ9-THC-treated vs. vehicle-treated macaques, though differences between HIV proviral DNA between groups did not statistically differ [110]. Elevated duodenal integrin β7+CD4+ and CD8+ central memory T cells and preferential Th2 cytokine levels were also noted between Δ9-THC-treated vs. control groups [110]. Gene array analysis identified six genes important for apoptosis, cell survival, proliferation and morphogenesis and energy and substrate metabolic processesthat were increased in the Δ9-THC-treated vs. control groups [110]. Immunohistochemical staining showed attenuated apoptosis in epithelial crypt cells of THC-treated macaques [110]. The same group also investigated cannabinoids including Δ9-THC on microRNA (miRNA) expression in the gut. Compared with Indian-origin rhesus macaques administered vehicle/SIV, Δ9-THC administration to SIV-infected animals resulted in the selective upregulation of a cluster of the six miRNAs previously shown to have anti-inflammatory effects [111]. They characterized the functional relevance of miR-99b and found its expression increased in SIV-infected macaques [111]. The same group then studied miRNA expression sequentially in intestinal lamia propria leukocytes of eight Indian origin rhesus macaques before and at 21, 90 and 180 days postinoculation with SIV [112]. They demonstrated miR-150 downregulation during T-cell activation, disrupting IL-1 receptor kinase 1 translational control, fostering persistent gastrointestinal inflammation [112].

As epithelial barrier function is epigenetically regulated by inflammation-induced microRNAs [132], Kumar et al.[113] examined the epigenetic regulation of miRNAs when colonic tissue from SIV-infected Indian-origin rhesus macaques was treated ex vivo with Δ9-THC. They found that dysregulated miR-130a and miR-212 expression in colonic epithelium increased epithelial barrier disturbance via modulation of peroxisome proliferator-activated receptor gamma (PPARγ) and occludin expression [113]. Δ9-THC treatment of colonic tissue was able to recover transepithelial electrical resistance (TEER) levels to that observed in control's miRNA mimics [113]. PPARγ mRNA expression was increased when colonic tissue was treated with Δ9-THC, showcasing the therapeutic potential of cannabinoids to restore gut epithelial barrier integrity [113]. Yet another study demonstrated that 10 miRNAs were significantly upregulated in Δ9-THC-treated SIV-infected macaques vs. vehicle-treated SIV-infected macaques. Among these, miR-204 was confirmed to directly target matrix metalloproteinase (MMP)-8, an extracellular matrix-degrading collagenase that was significantly downregulated in Δ9-THC-treated SIV rhesus macaques [114]. Moreover, Δ9-THC-treated SIV rhesus macaques failed to upregulate pro-inflammatory miR-21, miR-141 and miR-222 and alpha/beta defensins, suggesting reduced intestinal inflammation [114]. Δ9-THC-treated SIV rhesus macaques also had increased expression of tight junction proteins (occludin, claudin-3), anti-inflammatory MUC13, keratin 8 (stress protection), prominin 1 (PROM1) (epithelial proliferation) and anti-HIV CCL5 [114]. There was significant collagen deposition in the paracortex and B-cell follicular lymph nodes from all vehicle-treated SIV-infected but not in the THC-treated, SIV-infected macaques, underscoring the ability of Δ9-THC to prevent lymph node fibrosis [114]. This finding is noteworthy, given that lymph node fibrosis is an irreversible consequence of HIV-induced chronic inflammation [114]. Furthermore, Δ9-THC suppressed gut T-cell proliferation and activation as demonstrated by Ki67, HLA-DR and PD-1 expression, and increased percentages of anti-inflammatory CD163+ macrophages [114]. Although Δ9-THC did not affect levels of CD4+ T cells, it significantly reduced levels of CD8+ T cells in peripheral blood at 14 and 150 days post-SIV infection [114]. Taken together, these findings support a role for selective miRNA and gene induction, and reduction in T-cell activation, in Δ9-THC-mediated suppression of intestinal inflammation in SIV [114].

The SIV/HIV reservoir is of importance because of its implications for HIV cure. Molina et al.[133] examined the effect of Δ9-THC treatment on the SIV reservoir within macaque spleens. Two long terminal repeat (LTR) DNA circles are markers of recent viral integration and provide a measure of viral DNA entry into the nucleus after integration of proviral DNA into host cell chromosomes. They demonstrated that episomal SIV 2-long terminal repeat (2-LTR) DNA circles were much lower in Δ9-THC-treated SIV-infected macaques compared with SIV-infected macaques not treated with Δ9-THC [133]. These findings suggest that Δ9-THC treatment may reduce SIV replication within tissue reservoirs, such as the spleen [133] -- a tissue very difficult to access in humans because of the risk of haemorrhage. In a study currently being led by Dr Nichole Klatt, her group is testing the hypothesis that cannabinoid administration to chronic SIV-infected nonhuman primates will decrease inflammation and the SIV reservoir [134]. Furthermore, they are determining cannabinoid levels and kinetics in the blood vs. gastrointestinal tract [134].

Human studies on cannabinoids and HIV infection: from observational studies to randomized controlled trials

Observational studies

Moving from simians to humans, variable results have been obtained from cross-sectional observational studies examining the association between CD4+ and CD8+ T-cell count and cannabis use [1,115–119]. In a longitudinal study involving PLWH and hepatitis C co-infection, cannabis use was not associated with reduction in CD4+ T-cell count [120]. No association has been found between cannabis and progression of HIV disease in the pre-ART era [122]. In the ART era, cannabis use has not been associated with 5-year mortality risk among over 3000 veterans followed between 2002 and 2010 [123]. Furthermore, cannabis smoking did not accelerate progression of liver disease in a cohort of PLWH and hepatitis C co-infection followed longitudinally [135].

In a cross-sectional observational study, which performed detailed immunological analyses, Manuzak et al.[124] examined the impact of cannabis use on peripheral immune cell frequency, activation and function in an observational study including 198 HIV-infected ART-treated PLWH. The overall frequency of CD4+ and CD8+ T cells among PBMCs was not altered in moderate or heavy cannabis-using individuals, compared with noncannabis-using individuals. However, the frequency of activated (HLA-DR+CD38+) T cells was significantly lower in the heavy cannabis-using group compared with the noncannabis-using individuals for both CD4+ and CD8+ T cells [124]. They also examined the frequency of intermediate monocytes (CD14+CD16+), which contribute to end-organ complications in PLWH because of their secretion of pro-inflammatory cytokines [136,137]. Intermediate monocyte frequency was significantly lower in the heavy cannabis-using group compared with noncannabis users. Finally, the frequency of nonclassical monocytes (CD14+CD16+), which also secrete pro-inflammatory cytokines and promote T-cell activation [136,137], was significantly reduced in the moderate and heavy cannabis-using group compared with the frequencies among noncannabis users [124]. Following in-vitro stimulation, frequencies of IL-10, IL-23, TNF-α-producing antigen-presenting cells were also much lower in heavy cannabis-using individuals vs. noncannabis using individuals [124]. High frequencies of CD16+ monocytes in circulation are of interest as they are elevated in individuals suffering from HIV-associated dementia [125,138]. Furthermore, in the central nervous system, specimens from patients with HIV-associated neurocognitive disorders obtained postmortem, high levels of CD16+ monocytes staining positive for HIV viral capsid protein p24 have been described [138]. In another observational study, Rizzo et al.[125] also examined frequencies of CD16+ monocytes between HIV-infected individuals using cannabis and HIV-infected donors not using cannabis. They observed lower numbers of CD16+CD163+ monocytes in HIV-infected persons using cannabis compared with donors when compared with HIV-infected persons not using cannabis donors [125]. In addition, plasma inducible protein 10 was also significantly lower in HIV-infected cannabis users than noncannabis users. Δ9-THC significantly decreased the percentage of monocytes expressing the CD16+, CD163+ and CD16+/CD163+ whereas CBD at the same concentration elicited no significant effects on the percentage of monocytes expressing CD16+, CD163+ and CD16+/CD163+[125]. These findings are relevant given that co-expression of CD16+ and CD163+ on monocytes is increased in postmortem brain tissue of persons with cognitive impairment [139].

In another cross-sectional study, Vidot et al.[126] examined markers of gut microbial translocation in addition to immune activation in ART-treated PLWH who use methamphetamines, which have been associated within greater monocyte activation and inflammation [100,140]. However, hazardous cannabis users (as determined by a score of eight or more on the Cannabis Use Disorder Identification Test-Revised (CUDIT-R) [141] had increased soluble CD14+ (sCD14+) compared with noncannabis users. Furthermore, there was no association between cannabis use severity with intestinal barrier integrity protein (iFABP), a marker of gastrointestinal enterocyte cell death [126]. Therefore, hazardous cannabis use in ART-treated methamphetamine users is unlikely to counteract the markers of gut mucosal damages [126]. However, because of inherent differences between populations, these results should not be generalized to individuals using cannabinoids for medicinal or recreational reasons in the absence of harzardous cannabis or methamphetamine use.

Given the importance of seminal fluid for HIV transmission, one group examined the association between cannabis use and seminal plasma viral load. In HIV-infected MSM on ART for at least 6 months, Ghosn et al.[127] measured HIV-RNA in seminal plasma and in blood plasma from two paired samples obtained 4 weeks apart. Out of 157 participants, 33 (21%) of individuals had STIs and residual blood plasma viral load. Multivariate adjustments were performed and PMBC-associated HIV-DNA and cannabis use during sexual intercourse were the only factors associated with significantly with seminal plasma viral load [OR 2.9 (1.2; 7.3)] [127]. However, because of the observational nature of this study, the high prevalence of STIs (which increase the seminal plasma viral load [142]) and the tendency of many individuals to use cannabis in combination with other substances, no causal relationship can be implied [127]. Indeed, cannabis use is more likely a by-proxy for factors contributing to increase seminal plasma viral load [127]. Furthermore, based on the evidence available to date, there is no biological basis to suggest that cannabis use induces inflammation in the genital tract, therefore more study is required before conclusions can be drawn.

In the aforementioned NIH-funded, ongoing study currently being led by Dr Nichole Klatt, her group will also test the hypothesis that cannabis use in the context of chronic HIV infection may decrease inflammation and the HIV reservoir in humans [134]. They will measure markers of inflammation, immunity, and the HIV reservoir from both blood and gastrointestinal tissues of PLWH reporting using cannabis daily compared with those reporting no drug use [134]. They will also assess ex vivo the mechanisms of cannabinoid anti-inflammatory activity through the use of cannabinoid receptor agonists in co-cultures [134].

Randomized-controlled trials in people living with HIV with primary endpoints outside of immunology

Moving onto randomized-controlled trials (RCTs) of cannabis in PLWH, two previous studies were performed to examine the effects of cannabinoids on peripheral neuropathy. Abrams et al.[143] randomly assigned 50 PLWH to smoke either cannabis (3.56% Δ9-THC) or identical placebo cigarettes, with the cannabinoids extracted three times daily for 5 days. Although cannabis was effective to reduce chronic HIV-associated neuropathic pain, effect on immunological parameters were not examined [143]. In another study examining neuropathic pain in PLWH, Coates et al. performed a placebo controlled double-blind randomized crossover trial of 1–8% Δ9-THC and placebo cigarettes administered four times daily for 5 days [144]. Although cannabis improved neuropathic pain vs. placebo, there were no significant effects on viral load or CD4+ T-cell counts reported [144]. The authors of a systematic review on the use of cannabis for reducing morbidity and mortality in HIV concluded that evidence for substantial effects on morbidity and mortality is currently limited and evidence for safety and efficacy of cannabis is lacking. Studies have been of short duration, in small numbers of patients and have focused on short-term measures of efficacy [145]. Furthermore, no study examined effect on inflammatory markers or HIV reservoir size.

Clinical trials in the context of HIV immunology and virology

With the goal of describing the short-term effects of cannabinoids on PLWH, 67 individuals who had been on ART for at least 8 weeks prior to the study were enrolled and randomized to either 3.95% of Δ9-THC cannabis cigarettes, 2.5 mg of dronabinol-containing capsule or placebo capsules three times daily prior to meals. In this study published in 2003, at baseline HIV-RNA level was under 50 copies/ml for 36 participants. Nearly half of participants were on either nelfinavir (55%) or indinavir (45%). This was notable because of concern for a potential drug interaction via hepatic cytochrome P-450 enzyme systems. However, neither smoked nor oral cannabinoids significantly changed HIV RNA levels, CD4+ and CD8+ T cells or protease inhibitor levels over a 21-day course of treatment [129]. Furthermore, other T-cell subpopulations, B cells, natural killer (NK) cells were not significantly altered. In addition, immune cell function appeared preserved based on assays for induced cytokine production, NK cell function and lymphocyte proliferation, as performed at baseline and weekly thereafter [130]. Bredt et al.[130] concluded that the few changes, which were statistically significant did not constitute any meaningful pattern of changes in immune phenotype or function.

In a phase 1 trial supported by the Canadian Institutes of Health Research (CIHR) Canadian HIV Trials Network (CTN-PT028), our group plans to examine the safety and tolerability of cannabinoid capsules with capsules of Δ9-THC:CBD in a 1 : 1 ratio or capsules of Δ9-THC:CBD in a 1 : 9 ratio when taken by PLWH with fully suppressed viral load for at least 3 years [146]. Questionnaires assessing quality of life and mood will be administered and effect on inflammatory markers and markers of peripheral blood HIV reservoir size will be assessed before and after study completion [146]. As this is a pilot study, a goal is to identify which steps in the protocol run smoothly vs. which steps require modification when designing a larger clinical trial powered to answer specific outcome questions related to human immunology and the HIV reservoir [146]. Studies examining the effects of cannabis in the context of HIV/SIV infection are summarized in Table 3.

Challenges of performing randomized-controlled trials with cannabinoid-based medicines

Due to confounding from observational studies, which limit assessment of cause and effect relationships, randomized, double-blinded, placebo-controlled clinical trials are the ideal method to study the effect of a drug on immune function. However, because of the psychological effects of Δ9-THC, it may be difficult if not impossible to blind participants and staff to treatment group allocation by having a placebo group. There are also concerns that giving participants agents, which could impede their cognitive function could be problematic for persons in certain types of jobs or while driving [147]. There is ongoing stigma related to cannabis and CBMs within and outside the medical community. Moreover, regulatory issues, with long time delays before receiving all the approvals necessary to execute a clinical trials, remain another impediment to conducting RCTs of CBMs.

The legalization of recreational cannabis in some US states and in Canada on 17 October 2018 (under The Cannabis Act) has brought along with it an increased interest by both the clinicians, researchers, public health authorities, government and the general public. Since the legalization of recreational cannabis by the Canadian Federal government, there has been a rise in funding opportunities available to better understand the health effects of cannabis, with millions of dollars in federal funding for cannabis research projects across the country being recently announced [148]. However, despite the legalization of recreational cannabis, performing clinical trials in Canada remains challenging because of lengthy regulatory hurdles.

Conclusion

Although the reasons for cannabis use have changed since the beginning of the HIV epidemic, for PLWH adherent to ART prevention and management of comorbidities is the paramount concern. Gut microbial translocation results in chronic systemic inflammation, driving these comorbidities. In-vitro studies show that CB2 activation of CD4+ T cells impairs productive HIV infection and CB2 agonists can reduce HIV reverse transcriptase activity. Simian studies have shown beneficial effects of Δ9-THC on reducing morbidity and mortality in SIV-infected macaques, and have demonstrated the ability of cannabinoids to restore gut--epithelial barrier integrity. In humans, the majority of cross-sectional studies neither demonstrate any harmful effect of cannabis or CBM on peripheral blood CD4+ or CD8+ T-cell count nor on HIV viral load. Cross-sectional studies in PLWH have also demonstrated favourable associations between cannabis use and a reduction in intermediate and nonclassical monocytes, which are harmful because of their ability to secrete pro-inflammatory cytokines and promote T-cell activation. Despite regulatory hurdles, further exploration of the potential benefits of cannabis and CBM on the immune system of PLWH in the context of randomized clinical trials will be helpful to elucidate the therapeutic potential of these agents in the management of chronic HIV. Ultimately, cannabis and CBM may play a role in slowing the development of HIV-associated morbidity and mortality, as an adjunct to ART, if their immunomodulatory properties can properly be exploited.

Acknowledgements

Funding: C.T.C. is supported by a Fonds de recherche du Québec-Santé (FRQ-S) clinician-researcher Junior 1 salary award. M.A.J. holds the CIHR Canada Research Chair tier 2 in Immuno-Virology. The CTN PT028 study is supported by the CIHR Canadian HIV Trials Network.

The authors have received free product from Tilray Inc. for use in a clinical trial (Canadian HIV Trials Network PT028), but no financial support was received for this article or other studies.

Conflicts of interest

There are no conflicts of interest.

References

1. Furler MD, Einarson TR, Millson M, Walmsley S, Bendayan R. Medicinal and recreational marijuana use by patients infected with HIV. AIDS Patient Care STDs 2004; 18:215–228.
2. Belle-Isle L, Hathaway A. Barriers to access to medical cannabis for Canadians living with HIV/AIDS. AIDS Care 2007; 19:500–506.
3. Armah KA, McGinnis K, Baker J, Gibert C, Butt AA, Bryant KJ, et al. HIV status, burden of comorbid disease, and biomarkers of inflammation, altered coagulation, and monocyte activation. Clin Infect Dis 2012; 55:126–136.
4. Aberg JA. Aging, inflammation, and HIV infection. Top Antivir Med 2012; 20:101–105.
5. Hsue PY. Mechanisms of cardiovascular disease in the setting of HIV infection. Can J Cardiol 2019; 35:238–248.
6. Deeks SG, Tracy R, Douek DC. Systemic effects of inflammation on health during chronic HIV infection. Immunity 2013; 39:633–645.
7. Harris GE, Dupuis L, Mugford GJ, Johnston L, Haase D, Page G, et al. Patterns and correlates of cannabis use among individuals with HIV/AIDS in Maritime Canada. Can J Infect Dis Med Microbiol 2014; 25:e1–e7.
8. Maartens G, Celum C, Lewin SR. HIV infection: epidemiology, pathogenesis, treatment, and prevention. Lancet 2014; 384:258–271.
9. Antiretroviral Therapy Cohort Collaboration. Life expectancy of individuals on combination antiretroviral therapy in high-income countries: a collaborative analysis of 14 cohort studies. Lancet 2008; 372:293–299.
10. Antiretroviral Therapy Cohort Collaboration. Survival of HIV-positive patients starting antiretroviral therapy between 1996 and 2013: a collaborative analysis of cohort studies. Lancet HIV 2017; 4:e349–e356.
11. Hileman CO, Funderburg NT. Inflammation, immune activation, and antiretroviral therapy in HIV. Curr HIV/AIDS Rep 2017; 14:93–100.
12. Funderburg NT, Mayne E, Sieg SF, Asaad R, Jiang W, Kalinowska M, et al. Increased tissue factor expression on circulating monocytes in chronic HIV infection: relationship to in vivo coagulation and immune activation. Blood 2010; 115:161–167.
13. Worm SW, Bower M, Reiss P, Bonnet F, Law M, Fatkenheuer G, et al. Non-AIDS defining cancers in the D:A:D Study–time trends and predictors of survival: a cohort study. BMC Infect Dis 2013; 13:471.
14. Deeks SG, Phillips AN. HIV infection, antiretroviral treatment, ageing, and non-AIDS related morbidity. BMJ 2009; 338:a3172.
15. Costiniuk CT, Angel JB. Human immunodeficiency virus and the gastrointestinal immune system: does highly active antiretroviral therapy restore gut immunity?. Mucosal Immunol 2012; 5:596–604.
16. Dandekar S. Pathogenesis of HIV in the gastrointestinal tract. Curr HIV/AIDS Rep 2007; 4:10–15.
17. Guadalupe M, Reay E, Sankaran S, Prindiville T, Flamm J, McNeil A, et al. Severe CD4+ T-cell depletion in gut lymphoid tissue during primary human immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J Virol 2003; 77:11708–11717.
18. Deeks SG. Immune dysfunction, inflammation, and accelerated aging in patients on antiretroviral therapy. Top HIV Med 2009; 17:118–123.
19. Brenchley JM, Price DA, Schacker TW, Asher TE, Silvestri G, Rao S, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med 2006; 12:1365–1371.
20. Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE, Schatz AR, et al. Identification of an endogenous 2-monoglyceride, present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol 1995; 50:83–90.
21. Khanolkar AD, Abadji V, Lin S, Hill WA, Taha G, Abouzid K, et al. Head group analogs of arachidonylethanolamide, the endogenous cannabinoid ligand. J Med Chem 1996; 39:4515–4519.
22. Pacher P, Batkai S, Kunos G. The endocannabinoid system as an emerging target of pharmacotherapy. Pharmacol Rev 2006; 58:389–462.
23. Lee Y, Jo J, Chung HY, Pothoulakis C, Im E. Endocannabinoids in the gastrointestinal tract. Am J Physiol Gastrointest Liver Physiol 2016; 311:G655–G666.
24. Ben-Shabat S, Fride E, Sheskin T, Tamiri T, Rhee MH, Vogel Z, et al. An entourage effect: inactive endogenous fatty acid glycerol esters enhance 2-arachidonoyl-glycerol cannabinoid activity. Eur J Pharmacol 1998; 353:23–31.
25. DiPatrizio NV. Endocannabinoids in the gut. Cannabis Cannabinoid Res 2016; 1:67–77.
26. Hanus L, Abu-Lafi S, Fride E, Breuer A, Vogel Z, Shalev DE, et al. 2-arachidonyl glyceryl ether, an endogenous agonist of the cannabinoid CB1 receptor. Proc Natl Acad Sci USA 2001; 98:3662–3665.
27. Herkenham M, Lynn AB, Little MD, Johnson MR, Melvin LS, de Costa BR, et al. Cannabinoid receptor localization in brain. Proc Natl Acad Sci USA 1990; 87:1932–1936.
28. Battista N, Di Tommaso M, Bari M, Maccarrone M. The endocannabinoid system: an overview. Front Behav Neurosci 2012; 6:9.
29. Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature 1993; 365:61–65.
30. Van Sickle MD, Duncan M, Kingsley PJ, Mouihate A, Urbani P, Mackie K, et al. Identification and functional characterization of brainstem cannabinoid CB2 receptors. Science 2005; 310:329–332.
31. Simon V, Cota D. Mechanisms in endocrinology: endocannabinoids and metabolism: past, present and future. Eur J Endocrinol 2017; 176:R309–R324.
32. Basavarajappa BS. Critical enzymes involved in endocannabinoid metabolism. Protein Pept Lett 2007; 14:237–246.
33. Chiurchiu V, Battistini L, Maccarrone M. Endocannabinoid signalling in innate and adaptive immunity. Immunology 2015; 144:352–364.
34. Specter S, Lancz G, Goodfellow D. Suppression of human macrophage function in vitro by delta 9-tetrahydrocannabinol. J Leukoc Biol 1991; 50:423–426.
35. Yao B, Mackie K. Endocannabinoid receptor pharmacology. Curr Top Behav Neurosci 2009; 1:37–63.
36. Chanda D, Neumann D, Glatz JFC. The endocannabinoid system: overview of an emerging multifaceted therapeutic target. Prostaglandins Leukot Essent Fatty Acids 2019; 140:51–56.
37. Tahamtan A, Tavakoli-Yaraki M, Rygiel TP, Mokhtari-Azad T, Salimi V. Effects of cannabinoids and their receptors on viral infections. J Med Virol 2016; 88:1–12.
38. Smith TH, Sim-Selley LJ, Selley DE. Cannabinoid CB1 receptor-interacting proteins: novel targets for central nervous system drug discovery?. Br J Pharmacol 2010; 160:454–466.
39. Herkenham M, Lynn AB, de Costa BR, Richfield EK. Neuronal localization of cannabinoid receptors in the basal ganglia of the rat. Brain Res 1991; 547:267–274.
40. Morena M, Patel S, Bains JS, Hill MN. Neurobiological interactions between stress and the endocannabinoid system. Neuropsychopharmacology 2016; 41:80–102.
41. Hasenoehrl C, Taschler U, Storr M, Schicho R. The gastrointestinal tract - a central organ of cannabinoid signaling in health and disease. Neurogastroenterol Motil 2016; 28:1765–1780.
42. Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: delta9-tetrahydrocannabinol, cannabidiol and delta9-tetrahydrocannabivarin. Br J Pharmacol 2008; 153:199–215.
43. Nagarkatti P, Pandey R, Rieder SA, Hegde VL, Nagarkatti M. Cannabinoids as novel anti-inflammatory drugs. Future Med Chem 2009; 1:1333–1349.
44. Keereetaweep J, Chapman KD. Lipidomic analysis of endocannabinoid signaling: targeted metabolite identification and quantification. Neural Plast 2016; 2016:2426398.
45. Di Marzo V, Fontana A, Cadas H, Schinelli S, Cimino G, Schwartz JC, Piomelli D. Formation and inactivation of endogenous cannabinoid anandamide in central neurons. Nature 1994; 372:686–691.
46. Muccioli GG, Naslain D, Backhed F, Reigstad CS, Lambert DM, Delzenne NM, Cani PD. The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst Biol 2010; 6:392.
47. Gyires K, Zadori ZS. Role of cannabinoids in gastrointestinal mucosal defense and inflammation. Curr Neuropharmacol 2016; 14:935–951.
48. Lewis MA, Russo EB, Smith KM. Pharmacological foundations of cannabis chemovars. Planta Med 2018; 84:225–233.
49. Russo EB. The Case for the entourage effect and conventional breeding of clinical cannabis: No “Strain,” No Gain’. Front Plant Sci 1969; 9: doi: 10.3389/fpls.2018.01969.
50. Mechoulam R, Ben-Shabat S. From gan-zi-gun-nu to anandamide and 2-arachidonoylglycerol: the ongoing story of cannabis. Nat Prod Rep 1999; 16:131–143.
51. Russo EB. Taming THC: potential cannabis synergy and phytocannabinoid-terpenoid entourage effects. Br J Pharmacol 2011; 163:1344–1364.
52. Wu HY, Chang AC, Wang CC, Kuo FH, Lee CY, Liu DZ, Jan TR. Cannabidiol induced a contrasting pro-apoptotic effect between freshly isolated and precultured human monocytes. Toxicol Appl Pharmacol 2010; 246:141–147.
53. Kishimoto S, Gokoh M, Oka S, Muramatsu M, Kajiwara T, Waku K, et al. 2-arachidonoylglycerol induces the migration of HL-60 cells differentiated into macrophage-like cells and human peripheral blood monocytes through the cannabinoid CB2 receptor-dependent mechanism. J Biol Chem 2003; 278:24469–24475.
54. 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 into macrophage-like cells and human peripheral blood monocytes. FEBS Lett 2005; 579:6473–6478.
55. Kishimoto S, Kobayashi Y, Oka S, Gokoh M, Waku K, Sugiura T. 2-Arachidonoylglycerol, an endogenous cannabinoid receptor ligand, induces accelerated production of chemokines in HL-60 cells. J Biochem 2004; 135:517–524.
56. Gokoh M, Kishimoto S, Oka S, Sugiura T. 2-Arachidonoylglycerol enhances the phagocytosis of opsonized zymosan by HL-60 cells differentiated into macrophage-like cells. Biol Pharm Bull 2007; 30:1199–1205.
57. Stefano GB, Bilfinger TV, Rialas CM, Deutsch DG. 2-arachidonyl-glycerol stimulates nitric oxide release from human immune and vascular tissues and invertebrate immunocytes by cannabinoid receptor 1. Pharmacol Res 2000; 42:317–322.
58. Matias I, Pochard P, Orlando P, Salzet M, Pestel J, Di Marzo V. Presence and regulation of the endocannabinoid system in human dendritic cells. Eur J Biochem 2002; 269:3771–3778.
59. Roth MD, Castaneda JT, Kiertscher SM. Exposure to delta9-tetrahydrocannabinol impairs the differentiation of human monocyte-derived dendritic cells and their capacity for T cell activation. J Neuroimmune Pharmacol 2015; 10:333–343.
60. Chiurchiu V, Cencioni MT, Bisicchia E, De Bardi M, Gasperini C, Borsellino G, et al. Distinct modulation of human myeloid and plasmacytoid dendritic cells by anandamide in multiple sclerosis. Ann Neurol 2013; 73:626–636.
61. Srivastava MD, Srivastava BI, Brouhard B. Delta9 tetrahydrocannabinol and cannabidiol alter cytokine production by human immune cells. Immunopharmacology 1998; 40:179–185.
62. Specter S, Rivenbark M, Newton C, Kawakami Y, Lancz G. Prevention and reversal of delta-9-tetrahydrocannabinol induced depression of natural killer cell activity by interleukin-2. Int J Immunopharmacol 1989; 11:63–69.
63. Sugiura T, Oka S, Gokoh M, Kishimoto S, Waku K. New perspectives in the studies on endocannabinoid and cannabis: 2-arachidonoylglycerol as a possible novel mediator of inflammation. J Pharmacol Sci 2004; 96:367–375.
64. Kishimoto S, Muramatsu M, Gokoh M, Oka S, Waku K, Sugiura T. Endogenous cannabinoid receptor ligand induces the migration of human natural killer cells. J Biochem 2005; 137:217–223.
65. Trisler K, Specter S. Delta-9-tetrahydrocannabinol treatment results in a suppression of interleukin-2-induced cellular activities in human and murine lymphocytes. Int J Immunopharmacol 1994; 16:593–603.
66. Herrera B, Carracedo A, Diez-Zaera M, Gomez del Pulgar T, Guzman M, Velasco G. The CB2 cannabinoid receptor signals apoptosis via ceramide-dependent activation of the mitochondrial intrinsic pathway. Exp Cell Res 2006; 312:2121–2131.
67. Jia W, Hegde VL, Singh NP, Sisco D, Grant S, Nagarkatti M, et al. Delta9-tetrahydrocannabinol-induced apoptosis in Jurkat leukemia T cells is regulated by translocation of Bad to mitochondria. Mol Cancer Res 2006; 4:549–562.
68. Ngaotepprutaram T, Kaplan BL, Kaminski NE. Impaired NFAT and NFkappaB activation are involved in suppression of CD40 ligand expression by Delta(9)-tetrahydrocannabinol in human CD4(+) T cells. Toxicol Appl Pharmacol 2013; 273:209–218.
69. Schwarz H, Blanco FJ, Lotz M. Anadamide, an endogenous cannabinoid receptor agonist inhibits lymphocyte proliferation and induces apoptosis. J Neuroimmunol 1994; 55:107–115.
70. Sancho R, Calzado MA, Di Marzo V, Appendino G, Munoz E. Anandamide inhibits nuclear factor-kappaB activation through a cannabinoid receptor-independent pathway. Mol Pharmacol 2003; 63:429–438.
71. Rockwell CE, Raman P, Kaplan BL, Kaminski NE. A COX-2 metabolite of the endogenous cannabinoid, 2-arachidonyl glycerol, mediates suppression of IL-2 secretion in activated Jurkat T cells. Biochem Pharmacol 2008; 76:353–361.
72. Joseph J, Niggemann B, Zaenker KS, Entschladen F. Anandamide is an endogenous inhibitor for the migration of tumor cells and T lymphocytes. Cancer Immunol Immunother 2004; 53:723–728.
73. Malfitano AM, Matarese G, Pisanti S, Grimaldi C, Laezza C, Bisogno T, et al. Arvanil inhibits T lymphocyte activation and ameliorates autoimmune encephalomyelitis. J Neuroimmunol 2006; 171:110–119.
74. Rockwell CE, Snider NT, Thompson JT, Vanden Heuvel JP, Kaminski NE. Interleukin-2 suppression by 2-arachidonyl glycerol is mediated through peroxisome proliferator-activated receptor gamma independently of cannabinoid receptors 1 and 2. Mol Pharmacol 2006; 70:101–111.
75. Derocq JM, Segui M, Marchand J, Le Fur G, Casellas P. Cannabinoids enhance human B-cell growth at low nanomolar concentrations. FEBS Lett 1995; 369:177–182.
76. Cencioni MT, Chiurchiu V, Catanzaro G, Borsellino G, Bernardi G, Battistini L, Maccarrone M. Anandamide suppresses proliferation and cytokine release from primary human T-lymphocytes mainly via CB2 receptors. PloS One 2010; 5:e8688.
77. Pross SH, Klein TW, Newton CA, Smith J, Widen R, Friedman H. Differential suppression of T-cell subpopulations by thc (delta-9-tetrahydrocannabinol). Int J Immunopharmacol 1990; 12:539–544.
78. Pross S, Newton C, Klein T, Widen R, Smith J, Friedman H. Suppression of T lymphocyte subpopulations by THC. Adv Exp Med Biol 1991; 288:113–117.
79. Levy JA, Heppner GH. Alterations of immune reactivity by haloperidol and delta-9-tetrahydrocannabinol. J Immunopharmacol 1981; 3:93–109.
80. Pross SH, Klein TW, Newton C, Smith J, Widen R, Friedman H. Age-related suppression of murine lymphoid cell blastogenesis by marijuana components. Dev Comp Immunol 1990; 14:131–137.
81. Massi P, Sacerdote P, Ponti W, Fuzio D, Manfredi B, Vigano D, et al. Immune function alterations in mice tolerant to delta9-tetrahydrocannabinol: functional and biochemical parameters. J Neuroimmunol 1998; 92:60–66.
82. Borner C, Smida M, Höllt V, Schraven B, Kraus J. Cannabinoid receptor type 1- and 2-mediated increase in cyclic AMP inhibits T cell receptor-triggered signaling. J Biol Chem 2009; 284:35450–35460.
83. Eisenstein TK, Meissler JJ. Effects of cannabinoids on T-cell function and resistance to infection. J Neuroimmune Pharmacol 2015; 10:204–216.
84. Yang X, Hegde VL, Rao R, Zhang J, Nagarkatti PS, Nagarkatti M. Histone modifications are associated with Δ9-tetrahydrocannabinol-mediated alterations in antigen-specific T cell responses. J Biol Chem 2014; 289:18707–18718.
85. Zhu LX, Sharma S, Stolina M, Gardner B, Roth MD, Tashkin DP, et al. Delta-9-tetrahydrocannabinol inhibits antitumor immunity by a CB2 receptor-mediated, cytokine-dependent pathway. J Immunol 2000; 165:373–380.
86. Gardner B, Zu LX, Sharma S, Liu Q, Makriyannis A, Tashkin DP, Dubinett SM. Autocrine and paracrine regulation of lymphocyte CB2 receptor expression by TGF-beta. Biochem Biophys Res Commun 2002; 290:91–96.
87. Burstein SH, Zurier RB. Cannabinoids, endocannabinoids, and related analogs in inflammation. AAPS J 2009; 11:109–119.
88. Burstein S. Cannabidiol (CBD) and its analogs: a review of their effects on inflammation. Bioorg Med Chem 2015; 23:1377–1385.
89. Ribeiro A, Ferraz-de-Paula V, Pinheiro ML, Vitoretti LB, Mariano-Souza DP, Quinteiro-Filho WM, et al. Cannabidiol, a nonpsychotropic plant-derived cannabinoid, decreases inflammation in a murine model of acute lung injury: role for the adenosine A(2A) receptor. Eur J Pharmacol 2012; 678:78–85.
90. Ribeiro A, Almeida VI, Costola-de-Souza C, Ferraz-de-Paula V, Pinheiro ML, Vitoretti LB, et al. Cannabidiol improves lung function and inflammation in mice submitted to LPS-induced acute lung injury. Immunopharmacol Immunotoxicol 2015; 37:35–41.
91. Malfait AM, Gallily R, Sumariwalla PF, Malik AS, Andreakos E, Mechoulam R, et al. The nonpsychoactive cannabis constituent cannabidiol is an oral antiarthritic therapeutic in murine collagen-induced arthritis. Proc Natl Acad Sc USA 2000; 97:9561–9566.
92. Esposito G, Scuderi C, Valenza M, Togna GI, Latina V, De Filippis D, et al. Cannabidiol reduces Abeta-induced neuroinflammation and promotes hippocampal neurogenesis through PPARgamma involvement. PloS One 2011; 6:e28668.
93. Esposito G, Scuderi C, Savani C, Steardo L Jr, De Filippis D, Cottone P, et al. Cannabidiol in vivo blunts beta-amyloid induced neuroinflammation by suppressing IL-1beta and iNOS expression. Br J Pharmacol 2007; 151:1272–1279.
94. Borrelli F, Aviello G, Romano B, Orlando P, Capasso R, Maiello F, et al. Cannabidiol, a safe and nonpsychotropic ingredient of the marijuana plant Cannabis sativa, is protective in a murine model of colitis. J Mol Med (Berl) 2009; 87:1111–1121.
95. Aso E, Sanchez-Pla A, Vegas-Lozano E, Maldonado R, Ferrer I. Cannabis-based medicine reduces multiple pathological processes in AbetaPP/PS1 mice. J Alzheimers Dis 2015; 43:977–991.
96. Lu HC, Mackie K. An introduction to the endogenous cannabinoid system. Biol Psychiatry 2016; 79:516–525.
97. Zgair A, Lee JB, Wong JCM, Taha DA, Aram J, Di Virgilio D, et al. Oral administration of cannabis with lipids leads to high levels of cannabinoids in the intestinal lymphatic system and prominent immunomodulation. Sci Rep 2017; 7:14542.
98. Katona S, Kaminski E, Sanders H, Zajicek J. Cannabinoid influence on cytokine profile in multiple sclerosis. Clin Exp Immunol 2005; 140:580–585.
99. Zgair A, Wong JC, Lee JB, Mistry J, Sivak O, Wasan KM, et al. Dietary fats and pharmaceutical lipid excipients increase systemic exposure to orally administered cannabis and cannabis-based medicines. Am J Transl Res 2016; 8:3448–3459.
100. Massanella M, Gianella S, Schrier R, Dan JM, Perez-Santiago J, Oliveira MF, et al. Methamphetamine use in HIV-infected individuals affects T-cell function and viral outcome during suppressive antiretroviral therapy. Sci Rep 2015; 5:13179.
101. Trevaskis NL, Charman WN, Porter CJ. Targeted drug delivery to lymphocytes: a route to site-specific immunomodulation?. Mol Pharm 2010; 7:2297–2309.
102. Costantino CM, Gupta A, Yewdall AW, Dale BM, Devi LA, Chen BK. Cannabinoid receptor 2-mediated attenuation of CXCR4-tropic HIV infection in primary CD4+ T cells. PloS One 2012; 7:e33961.
103. Ramirez SH, Reichenbach NL, Fan S, Rom S, Merkel SF, Wang X, et al. Attenuation of HIV-1 replication in macrophages by cannabinoid receptor 2 agonists. J Leukoc Biol 2013; 93:801–810.
104. Lu TS, Avraham HK, Seng S, Tachado SD, Koziel H, Makriyannis A, Avraham S. Cannabinoids inhibit HIV-1 Gp120-mediated insults in brain microvascular endothelial cells. J Immunol 2008; 181:6406–6416.
105. Peterson PK, Gekker G, Hu S, Cabral G, Lokensgard JR. Cannabinoids and morphine differentially affect HIV-1 expression in CD4(+) lymphocyte and microglial cell cultures. J Neuroimmunol 2004; 147:123–126.
106. Chen W, Crawford RB, Kaplan BL, Kaminski NE. Modulation of HIVGP120 antigen-specific immune responses in vivo by Δ9-tetrahydrocannabinol. J Neuroimmune Pharmacol 2015; 10:344–355.
107. Lombard C, Hegde VL, Nagarkatti M, Nagarkatti PS. Perinatal exposure to Δ9-tetrahydrocannabinol triggers profound defects in T cell differentiation and function in fetal and postnatal stages of life, including decreased responsiveness to HIV antigens. J Pharmacol Exp Ther 2011; 339:607–617.
108. Molina PE, Winsauer P, Zhang P, Walker E, Birke L, Amedee A, et al. Cannabinoid administration attenuates the progression of simian immunodeficiency virus. AIDS Res Hum Retroviruses 2011; 27:585–592.
109. Wei Q, Liu L, Cong Z, Wu X, Wang H, Qin C, et al. Chronic Δ(9)-tetrahydrocannabinol administration reduces IgE(+)B cells but unlikely enhances pathogenic SIVmac251 infection in male rhesus macaques of Chinese origin. J Neuroimmune Pharmacol 2016; 11:584–591.
110. Molina PE, Amedee AM, LeCapitaine NJ, Zabaleta J, Mohan M, Winsauer PJ, et al. Modulation of gut-specific mechanisms by chronic Δ(9)-tetrahydrocannabinol administration in male rhesus macaques infected with simian immunodeficiency virus: a systems biology analysis. AIDS Res Hum Retroviruses 2014; 30:567–578.
111. Chandra LC, Kumar V, Torben W, Vande Stouwe C, Winsauer P, Amedee A, et al. Chronic administration of Δ9-tetrahydrocannabinol induces intestinal anti-inflammatory microRNA expression during acute simian immunodeficiency virus infection of rhesus macaques. J Virol 2015; 89:1168–1181.
112. Kumar V, Torben W, Kenway CS, Schiro FR, Mohan M. Longitudinal examination of the intestinal lamina propria cellular compartment of simian immunodeficiency virus-infected rhesus macaques provides broader and deeper insights into the link between aberrant microRNA expression and persistent immune activation. J Virol 2016; 90:5003–5019.
113. Kumar V, Mansfield J, Fan R, MacLean A, Li J, Mohan M. miR-130a and miR-212 disrupt the intestinal epithelial barrier through modulation of PPARgamma and occludin expression in chronic simian immunodeficiency virus-infected rhesus macaques. J Immunol 2018; 200:2677–2689.
114. Kumar VTW, Mansfield J, Alvarez Z, Stowe CV, Li J, Byrareddy SN, et al. Cannabinoid attenuation of intestinal inflammation in chronic SIV-infected rhesus macaques involves T cell modulation and differential expression of micro-RNAs and pro-inflammatory genes. Front ImmunolV 10 2019; 914.
115. Keen L 2nd, Abbate A, Blanden G, Priddie C, Moeller FG, Rathore M. Confirmed marijuana use and lymphocyte count in black people living with HIV. Drug Alcohol Depend 2019; 198:112–115.
116. Okafor CN, Zhou Z, Burrell LE 2nd, Kelso NE, Whitehead NE, Harman JS, et al. Marijuana use and viral suppression in persons receiving medical care for HIV-infection. Am J Drug Alcohol Abuse 2017; 43:103–110.
117. Bonn-Miller MO, Oser ML, Bucossi MM, Trafton JA. Cannabis use and HIV antiretroviral therapy adherence and HIV-related symptoms. J Behav Med 2014; 37:1–10.
118. Chao C, Jacobson LP, Tashkin D, Martinez-Maza O, Roth MD, Margolick JB, et al. Recreational drug use and T lymphocyte subpopulations in HIV-uninfected and HIV-infected men. Drug Alcohol Depend 2008; 94:165–171.
119. Kelly EM, Dodge JL, Sarkar M, French AL, Tien PC, Glesby MJ, et al. Marijuana use is not associated with progression to advanced liver fibrosis in HIV/hepatitis C virus-coinfected women. Clin Infect Dis 2016; 63:512–518.
120. Marcellin F, Lions C, Rosenthal E, Roux P, Sogni P, Wittkop L, et al. HEPAVIH ANRS CO13 Study Group*. No significant effect of cannabis use on the count and percentage of circulating CD4 T-cells in HIV-HCV co-infected patients (ANRS CO13-HEPAVIH French cohort). Drug Alcohol Rev 2017; 36:227–238.
121. Milloy MJ, Marshall B, Kerr T, Richardson L, Hogg R, Guillemi S, et al. High-intensity cannabis use associated with lower plasma human immunodeficiency virus-1 RNA viral load among recently infected people who use injection drugs. Drug Alcohol Rev 2015; 34:135–140.
122. Coates RA, Farewell VT, Raboud J, Read SE, MacFadden DK, Calzavara LM, et al. Cofactors of progression to acquired immunodeficiency syndrome in a cohort of male sexual contacts of men with human immunodeficiency virus disease. Am J Epidemiol 1990; 132:717–722.
123. Adams JW, Bryant KJ, Edelman EJ, Fiellin DA, Gaither JR, Gordon AJ, et al. Association of cannabis, stimulant, and alcohol use with mortality prognosis among HIV-infected men. AIDS Behav 2018; 22:1341–1351.
124. Manuzak JA, Gott TM, Kirkwood JS, Coronado E, Hensley-McBain T, Miller C, et al. Heavy cannabis use associated with reduction in activated and inflammatory immune cell frequencies in antiretroviral therapy-treated human immunodeficiency virus-infected individuals. Clin Infect Dis 2018; 66:1872–1882.
125. Rizzo MD, Crawford RB, Henriquez JE, Aldhamen YA, Gulick P, Amalfitano A, et al. HIV-infected cannabis users have lower circulating CD16+ monocytes and IFN-gamma-inducible protein 10 levels compared with nonusing HIV patients. AIDS 2018; 32:419–429.
126. Vidot DC, Manuzak JA, Klatt NR, Pallikkuth S, Roach M, Dilworth SE, et al. Hazardous cannabis use and monocyte activation among methamphetamine users with treated HIV infection. J Acquir Immune Defic Syndr 2019; 81:361–364.
127. Ghosn J, Leruez-Ville M, Blanche J, Delobelle A, Beaudoux C, Mascard L, et al. Evarist–ANRS EP 49 Study Group. HIV-1 DNA levels in peripheral blood mononuclear cells and cannabis use are associated with intermittent HIV shedding in semen of men who have sex with men on successful antiretroviral regimens. Clin Infect Dis 2014; 58:1763–1770.
128. Chaillon A, Nakazawa M, Anderson C, Christensen-Quick A, Ellis RJ, Franklin D, et al. Effect of cannabis use on HIV DNA during suppressive ART. Clin Infect Dis 2019; pii: ciz387. doi: 10.1093/cid/ciz387. [Epub ahead of print].
129. Abrams DI, Hilton JF, Leiser RJ, Shade SB, Elbeik TA, Aweeka FT, et al. Short-term effects of cannabinoids in patients with HIV-1 infection: a randomized, placebo-controlled clinical trial. Ann Intern Med 2003; 139:258–266.
130. Bredt BM, Higuera-Alhino D, Shade SB, Hebert SJ, McCune JM, Abrams DI. Short-term effects of cannabinoids on immune phenotype and function in HIV-1-infected patients. J Clin Pharmacol 2002; 42:82S–89S.
131. Amedee AM, Nichols WA, LeCapitaine NJ, Stouwe CV, Birke LL, Lacour N, et al. Chronic Δ (9)-tetrahydrocannabinol administration may not attenuate simian immunodeficiency virus disease progression in female rhesus macaques. AIDS Res Hum Retroviruses 2014; 30:1216–1225.
132. Cichon C, Sabharwal H, Ruter C, Schmidt MA. MicroRNAs regulate tight junction proteins and modulate epithelial/endothelial barrier functions. Tissue Barriers 2014; 2:e944446.
133. Molina PE, Amedee A, LeCapitaine NJ, Zabaleta J, Mohan M, Winsauer P, Vande Stouwe C, et al. Cannabinoid neuroimmune modulation of SIV disease. J Neuroimmune Pharmacol 2011; 6:516–527.
134. Impact of cannabis on inflammation and viral persistence in treated HIV/SIV. National Institutes of Health Grants Pages. [Accessed 1 June from http://grantome.com/grant/NIH/DP1-DA037979-01]
135. Brunet L, Moodie EE, Rollet K, Cooper C, Walmsley S, Potter M, et al. Marijuana smoking does not accelerate progression of liver disease in HIV-hepatitis C coinfection: a longitudinal cohort analysis. Clin Infect Dis 2013; 57:663–670.
136. Wong KL, Yeap WH, Tai JJ, Ong SM, Dang TM, Wong SC. The three human monocyte subsets: implications for health and disease. Immunol Res 2012; 53:41–57.
137. Ziegler-Heitbrock L. The CD14+ CD16+ blood monocytes: their role in infection and inflammation. J Leuk Biol 2007; 81:584–592.
138. Han J, Wang B, Han N, Zhao Y, Song C, Feng X, et al. CD14(high)CD16(+) rather than CD14(low)CD16(+) monocytes correlate with disease progression in chronic HIV-infected patients. J Acquir Immune Defic Syndr 2009; 52:553–559.
139. Fischer-Smith T, Croul S, Sverstiuk AE, Capini C, L’Heureux D, Regulier EG, et al. CNS invasion by CD14+/CD16+ peripheral blood-derived monocytes in HIV dementia: perivascular accumulation and reservoir of HIV infection. J Neurovirol 2001; 7:528–541.
140. Carrico AW, Cherenack EM, Roach ME, Riley ED, Oni O, Dilworth SE, et al. Substance-associated elevations in monocyte activation among methamphetamine users with treated HIV infection. Aids 2018; 32:767–771.
141. Adamson SJ, Kay-Lambkin FJ, Baker AL, Lewin TJ, Thornton L, Kelly BJ, et al. An improved brief measure of cannabis misuse: the Cannabis Use Disorders Identification Test-Revised (CUDIT-R). Drug Alcohol Depend 2010; 110:137–143.
142. Cohen MS, Hoffman IF, Royce RA, Kazembe P, Dyer JR, Daly CC, et al. Reduction of concentration of HIV-1 in semen after treatment of urethritis: implications for prevention of sexual transmission of HIV-1. AIDSCAP Malawi Research Group. Lancet 1997; 349:1868–1873.
143. Abrams DI, Jay CA, Shade SB, Vizoso H, Reda H, Press S, et al. Cannabis in painful HIV-associated sensory neuropathy: a randomized placebo-controlled trial. Neurology 2007; 68:515–521.
144. Ellis RJ, Toperoff W, Vaida F, van den Brande G, Gonzales J, Gouaux B, et al. Smoked medicinal cannabis for neuropathic pain in HIV: a randomized, crossover clinical trial. Neuropsychopharmacology 2009; 34:672–680.
145. Lutge EE, Gray A, Siegfried N. The medical use of cannabis for reducing morbidity and mortality in patients with HIV/AIDS. Cochrane Database Syst Rev 2013; 4:CD005175.
146. Costiniuk CT, Saneei Z, Routy JP, Margolese S, Mandarino E, Singer J, et al. Oral cannabinoids in people living with HIV on effective antiretroviral therapy: CTN PT028-study protocol for a pilot randomised trial to assess safety, tolerability and effect on immune activation. BMJ Open 2019; 9:e024793.
147. Windle SB, Wade K, Filion KB, Kimmelman J, Thombs BD, Eisenberg MJ. Potential harms from legalization of recreational cannabis use in Canada. Can J Public Health 2019; 110:222–226.
148. Canadian Broadcast Company (CBC) news: Legalization leaves Canada poised to lead on cannabis research ms [Accessed 22 May]
Keywords:

AIDS; cannabis; HIV; inflammation; marijuana; medical cannabinoids

Copyright © 2019 Wolters Kluwer Health, Inc.