Canna~Fangled Abstracts

Could we co-opt the cannabinoid system for asthma therapy?

By July 11, 2023No Comments


Altmetric

Editorial
Received 10 Apr 2023, Accepted 06 Jul 2023, Accepted author version posted online: 07 Jul 2023, Published online: 10 Jul 2023

KEYWORDS:

1. Body of the article

Asthma is a major health problem of increasing prevalence affecting both children and adults with a high socio-economic impact [Citation1]. Around 350 million people suffer from asthma, leading to a significant impairment of the patients’ quality of life and to an annual care costs of around €20 billion in Europe [Citation1–3]. Asthma is a chronic inflammatory disorder of the airways characterized by variable and recurring symptoms (wheezing, reversible airflow obstruction, chest tightness and cough), bronchial hyperresponsiveness (BHR) and chronic airway inflammation [Citation4,Citation5]. Asthma is a highly heterogeneous syndrome encompassing different phenotypes/endotypes. According to the expression levels of type 2 biomarkers and clinical features, asthma can be categorized as follows: i) type 2 immune-mediated asthma, which is characterized by blood eosinophilia (>150 cells/µL−1), tissue eosinophilia (>2% in sputum), elevated serum IgE levels and clinically allergen-driven, elevated FeNO (>19.5 ppb) and upper airways comorbidities; ii) non-type 2 asthma encompassing neutrophilic, paucigranulocytic and obesity-associated asthma [Citation4,Citation6,Citation7]. Alarmins TSLP, IL-33 and IL-25 produced by bronchial epithelial cells (BECs) activate dendritic cells (DCs) and type 2 innate lymphoid cells (ILC2s), thus initiating and perpetuating type 2 inflammatory responses, characterized by increased numbers of CD4+ Th2 cells and ILC2s in lungs and peripheral blood [Citation5]. Type 2 cytokines (IL-4, IL-13, IL-5 and IL-9) contribute to eosinophilia, BHR, mucus production and IgE levels [Citation5,Citation8,Citation9]. Non-type 2 asthma is characterized by increased levels of Th1 and/or Th17 cells, which together with macrophages, ILC subsets, fibroblasts, smooth muscle cells and BECs’ oxidative stress promote apoptosis of bronchial epithelium and neutrophil recruitment, leading to severe clinical manifestations [Citation6,Citation10]. Allergic sensitization and infections are risk factors for asthma development and progression to difficult-to-treat and severe asthma [Citation11,Citation12]. Severe asthma represent 5–10% of the entire asthma population but it is associated with significant comorbidity (including oral corticosteroid side (OCS) effects) and with the highest healthcare resource use [Citation1,Citation4,Citation13]. Corticosteroids and bronchodilators represent the mainstay treatment for asthma and many patients remain controlled with them. However, the management of severe asthma continues to be challenging. The recent advent of biologicals was a major breakthrough in the treatment of severe type 2 asthma [Citation1]. Biologicals show efficacy in reducing exacerbations, improving lung function, and reducing OCS, but the main drawbacks include the high costs of the treatment and the lack of suitable predictive biomarkers for patient selection. Importantly, they show poor efficacy for severe non-type 2 asthmatic patients. Therefore, there is an unmet need for the development of novel therapies for the prevention and treatment of severe asthma.

The human endogenous cannabinoid system (ECS) is a complex signaling network involved in a large number of physiological processes [Citation14,Citation15]. The ECS comprises the endocannabinoid ligands (anandamide (AEA), 2-arachidonoylglycerol (2-AG) as well as other minor ω-6 (n-6) and ω-3 (n-3) fatty acids compounds), the proteins related to their synthesis (N-acyl-phosphatidylethanolamine (NAPE)-hydrolyzing phospholipase D (NAPE-PLD) and sn-1-specific diacylglycerol lipase (DAGL)) and degradation (fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL)) and the cannabinoid receptors (CBRs). Endocannabinoids bind to the canonical G-protein coupled cannabinoid receptor 1 (CB1) and 2 (CB2). CB1 is largely expressed in the central nervous system but also in peripheral tissues and immune cells [Citation16]. CB2 is mainly expressed in immune cells, but also in neurons and other cell types [Citation17]. Alternative CBRs include other orphan GPCRs, TRPVs or PPARs [Citation14,Citation15]. Phytocannabinoids from Cannabis sativa L., marijuana, such as Δ9-tetrahydrocannabinol (Δ9-THC), also activate CBRs and downstream signaling pathways regulating proliferation, differentiation, cell survival, metabolism or immunity [Citation15]. Similarly, a plethora of functional synthetic cannabinoids have been generated, including agonists with similar affinity for CB1/CB2 (WIN55212–2 or HU210), CB1-selective (R-(+)-methanandamide), CB2-selective (HU308) and selective antagonists for CB1 (rimonabant) or CB2 (AM630), among others [Citation14,Citation15]. Alterations in the ECS signaling pathways and changes in the endocannabinoid levels are associated to different diseases such as, neurological and neuropsychiatric conditions, pain and inflammation, obesity, metabolic disorders, septic shock and other inflammatory immune-mediated diseases, including asthma [Citation14,Citation15].

Most animal studies showed cannabinoids as potent suppressors of immune-mediated responses in asthma, but some contradictory results have been reported [Citation15]. Δ9-THC prevents the IgE sensitization and pro-inflammatory cytokines in the lung [Citation18]. Similarly, cannabidiol (CBD), a major active ingredient of marijuana that does not produce psychotropic effects, restores lung function and impairs asthma features in a mouse model of allergic asthma by reducing cytokine production, airway remodeling and BHR [Citation19]. Supporting these data, FAAH and MAGL inhibitor treatments impairs airway inflammation and BHR in an OVA-induced allergic inflammation guinea pig model [Citation20]. Different studies convincingly demonstrated that endocannabinoids, synthetic cannabinoids and phytocannabinoids can also reduce neuroinflammation by inhibiting afferent firing and inflammatory neuropeptide release [Citation21]. Some of the beneficial effects of cannabinoids in these asthma models might be partially due to their reported bronchodilatory properties. For example, inhaled AEA reduces leukotriene D4-induced airway obstruction in guinea pigs [Citation22] and similar effects have been also reported for CBD [Citation23]. In contrast, other mice studies showed that CB2 enhances eosinophil recruitment and responsiveness in the lung, thus worsening allergen-induced pulmonary inflammation [Citation24]. In the same line, CB2 knockout mice display reduced airway inflammation than wild-type mice upon house dust mite sensitization and provocation, which dependent on the negative regulation exerted by NK cells on ILC2s in a CB2-dependent manner [Citation25]. More recently, it was shown that CB2 directly regulates ILC2s by promoting their expansion and functional activation as key contributors of airway inflammation both in mice and humans [Citation26]. However, other studies in guinea pig models demonstrated potential beneficial effects of CB2 activation in the inhibition of antigen-induced plasma extravasation in the airways, inhibition of neurogenic inflammation as well as in the prevention of asthma development [Citation27–29]. Collectively, these data indicate that cannabinoids suppress asthma features in the airways, but there might be conditions under which cannabinoids might also exert pro-inflammatory and deleterious properties.

The first evidence of the potential therapeutic role of cannabinoids in asthma was reported in the 1970s when initial studies demonstrated bronchodilatory properties of marijuana smoke and oral administration of Δ9-THC [Citation30]. However, in some asthmatic patients aerosolized Δ9-THC induced bronchospasm [Citation31], which together with concerns related to its psychotropic effects have hampered further clinical developments. Mechanistically, CB1 activation control nerve-mediated cholinergic contractions and prevented BHR both in mice and humans, confirming a probable protective role in asthma [Citation32–34]. AEA can exert dual effects on bronchial responsiveness in rodents. AEA inhibits capsaicin-induced BHR in axon terminals of airways nerves in a CB1 dependent manner but induces bronchospasm when the constricting tone exerted by the vagus nerve is removed [Citation35], which might explain some of the paradoxical bronchoconstrictory responses reported in asthmatic patients. Discrepancies noted between mice and human data might be also related to the capacity of allergenic proteins contained in cannabis to induce allergic responses in sensitized marijuana smokers [Citation36]. At this regard, it is important to clearly differentiate between the role of allergenic proteins contained in cannabis able to trigger allergic reactions and cannabinoids lipid mediators as small molecules with anti-inflammatory and immunomodulatory features. The use of proper synthetic cannabinoids as potential therapeutic drugs for asthma might completely overcome potential risk associated to cannabis allergic sensitization. Although data in humans are still scarce compare to animal models [Citation15], CB1 plays a potent inhibitory role on human mast cell activation in the airway mucosa [Citation37]. Increased levels of AEA in the bronchoalveolar lavage fluid of asthma patients upon allergen challenge and increased mRNA levels of CB1 in peripheral blood mononuclear cells and tonsils from atopic and asthma patients have been reported [Citation38,Citation39], suggesting that the ECS might play an important role in the pathogenesis of asthma in humans. We have recently shown that the synthetic cannabinoid WIN55212–2 induces human tolerogenic DCs with the capacity of generating highly suppressive regulatory T (Treg) cells by mechanisms depending on autophagy induction and metabolic reprogramming [Citation40]. Human T cells, B cells and different subsets of DCs express functional CBRs [Citation39]. WIN55212–2 also suppress T cell responses and promote Treg cells in human tonsils [Citation41], impair inflammatory properties in human macrophages [Citation42] and prevent peanut-allergic sensitization by promoting the generation of functional Treg cells [Citation43]. The generation and maintenance of functional Treg cells plays a key role in the restoration of healthy immune responses in the airways of asthma patients and other inflammatory diseases [Citation8,Citation44]. Remarkably, WIN55212–2 restores rhinovirus-induced epithelial barrier disruption [Citation45], suggesting that this synthetic cannabinoid also directly acts on human BECs favoring their restoration upon viral infections, which together with allergic sensitization constitute a major risk factor for asthma development. This barrier protective effects and its immunomodulatory and anti-inflammatory properties, point out WIN55212–2 as a potential suitable cannabinoid for asthma treatment.

2. Expert opinion

Compelling experimental evidence both in animal models and humans demonstrates that the ECS plays an important role in the pathophysiology of asthma by regulating immune system cells and different key structural cells, such as smooth muscle cells, endothelial cells, BECs, fibroblasts and neurons in the airways. There is a potential scenario for cannabinoids or molecules targeting ECS as novel therapeutic interventions in asthma, but a better understanding of the role played by specific cannabinoid ligands and CBRs on the different cell types is required. The generation or modification of already existing synthetic cannabinoids able to target specific CBRs in the periphery avoiding CNS-mediated psychotropic effects might well provide the suitable framework for the development of more selective cannabinoid-based therapeutic interventions for asthma patients. At this regard, several compounds targeting CB2, peripherally restricted CB1 and endocannabinoids degrading enzymes have demonstrated to attenuate neuropathic pain with limited side effects [Citation46,Citation47]. Importantly, large-scale clinical trials to determine whether specific cannabinoids might well be safe and effective drugs for asthma treatment are warranted.

Acknowledgments

The author’s laboratory is supported by grant PID2020-114396RB-I00 to OP from MINECO, Spain.

Declaration of interest

O Palomares has received fees for lectures or participation in Advisory Boards from AstraZeneca, GSK, Pfizer, Inmunotek SL, Novartis, Sanofi Genzyme, and Regeneron. OP has received research grants from Inmunotek SL, Novartis SL, Amgen-AstraZeneca, MINECO, MICINNIN and CAM. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Additional information

Funding

This paper was not funded.

References

  • Agache IAkdis CAAkdis M, et al. EAACI biologicals guidelines—recommendations for severe asthma. Allergy. 2021;76(1):1444. doi: 10.1111/all.14425 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Vianello ACaminati MCrivellaro M, et al. Fatal asthma; is it still an epidemic? World Allergy Organ J. 2016;9:42. doi: 10.1186/s40413-016-0129-9 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Accordini SCorsico AGBraggion M, et al. The cost of persistent asthma in Europe: an international population-based study in adults. Int Arch Allergy Immunol. 2013;160(1):93101. doi: 10.1159/000338998 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Global Strategy for Asthma Management and Prevention, Global Initiative for Asthma (GINA) 2020. Availble from: http://www.ginasthma.org/Accessed: Dec, 2020. [Google Scholar]
  • Akdis CAArkwright PDBruggen MC, et al. Type 2 immunity in the skin and lungs. Allergy. 2020;75(7):15821605. doi: 10.1111/all.14318 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Wenzel SE. Asthma phenotypes: the evolution from clinical to molecular approaches. Nat Med. 2012;18(5):716725. doi: 10.1038/nm.2678 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Israel EReddel HKDrazen JMSevere and difficult-to-treat asthma in adults. N Engl J Med. 2017;377(10):965976. doi: 10.1056/NEJMra1608969 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Palomares OAkdis MMartin-Fontecha M, et al. Mechanisms of immune regulation in allergic diseases: the role of regulatory T and B cells. Immunol Rev. 2017;278(1):219236. doi: 10.1111/imr.12555 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Palomares OAkdis AC. Chapter 20: immunology of the asthmatic response. Elsevier2020. [Google Scholar]
  • Hudey SNLedford DKCardet JCMechanisms of non-type 2 asthma. Curr Opin Immunol. 2020;66:123128. doi: 10.1016/j.coi.2020.10.002 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Jackson DJGern JELemanske RF Jr. The contributions of allergic sensitization and respiratory pathogens to asthma inception. J Allergy Clin Immunol. 2016;137(3):659665. quiz 66. doi: 10.1016/j.jaci.2016.01.002 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Pavord IDBeasley RAgusti A, et al. After asthma: redefining airways diseases. Lancet. 2018;391(10118):350400. doi: 10.1016/S0140-6736(17)30879-6 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Custovic AJohnston SLPavord I, et al. EAACI position statement on asthma exacerbations and severe asthma. Allergy. 2013;68(12):15201531. doi: 10.1111/all.12275 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Velasco GSanchez CGuzman MAnticancer mechanisms of cannabinoids. Curr Oncol. 2016;23(11):S2332. doi: 10.3747/co.23.3080 [Crossref][PubMed][Google Scholar]
  • Angelina APerez-Diego MLopez-Abente J, et al. The role of cannabinoids in allergic diseases. Int Arch Allergy Immunol. 2020;181(8):565584. [Crossref][PubMed][Google Scholar]
  • Busquets Garcia ASoria-Gomez EBellocchio L, et al. Cannabinoid receptor type-1: breaking the dogmas. F1000Res. 2016;5:990. doi: 10.12688/f1000research.8245.1 [Crossref][Google Scholar]
  • Simard MRakotoarivelo VDi Marzo V, et al. Expression and functions of the CB(2) receptor in human leukocytes. Front Pharmacol. 2022;13:826400. doi: 10.3389/fphar.2022.826400 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Jan TRFarraj AKHarkema JR, et al. Attenuation of the ovalbumin-induced allergic airway response by cannabinoid treatment in A/J mice. Toxicol Appl Pharmacol. 2003;188(1):2435. doi: 10.1016/S0041-008X(03)00010-3 [Crossref][PubMed][Web of Science ®][Google Scholar]

    •• This study shows that cannabinoid treatment impair allergen-induced airway inflamation and asthma features in vivo in a mouse model of OVA-induced allergy.

  • Vuolo FAbreu SCMichels M, et al. Cannabidiol reduces airway inflammation and fibrosis in experimental allergic asthma. Eur J Pharmacol. 2019;843:251259. doi: 10.1016/j.ejphar.2018.11.029 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Abohalaka RKaraman YRecber T, et al. Endocannabinoid metabolism inhibition ameliorates ovalbumin-induced allergic airway inflammation and hyperreactivity in Guinea pigs. Life Sci. 2022;306:120808. doi: 10.1016/j.lfs.2022.120808 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • McKenna MMcDougall JJCannabinoid control of neurogenic inflammation. Br J Pharmacol. 2020;177(19):43864399. doi: 10.1111/bph.15208 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Stengel PWCockerham SLSilbaugh SAInhaled anandamide reduces leukotriene D4-induced airway obstruction in guinea pigs. Eur J Pharmacol. 2007;557(1):6668. doi: 10.1016/j.ejphar.2006.11.025 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Dudasova AKeir SDParsons ME, et al. The effects of cannabidiol on the antigen-induced contraction of airways smooth muscle in the guinea-pig. Pulm Pharmacol Ther. 2013;26(3):373379. doi: 10.1016/j.pupt.2013.02.002 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Frei RBLuschnig PParzmair GP, et al. Cannabinoid receptor 2 augments eosinophil responsiveness and aggravates allergen-induced pulmonary inflammation in mice. Allergy. 2016;71(7):944956. doi: 10.1111/all.12858 [Crossref][PubMed][Web of Science ®][Google Scholar]

    •• This study shows that CB2 activation promotes eosinophil activation in the airways and worsen airway inflammation triggered by allergens in vivo in mice.

  • Ferrini MEHong SStierle A, et al. CB2 receptors regulate natural killer cells that limit allergic airway inflammation in a murine model of asthma. Allergy. 2017;72(6):937947. doi: 10.1111/all.13107 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Hurrell BPHelou DGShafiei-Jahani P, et al. Cannabinoid receptor 2 engagement promotes group 2 innate lymphoid cell expansion and enhances airway hyperreactivity. J Allergy Clin Immunol. 2022;149(5):162842 e10. doi: 10.1016/j.jaci.2021.09.037 [Crossref][PubMed][Google Scholar]
  • Fukuda HAbe TYoshihara SThe cannabinoid receptor agonist WIN 55,212-2 inhibits antigen-induced plasma extravasation in guinea pig airways. Int Arch Allergy Immunol. 2010;152(3):295300. doi: 10.1159/000283042 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Yoshihara SMorimoto HOhori M, et al. Endogenous cannabinoid receptor agonists inhibit neurogenic inflammations in guinea pig airways. Int Arch Allergy Immunol. 2005;138(1):8087. doi: 10.1159/000087361 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Giannini LNistri SMastroianni R, et al. Activation of cannabinoid receptors prevents antigen-induced asthma-like reaction in guinea pigs. J Cell Mol Med. 2008;12(6a):23812394. doi: 10.1111/j.1582-4934.2008.00258.x [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Vachon LFitzGerald MXSolliday NH, et al. Single-dose effects of marihuana smoke. bronchial dynamics and respiratory-center sensitivity in normal subjects. N Engl J Med. 1973;288(19):985989. doi: 10.1056/NEJM197305102881902 [Crossref][PubMed][Google Scholar]
  • Tashkin DPReiss SShapiro BJ, et al. Bronchial effects of aerosolized delta 9-tetrahydrocannabinol in healthy and asthmatic subjects. Am Rev Respir Dis. 1977;115:5765. [PubMed][Web of Science ®][Google Scholar]
  • Bozkurt TEKaya YDurlu-Kandilci NT, et al. The effect of cannabinoids on dinitrofluorobenzene-induced experimental asthma in mice. Respir Physiol Neurobiol. 2016;231:713. doi: 10.1016/j.resp.2016.05.012 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Grassin-Delyle SNaline EBuenestado A, et al. Cannabinoids inhibit cholinergic contraction in human airways through prejunctional CB1 receptors. Br J Pharmacol. 2014;171(11):27672777. doi: 10.1111/bph.12597 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Bozkurt TELarsson OAdner MStimulation of cannabinoid CB1 receptors prevents nerve-mediated airway hyperreactivity in NGF-induced inflammation in mouse airways. Eur J Pharmacol. 2016;776:132138. doi: 10.1016/j.ejphar.2016.02.045 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Calignano AKatona IDesarnaud F, et al. Bidirectional control of airway responsiveness by endogenous cannabinoids. Nature. 2000;408(6808):96101. doi: 10.1038/35040576 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Toscano AEbo DGAbbas K, et al. A review of cannabis allergy in the early days of legalization. Ann Allergy Asthma Immunol. 2023;130(3):288295. doi: 10.1016/j.anai.2022.10.016 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Sugawara KZakany NHundt T, et al. Cannabinoid receptor 1 controls human mucosal-type mast cell degranulation and maturation in situ. J Allergy Clin Immunol. 2013;132(1):182193. doi: 10.1016/j.jaci.2013.01.002 [Crossref][PubMed][Google Scholar]

    • This study shows that CB1 activation on airway mast cells inhibits their maturation, degranulation and release of anaphylactogenic mediators in situ.

  • Zoerner AAStichtenoth DOEngeli S, et al. Allergen challenge increases anandamide in bronchoalveolar fluid of patients with allergic asthma. Clin Pharmacol Ther. 2011;90(3):388391. doi: 10.1038/clpt.2011.94 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Martin-Fontecha MAngelina ARuckert B, et al. A fluorescent probe to unravel functional features of cannabinoid receptor CB1 in human blood and tonsil immune system cells. Bioconjug Chem. 2018;29(2):382389. doi: 10.1021/acs.bioconjchem.7b00680 [Crossref][PubMed][Web of Science ®][Google Scholar]

    •• This study shows that a plethora of human hemotopoietic blood and tonsil cells express functional CB1 by using a specific fluorescence probe.

  • Angelina APerez-Diego MLopez-Abente J, et al. Cannabinoids induce functional tregs by promoting tolerogenic DCs via autophagy and metabolic reprograming. Mucosal Immunol. 2022;15(1):96108. doi: 10.1038/s41385-021-00455-x [Crossref][PubMed][Google Scholar]

    •• This study demostrates that synthetic cannabinoid WIN55212-2 promote the generation of human tolerogenic DCs able to polarize functional Treg cells by mechanisms depending on autophagy induction and subsquent metabolic reprogramming

  • Angelina APerez-Diego MMaldonado A, et al. The cannabinoid WIN55212-2 suppresses effector T-cell responses and promotes regulatory T cells in human tonsils. Allergy. 2022;77(3):10291032. doi: 10.1111/all.15160 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Perez-Diego MAngelina AMartín-Cruz L, et al. Cannabinoid WIN55,212-2 reprograms monocytes and macrophages to inhibit LPS-induced inflammation. Front Immunol. 2023;14:14. [Crossref][Google Scholar]

    • This study demostrates that synthetic cannabinoid WIN55212-2 also display antiinflammatory properties by acting on human macrophages and reprogramming monocytes

  • Angelina AJimenez-Saiz RPerez-Diego M, et al. Cannabinoid WIN55212-2 impairs peanut-allergic sensitization and promotes the generation of allergen-specific regulatory T cells. Clin Exp Allergy. 2022;52(4):540549. doi: 10.1111/cea.14092 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Palomares OElewaut DIrving PM, et al. Regulatory T cells and immunoglobulin E: a new therapeutic link for autoimmunity? Allergy. 2022;77(11):32933308. doi: 10.1111/all.15449 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Angelina AMartin-Fontecha MRuckert B, et al. The cannabinoid WIN55212-2 restores rhinovirus-induced epithelial barrier disruption. Allergy. 2020;76(6):19001902. doi: 10.1111/all.14707 [Crossref][PubMed][Google Scholar]

    •• This study demostrates that synthetic cannabinoid WIN55212-2 is able to restore the damage induced upon rhinovirus infection in human bronchial epithelial cells

  • Romero-Sandoval EAAsbill SPaige CA, et al. Peripherally restricted cannabinoids for the treatment of pain. Pharmacotherapy. 2015;35(10):917925. doi: 10.1002/phar.1642 [Crossref][PubMed][Web of Science ®][Google Scholar]
  • Hossain MZAndo HUnno S, et al. Targeting peripherally restricted cannabinoid receptor 1, cannabinoid receptor 2, and endocannabinoid-degrading enzymes for the treatment of neuropathic pain including neuropathic orofacial pain. Int J Mol Sci. 2020;21(4):21. doi: 10.3390/ijms21041423 [Crossref][Web of Science ®][Google Scholar]

Leave a Reply