1
|
Meanti R, Bresciani E, Rizzi L, Molteni L, Coco S, Omeljaniuk RJ, Torsello A. Cannabinoid Receptor 2 (CB2R) as potential target for the pharmacological treatment of neurodegenerative diseases. Biomed Pharmacother 2025; 186:118044. [PMID: 40209306 DOI: 10.1016/j.biopha.2025.118044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 03/25/2025] [Accepted: 04/03/2025] [Indexed: 04/12/2025] Open
Abstract
The endocannabinoid system (ECS) is a ubiquitous physiological system that plays a crucial role in maintaining CNS homeostasis and regulating its functions. It includes cannabinoid receptors (CBRs), endogenous cannabinoids (eCBs), and the enzymes responsible for their synthesis and degradation. In recent years, growing evidence has highlighted the therapeutic potential of the ECS and CBRs, in a wide range of severe diseases and pathological conditions, including Alzheimer's and Parkinson's diseases, Amyotrophic Lateral Sclerosis, Multiple Sclerosis, Huntington's Disease, HIV-1 associated neurocognitive disorders, neuropathic pain and migraine. Targeting the cannabinoid type 2 receptor (CB2R) has gained attention due to its ability to (i) mitigate neuroinflammatory responses, (ii) regulate mitochondrial function and (iii) provide trophic support, all without eliciting the psychotropic actions associated with CB1R activation. This review aims to explore the potential of CB2R modulation as a strategy for the prevention and treatment of neurologic disorders, exploring both preclinical and clinical findings.
Collapse
Affiliation(s)
- Ramona Meanti
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Elena Bresciani
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Laura Rizzi
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Laura Molteni
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Silvia Coco
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| | - Robert J Omeljaniuk
- Department of Biology, Lakehead University, 955 Oliver Rd, Thunder Bay, Ontario P7B 5E1, Canada.
| | - Antonio Torsello
- School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, Monza 20900, Italy.
| |
Collapse
|
2
|
Kremsmayr T, Schober G, Kaltenböck M, Hoare BL, Brierley SM, Muttenthaler M. Oxytocin Analogues for the Oral Treatment of Abdominal Pain. Angew Chem Int Ed Engl 2024; 63:e202415333. [PMID: 39384545 DOI: 10.1002/anie.202415333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/11/2024]
Abstract
Abdominal pain presents an onerous reality for millions of people affected by gastrointestinal disorders such as irritable bowel syndrome (IBS) and inflammatory bowel diseases (IBD). The oxytocin receptor (OTR) has emerged as a new analgesic drug target with OTR expression upregulated on colon-innervating nociceptors in chronic visceral hypersensitivity states, accessible via luminal delivery. However, the low gastrointestinal stability of OTR's endogenous peptide ligand oxytocin (OT) is a bottleneck for therapeutic development. Here, we report the development of potent and fully gut-stable OT analogues, laying the foundation for a new area of oral gut-specific peptide therapeutics. Ligand optimisation guided by structure-gut-stability-activity relationships yielded highly stable analogues (t1/2>24 h, compared to t1/2<10 min of OT in intestinal fluid) equipotent to OT (~3 nM) and with enhanced OTR selectivity. Intra-colonic administration of the lead ligand significantly reduced colonic mechanical hypersensitivity in a concentration-dependent manner in a mouse model of chronic abdominal pain. Moreover, oral administration of the lead ligand also displayed significant analgesia in this abdominal pain mouse model. The generated ligands and employed strategies could pave the way to a new class of oral gut-specific peptides to study and combat chronic gastrointestinal disorders, an area with substantial unmet medical needs.
Collapse
Affiliation(s)
- Thomas Kremsmayr
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Straße 38, 1090, Vienna, Austria
| | - Gudrun Schober
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
| | - Matthias Kaltenböck
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Straße 38, 1090, Vienna, Austria
| | - Bradley L Hoare
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Straße 38, 1090, Vienna, Austria
| | - Stuart M Brierley
- Visceral Pain Research Group, Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, South Australia, 5000, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide, South Australia, 5000, Australia
| | - Markus Muttenthaler
- University of Vienna, Faculty of Chemistry, Institute of Biological Chemistry, Währinger Straße 38, 1090, Vienna, Austria
- Institute for Molecular Bioscience, The University of Queensland, 4072, Brisbane, Queensland, Australia
| |
Collapse
|
3
|
Ramsay S, Yew WP, Brookes S, Zagorodnyuk V. A combination of peripherally restricted CB 1 and CB 2 cannabinoid receptor agonists reduces bladder afferent sensitisation in cystitis. Eur J Pharmacol 2024; 985:177078. [PMID: 39532227 DOI: 10.1016/j.ejphar.2024.177078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/14/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Cannabinoid agonists can potentially ameliorate lower urinary tract symptoms (LUTS), including pain associated with interstitial cystitis/bladder pain syndrome (IC/BPS). This study aims to determine the contributions of the cannabinoid 1 receptors (CB1Rs) and CB2Rs in regulating the activity of different functional classes of afferents, comparing normal healthy bladder with bladders from guinea pigs with protamine/zymosan-induced cystitis. The mechanosensitivity of different functional afferent classes was determined by ex vivo single-unit extracellular recordings. Peripherally restricted CB1R preferential agonists, ACEA and PrNMI and peripherally restricted CB2R selective agonists, 4Q3C and olorinab all reduced the mechanosensitivity of mucosal bladder afferents. The potency and efficacy of these synthetic cannabinoid agonists were significantly increased in cystitis compared to controls. Combined application of CB1R agonists, ACEA or PrNMI with the CB2R agonist, 4Q3C produced additive inhibitory effects. ACEA and PrNMI also inhibited the stretch-induced firing of high-threshold muscular bladder afferents in animals with cystitis. In contrast, low- and high-threshold muscular-mucosal bladder afferents were unaffected by the CB1R and CB2R agonists in control and cystitis. Our data indicated that peripherally restricted CB1R and CB2R agonists effectively reduce the sensitisation of probable nociceptive afferents in the bladder in cystitis. The findings also suggest a potential benefit of simultaneously targeting both the CB1Rs and CB2Rs to ameliorate LUTS in cystitis.
Collapse
Affiliation(s)
- Stewart Ramsay
- Discipline of Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, South Australia, Australia
| | - Wai Ping Yew
- Discipline of Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, South Australia, Australia
| | - Simon Brookes
- Discipline of Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, South Australia, Australia
| | - Vladimir Zagorodnyuk
- Discipline of Human Physiology, Flinders Health & Medical Research Institute, College of Medicine and Public Health, Flinders University, South Australia, Australia.
| |
Collapse
|
4
|
Larauche M, Mulak A, Ha C, Million M, Arnett S, Germano P, Pearson JP, Currie MG, Taché Y. FAAH inhibitor URB597 shows anti-hyperalgesic action and increases brain and intestinal tissues fatty acid amides in a model of CRF 1 agonist mediated visceral hypersensitivity in male rats. Neurogastroenterol Motil 2024; 36:e14927. [PMID: 39344695 PMCID: PMC11781189 DOI: 10.1111/nmo.14927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/05/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024]
Abstract
BACKGROUND AND AIMS The endocannabinoid (eCB) system includes ligands (anandamide and 2-arachidonoyl glycerol, 2-AG), receptors and catabolizing enzymes (fatty acid amide hydrolase, FAAH and monoacylglycerol lipase) expressed in both the brain and gut. We investigated whether the FAAH inhibitor, URB597, influenced visceral pain to colorectal distension (CRD) in an acute stress-related model of visceral hypersensitivity induced by the selective corticotropin-releasing factor receptor subtype 1 (CRF1) agonist, cortagine. METHODS Male Sprague-Dawley rats were injected subcutaneously (SC) with URB597 (3 mg/kg) or vehicle and 2 h later, intraperitoneally with cortagine (10 μg/kg) or vehicle. The visceromotor responses (VMR) were assessed to a first CRD (baseline) before injections, and to a second CRD 15 min after the last treatment. Brain, jejunum, and proximal colon were collected from treated and naïve rats for levels quantification of three fatty acid amides (FAAs) [anandamide (arachidonyl-ethanolamide, AEA), oleoyl-ethanolamide (OEA) and palmitoyl-ethanolamide (PEA)], and 2-AG. In separate animals, defecation/diarrhea were monitored after URB597 and cortagine. KEY RESULTS URB597 inhibited cortagine-induced increased VMR at 40 mmHg (89.0 ± 14.8% vs. 132.5 ± 15.6% for vehicle SC, p < 0.05) and 60 mmHg (107.5 ± 16.1% vs. 176.9 ± 24.4% for vehicle SC, p < 0.001) while not influencing basal VMR. In URB597 plus cortagine group, FAAs levels increased in the brain and intestinal tissue while 2-AG did not change. URB597 did not modify cortagine-induced defecation/diarrhea versus vehicle. CONCLUSIONS AND INFERENCES URB597 shows efficacy to elevate brain and intestinal FAAs and to counteract the colonic hypersensitivity induced by peripheral activation of CRF1 signaling supporting a potential strategy of FAAH inhibitors to alleviate stress-related visceral hypersensitivity.
Collapse
Affiliation(s)
- Muriel Larauche
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Agata Mulak
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Present Affiliation: Department of Gastroenterology and Hepatology, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland
| | - Chrysanthy Ha
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Present Affiliation: Community Memorial Healthcare, Ventura, CA, USA
| | - Mulugeta Million
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Stacy Arnett
- Present Affiliation: Department of Pathology, St. Louis University, St. Louis, MO, USA
| | - Peter Germano
- Present Affiliation: Auron Therapeutics, Inc., Newton, MA, USA
| | - James P. Pearson
- Present Affiliation: Sea Pharmaceuticals LLC, Cambridge, MA, USA
| | - Mark G. Currie
- Present Affiliation: Sea Pharmaceuticals LLC, Cambridge, MA, USA
| | - Yvette Taché
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90056, USA
- VA Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| |
Collapse
|
5
|
Castro J, Maddern J, Erickson A, Harrington AM, Brierley SM. Peripheral and central neuroplasticity in a mouse model of endometriosis. J Neurochem 2024; 168:3777-3800. [PMID: 37165846 DOI: 10.1111/jnc.15843] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 04/24/2023] [Accepted: 04/26/2023] [Indexed: 05/12/2023]
Abstract
Chronic pelvic pain (CPP) is the most debilitating symptom of gynaecological disorders such as endometriosis. However, it remains unclear how sensory neurons from pelvic organs affected by endometriosis, such as the female reproductive tract, detect and transmit nociceptive events and how these signals are processed within the central nervous system (CNS). Using a previously characterized mouse model of endometriosis, we investigated whether the increased pain sensitivity occurring in endometriosis could be attributed to (i) changes in mechanosensory properties of sensory afferents innervating the reproductive tract, (ii) alterations in sensory input from reproductive organs to the spinal cord or (iii) neuroinflammation and sensitization of spinal neural circuits. Mechanosensitivity of vagina-innervating primary afferents was examined using an ex vivo single-unit extracellular recording preparation. Nociceptive signalling from the vagina to the spinal cord was quantified by phosphorylated MAP kinase ERK1/2 immunoreactivity. Immunohistochemistry was used to determine glial and neuronal circuit alterations within the spinal cord. We found that sensory afferents innervating the rostral, but not caudal portions of the mouse vagina, developed mechanical hypersensitivity in endometriosis. Nociceptive signalling from the vagina to the spinal cord was significantly enhanced in mice with endometriosis. Moreover, mice with endometriosis developed microgliosis, astrogliosis and enhanced substance P neurokinin-1 receptor immunoreactivity within the spinal cord, suggesting the development of neuroinflammation and sensitization of spinal circuitry in endometriosis. These results demonstrate endometriosis-induced neuroplasticity occurring at both peripheral and central sites of sensory afferent pathways. These findings may help to explain the altered sensitivity to pain in endometriosis and provide a novel platform for targeted pain relief treatments for this debilitating disorder.
Collapse
Affiliation(s)
- Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jessica Maddern
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Andelain Erickson
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Andrea M Harrington
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
6
|
Castro J, Maddern J, Chow CY, Tran P, Vetter I, King GF, Brierley SM. The voltage-gated sodium channel Na V1.7 underlies endometriosis-associated chronic pelvic pain. J Neurochem 2024; 168:3760-3776. [PMID: 36840383 DOI: 10.1111/jnc.15795] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023]
Abstract
Chronic pelvic pain (CPP) is the primary symptom of endometriosis patients, but adequate treatments are lacking. Modulation of ion channels expressed by sensory nerves innervating the viscera has shown promise for the treatment of irritable bowel syndrome and overactive bladder. However, similar approaches for endometriosis-associated CPP remain underdeveloped. Here, we examined the role of the voltage-gated sodium (NaV) channel NaV1.7 in (i) the sensitivity of vagina-innervating sensory afferents and investigated whether (ii) NaV1.7 inhibition reduces nociceptive signals from the vagina and (iii) ameliorates endometriosis-associated CPP. The mechanical responsiveness of vagina-innervating sensory afferents was assessed with ex vivo single-unit recording preparations. Pain evoked by vaginal distension (VD) was quantified by the visceromotor response (VMR) in vivo. In control mice, pharmacological activation of NaV1.7 with OD1 sensitised vagina-innervating pelvic afferents to mechanical stimuli. Using a syngeneic mouse model of endometriosis, we established that endometriosis sensitised vagina-innervating pelvic afferents to mechanical stimuli. The highly selective NaV1.7 inhibitor Tsp1a revealed that this afferent hypersensitivity occurred in a NaV1.7-dependent manner. Moreover, in vivo intra-vaginal treatment with Tsp1a reduced the exaggerated VMRs to VD which is characteristic of mice with endometriosis. Conversely, Tsp1a did not alter ex vivo afferent mechanosensitivity nor in vivo VMRs to VD in Sham control mice. Collectively, these findings suggest that NaV1.7 plays a crucial role in endometriosis-induced vaginal hyperalgesia. Importantly, NaV1.7 inhibition selectively alleviated endometriosis-associated CPP without the loss of normal sensation, suggesting that selective targeting of NaV1.7 could improve the quality of life of women with endometriosis.
Collapse
Affiliation(s)
- Joel Castro
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Jessica Maddern
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
| | - Chun Yuen Chow
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St Lucia, Queensland, Australia
| | - Poanna Tran
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- School of Pharmacy, The University of Queensland, Brisbane, Australia
| | - Glenn F King
- Institute for Molecular Bioscience, The University of Queensland, St Lucia, Queensland, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, St Lucia, Queensland, Australia
| | - Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
7
|
Rakotoarivelo V, Mayer TZ, Simard M, Flamand N, Di Marzo V. The Impact of the CB 2 Cannabinoid Receptor in Inflammatory Diseases: An Update. Molecules 2024; 29:3381. [PMID: 39064959 PMCID: PMC11279428 DOI: 10.3390/molecules29143381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
The emergence of inflammatory diseases is a heavy burden on modern societies. Cannabis has been used for several millennia to treat inflammatory disorders such as rheumatism or gout. Since the characterization of cannabinoid receptors, CB1 and CB2, the potential of cannabinoid pharmacotherapy in inflammatory conditions has received great interest. Several studies have identified the importance of these receptors in immune cell migration and in the production of inflammatory mediators. As the presence of the CB2 receptor was documented to be more predominant in immune cells, several pharmacological agonists and antagonists have been designed to treat inflammation. To better define the potential of the CB2 receptor, three online databases, PubMed, Google Scholar and clinicaltrial.gov, were searched without language restriction. The full texts of articles presenting data on the endocannabinoid system, the CB2 receptor and its role in modulating inflammation in vitro, in animal models and in the context of clinical trials were reviewed. Finally, we discuss the clinical potential of the latest cannabinoid-based therapies in inflammatory diseases.
Collapse
Affiliation(s)
- Volatiana Rakotoarivelo
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
| | - Thomas Z. Mayer
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, and Centre NUTRISS, École de Nutrition, Université Laval, Québec City, QC G1V 0V6, Canada
| | - Mélissa Simard
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
| | - Nicolas Flamand
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
| | - Vincenzo Di Marzo
- Centre de Recherche de l’Institut Universitaire De Cardiologie Et De Pneumologie de Québec, Département of Médecine, Université Laval, Québec City, QC G1V 4G5, Canada
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec City, QC G1V 0V6, Canada
- Institut sur la Nutrition et les Aliments Fonctionnels, and Centre NUTRISS, École de Nutrition, Université Laval, Québec City, QC G1V 0V6, Canada
- Joint International Unit between the CNR of Italy and Université Laval on Chemical and Biomolecular Research on the Microbiome and Its Impact on Metabolic Health and Nutrition (UMI-MicroMeNu), Québec City, QC G1V 0V6, Canada
| |
Collapse
|
8
|
Carruthers ER, Grimsey NL. Cannabinoid CB 2 receptor orthologues; in vitro function and perspectives for preclinical to clinical translation. Br J Pharmacol 2024; 181:2247-2269. [PMID: 37349984 DOI: 10.1111/bph.16172] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 05/01/2023] [Accepted: 05/22/2023] [Indexed: 06/24/2023] Open
Abstract
Cannabinoid CB2 receptor agonists are in development as therapeutic agents, including for immune modulation and pain relief. Despite promising results in rodent preclinical studies, efficacy in human clinical trials has been marginal to date. Fundamental differences in ligand engagement and signalling responses between the human CB2 receptor and preclinical model species orthologues may contribute to mismatches in functional outcomes. This is a tangible possibility for the CB2 receptor in that there is a relatively large degree of primary amino acid sequence divergence between human and rodent. Here, we summarise CB2 receptor gene and protein structure, assess comparative molecular pharmacology between CB2 receptor orthologues, and review the current status of preclinical to clinical translation for drugs targeted at the CB2 receptor, focusing on comparisons between human, mouse and rat receptors. We hope that raising wider awareness of, and proposing strategies to address, this additional challenge in drug development will assist in ongoing efforts toward successful therapeutic translation of drugs targeted at the CB2 receptor. LINKED ARTICLES: This article is part of a themed issue Therapeutic Targeting of G Protein-Coupled Receptors: hot topics from the Australasian Society of Clinical and Experimental Pharmacologists and Toxicologists 2021 Virtual Annual Scientific Meeting. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v181.14/issuetoc.
Collapse
Affiliation(s)
- Emma R Carruthers
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Natasha L Grimsey
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| |
Collapse
|
9
|
Ford AC, Vanner S, Kashyap PC, Nasser Y. Chronic Visceral Pain: New Peripheral Mechanistic Insights and Resulting Treatments. Gastroenterology 2024; 166:976-994. [PMID: 38325759 PMCID: PMC11102851 DOI: 10.1053/j.gastro.2024.01.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/15/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
Chronic visceral pain is one of the most common reasons for patients with gastrointestinal disorders, such as inflammatory bowel disease or disorders of brain-gut interaction, to seek medical attention. It represents a substantial burden to patients and is associated with anxiety, depression, reductions in quality of life, and impaired social functioning, as well as increased direct and indirect health care costs to society. Unfortunately, the diagnosis and treatment of chronic visceral pain is difficult, in part because our understanding of the underlying pathophysiologic basis is incomplete. In this review, we highlight recent advances in peripheral pain signaling and specific physiologic and pathophysiologic preclinical mechanisms that result in the sensitization of peripheral pain pathways. We focus on preclinical mechanisms that have been translated into treatment approaches and summarize the current evidence base for directing treatment toward these mechanisms of chronic visceral pain derived from clinical trials. The effective management of chronic visceral pain remains of critical importance for the quality of life of suffers. A deeper understanding of peripheral pain mechanisms is necessary and may provide the basis for novel therapeutic interventions.
Collapse
Affiliation(s)
- Alexander C Ford
- Leeds Institute of Medical Research at St. James's, University of |Leeds, Leeds, United Kingdom; Leeds Gastroenterology Institute, Leeds Teaching Hospitals National Health Service Trust, Leeds, United Kingdom
| | - Stephen Vanner
- Gastrointestinal Diseases Research Unit, Kingston General Hospital, Queen's University, Kingston, Ontario, Canada
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, Minnesota
| | - Yasmin Nasser
- Snyder Institute for Chronic Diseases, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
10
|
Mozaffari S, Nikfar S, Abdollahi M. Drugs of the future for diarrhea-predominant irritable bowel syndrome: an overview of current investigational drugs. Expert Opin Investig Drugs 2024; 33:219-228. [PMID: 38366822 DOI: 10.1080/13543784.2024.2320703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 02/15/2024] [Indexed: 02/18/2024]
Abstract
INTRODUCTION Irritable bowel syndrome (IBS) has a significant impact on society and quality of life. Current treatments are ineffective, and new investigational drugs are necessary. AREAS COVERED Numerous potential therapies are developing, targeting different areas such as cannabinoid signaling, opioid receptors, tachykinin (NK2) receptors, β3-adrenergic receptors, intestinal microbiota, inflammation, and 5HT receptors. Clinical trial evidence has shown that loperamide, eluxadoline, alosetron, ramosetron, bile acid sequestrants, and rifaximin can modulate GI alterations and benefit patients with IBS-D. Among the potential therapies, ibodutant, ibudilast, blautix, BOS-589, solabegron, vibegron, olorinab, ebastine, and ORP-101 have demonstrated possible effects but remain confirmed. EXPERT OPINION Individuals with IBS-D require cost-effective treatment options that do not impede their productivity or that of their caregivers. This is necessary for consistent healthcare and improved quality of life. Therefore, we should focus on developing new, efficient, and affordable medications for IBS-D. The government, insurers, and society must recognize this need and collaborate to ensure its fulfillment.
Collapse
Affiliation(s)
- Shilan Mozaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, and Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Shekoufeh Nikfar
- Department of Pharmacoeconomics and Pharmaceutical Administration, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Personalized Medicine Research Center, Endocrinology and Metabolism Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Abdollahi
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, and Pharmaceutical Sciences Research Center (PSRC), Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
11
|
Wardill HR, Wooley LT, Bellas OM, Cao K, Cross CB, van Dyk M, Kichenadasse G, Bowen JM, Zannettino ACW, Shakib S, Crawford GB, Boublik J, Davis MM, Smid SD, Price TJ. Supporting gut health with medicinal cannabis in people with advanced cancer: potential benefits and challenges. Br J Cancer 2024; 130:19-30. [PMID: 37884682 PMCID: PMC10781684 DOI: 10.1038/s41416-023-02466-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/08/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
The side effects of cancer therapy continue to cause significant health and cost burden to the patient, their friends and family, and governments. A major barrier in the way in which these side effects are managed is the highly siloed mentality that results in a fragmented approach to symptom control. Increasingly, it is appreciated that many symptoms are manifestations of common underlying pathobiology, with changes in the gastrointestinal environment a key driver for many symptom sequelae. Breakdown of the mucosal barrier (mucositis) is a common and early side effect of many anti-cancer agents, known to contribute (in part) to a range of highly burdensome symptoms such as diarrhoea, nausea, vomiting, infection, malnutrition, fatigue, depression, and insomnia. Here, we outline a rationale for how, based on its already documented effects on the gastrointestinal microenvironment, medicinal cannabis could be used to control mucositis and prevent the constellation of symptoms with which it is associated. We will provide a brief update on the current state of evidence on medicinal cannabis in cancer care and outline the potential benefits (and challenges) of using medicinal cannabis during active cancer therapy.
Collapse
Affiliation(s)
- Hannah R Wardill
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia.
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.
| | - Luke T Wooley
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Olivia M Bellas
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Katrina Cao
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- School of Public Health, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Courtney B Cross
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Supportive Oncology Research Group, Precision Cancer Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Madele van Dyk
- Flinders Centre for Innovation in Cancer, Flinders Medical Centre/Flinders University, SA Health, Adelaide, SA, Australia
| | - Ganessan Kichenadasse
- Flinders Centre for Innovation in Cancer, Flinders Medical Centre/Flinders University, SA Health, Adelaide, SA, Australia
- Northern Adelaide Local Health Network South Australia, SA Health, Adelaide, SA, Australia
| | - Joanne M Bowen
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Andrew C W Zannettino
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Sepehr Shakib
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Gregory B Crawford
- Northern Adelaide Local Health Network South Australia, SA Health, Adelaide, SA, Australia
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | | | - Mellar M Davis
- The Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | - Scott D Smid
- The School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
| | - Timothy J Price
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, SA, Australia
- Queen Elizabeth Hospital, Adelaide, SA, Australia
| |
Collapse
|
12
|
Camilleri M, Zheng T. Cannabinoids and the Gastrointestinal Tract. Clin Gastroenterol Hepatol 2023; 21:3217-3229. [PMID: 37678488 PMCID: PMC10872845 DOI: 10.1016/j.cgh.2023.07.031] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/18/2023] [Accepted: 07/25/2023] [Indexed: 09/09/2023]
Abstract
The synthesis and degradation of endocannabinoids, location of cannabinoid (CB) receptors, and cannabinoid mechanisms of action on immune/inflammatory, neuromuscular, and sensory functions in digestive organs are well documented. CB2 mechanisms are particularly relevant in immune and sensory functions. Increasing use of cannabinoids in the United States is impacted by social determinants of health including racial discrimination, which is associated with tobacco and cannabis co-use, and combined use disorders. Several conditions associated with emesis are related to cannabinoid use, including cannabinoid hyperemesis or withdrawal, cyclic vomiting syndrome, and nausea and vomiting of pregnancy. Cannabinoids generally inhibit gastrointestinal motor function; yet they relieve symptoms in patients with gastroparesis and diverse nausea syndromes. Cannabinoid effects on inflammatory mechanisms have shown promise in relatively small placebo-controlled studies in reducing disease activity and abdominal pain in patients with inflammatory bowel disease. Cannabinoids have been studied in disorders of motility, pain, and disorders of gut-brain interaction. The CB2-receptor agonist, cannabidiol, reduced the total Gastroparesis Cardinal Symptom Index and increases the ability to tolerate a meal in patients with gastroparesis appraised over 4 weeks of treatment. In contrast, predominant-pain end points in functional dyspepsia with normal gastric emptying were not improved significantly with cannabidiol. The CB2 agonist, olorinab, reduced abdominal pain in inflammatory bowel disease in an open-label trial and in constipation-predominant irritable bowel syndrome in a placebo-controlled trial. Cannabinoid mechanisms alter inflammation in pancreatic and liver diseases. In conclusion, cannabinoids, particularly agents affecting CB2 mechanisms, have potential for inflammatory, gastroparesis, and pain disorders; however, the trials require replication and further understanding of risk-benefit to enhance use of cannabinoids in gastrointestinal diseases.
Collapse
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota.
| | - Ting Zheng
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
13
|
Chang L, Cash BD, Lembo A, Kunkel DC, English BA, Lindstrom B, Gu G, Skare S, Gilder K, Turner S, Cataldi F, Lipkis D, Tack J. Efficacy and safety of olorinab, a full agonist of the cannabinoid receptor 2, for the treatment of abdominal pain in patients with irritable bowel syndrome: Results from a phase 2b randomized placebo-controlled trial (CAPTIVATE). Neurogastroenterol Motil 2023; 35:e14539. [PMID: 36740814 DOI: 10.1111/nmo.14539] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 12/19/2022] [Accepted: 01/11/2023] [Indexed: 02/07/2023]
Abstract
BACKGROUND Olorinab is a highly selective, peripherally acting, full agonist of cannabinoid receptor 2. This study assessed the efficacy and safety of olorinab to treat abdominal pain in patients with irritable bowel syndrome with diarrhea (IBS-D) and constipation (IBS-C). METHODS CAPTIVATE was a phase 2b, randomized, double-blind, placebo-controlled, parallel-group trial. Eligible participants aged 18-70 years with IBS-C and IBS-D diagnosed per Rome IV received olorinab 10 mg, 25 mg, or 50 mg three times daily (TID) or placebo TID for 12 weeks. The primary endpoint was the change in patient-reported average abdominal pain score (AAPS) from baseline to Week 12. KEY RESULTS A total of 273 participants were randomized to receive olorinab 10 mg (n = 67), olorinab 25 mg (n = 67), olorinab 50 mg (n = 69), or placebo (n = 70). Although a treatment response was observed across all groups, the weekly change in average AAPS from baseline to Week 12 was not significantly different between placebo and any olorinab dose. In a prespecified subgroup analysis of participants with a baseline AAPS ≥6.5, olorinab 50 mg (n = 35) significantly improved AAPS compared with placebo (n = 30) (p = 0.014). Adverse event rates were comparable between olorinab and placebo and there were no reported serious adverse events or deaths. CONCLUSION AND INFERENCES Although olorinab was well-tolerated and improved weekly AAPS, the primary endpoint was not met. However, in participants with moderate-to-severe pain at baseline (AAPS ≥6.5), olorinab 50 mg significantly improved weekly AAPS compared with placebo. CLINICALTRIALS gov: NCT04043455.
Collapse
Affiliation(s)
- Lin Chang
- David Geffen School of Medicine, UCLA, Los Angeles, California, USA
| | - Brooks D Cash
- University of Texas Health Science Center Houston, Houston, Texas, USA
| | - Anthony Lembo
- Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - David C Kunkel
- University of California San Diego, La Jolla, California, USA
| | | | | | - Guibao Gu
- Arena Pharmaceuticals, Inc., San Diego, California, USA
| | - Sharon Skare
- Arena Pharmaceuticals, Inc., San Diego, California, USA
| | - Kye Gilder
- Arena Pharmaceuticals, Inc., San Diego, California, USA
| | | | - Fabio Cataldi
- Arena Pharmaceuticals, Inc., San Diego, California, USA
| | - Donald Lipkis
- Sharp Grossmont Hospital, San Diego, California, USA
| | - Jan Tack
- University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
14
|
Niang LY, Heckroth M, Mathur P, Abell TL. Gastroparesis syndromes: emerging drug targets and potential therapeutic opportunities. Expert Opin Investig Drugs 2023; 32:245-262. [PMID: 36872904 DOI: 10.1080/13543784.2023.2186222] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023]
Abstract
INTRODUCTION Gastroparesis (Gp) and related disorders such as chronic unexplained nausea and vomiting and functional dyspepsia, known as gastropareis syndromes (GpS), have large unmet needs. Mainstays of GpS treatments are diet and drugs. AREAS COVERED The purpose of this review is to explore potential new medications and other therapies for gastroparesis. Before discussing possible new drugs, the currently used drugs are discussed. These include dopamine receptor antagonists, 5-hydroxytryptamine receptor agonists and antagonists, neurokinin-1 receptor antagonists and other anti-emetics. The article also considers future drugs that may be used for Gp, based on currently known pathophysiology. EXPERT OPINION Gaps in knowledge about the pathophysiology of gastroparesis and related syndromes are critical to developing therapeutic agents that will be successful. Recent major developments in the gastroparesis arena are related to microscopic anatomy, cellular function, and pathophysiology. The major challenges moving forward will be to develop the genetic and biochemical correlates of these major developments in gastroparesis research.
Collapse
Affiliation(s)
- Le Yu Niang
- Department of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, Kentucky, USA
| | - Matthew Heckroth
- Department of Medicine, University of Louisville, Louisville, Kentucky, USA
| | - Prateek Mathur
- Department of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, Kentucky, USA
| | - Thomas L Abell
- Department of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
15
|
Brierley SM, Greenwood-Van Meerveld B, Sarnelli G, Sharkey KA, Storr M, Tack J. Targeting the endocannabinoid system for the treatment of abdominal pain in irritable bowel syndrome. Nat Rev Gastroenterol Hepatol 2023; 20:5-25. [PMID: 36168049 DOI: 10.1038/s41575-022-00682-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2022] [Indexed: 12/27/2022]
Abstract
The management of visceral pain in patients with disorders of gut-brain interaction, notably irritable bowel syndrome, presents a considerable clinical challenge, with few available treatment options. Patients are increasingly using cannabis and cannabinoids to control abdominal pain. Cannabis acts on receptors of the endocannabinoid system, an endogenous system of lipid mediators that regulates gastrointestinal function and pain processing pathways in health and disease. The endocannabinoid system represents a logical molecular therapeutic target for the treatment of pain in irritable bowel syndrome. Here, we review the physiological and pathophysiological functions of the endocannabinoid system with a focus on the peripheral and central regulation of gastrointestinal function and visceral nociception. We address the use of cannabinoids in pain management, comparing them to other treatment modalities, including opioids and neuromodulators. Finally, we discuss emerging therapeutic candidates targeting the endocannabinoid system for the treatment of pain in irritable bowel syndrome.
Collapse
Affiliation(s)
- Stuart M Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute, Flinders University, Bedford Park, South Australia, Australia
- Hopwood Centre for Neurobiology, Lifelong Health, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia
| | | | - Giovanni Sarnelli
- Department of Clinical Medicine and Surgery, University of Naples "Federico II", Naples, Italy
| | - Keith A Sharkey
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.
| | - Martin Storr
- Department of Medicine, Ludwig-Maximilians University, Munich, Germany
- Zentrum für Endoskopie, Starnberg, Germany
| | - Jan Tack
- Translational Research Center for Gastrointestinal Disorders, University of Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Yu Y, Tsang QK, Jaramillo-Polanco J, Lomax AE, Vanner SJ, Reed DE. Cannabinoid 1 and mu-Opioid Receptor Agonists Synergistically Inhibit Abdominal Pain and Lack Side Effects in Mice. J Neurosci 2022; 42:6313-6324. [PMID: 35790401 PMCID: PMC9398536 DOI: 10.1523/jneurosci.0641-22.2022] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 05/26/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
While effective in treating abdominal pain, opioids have significant side effects. Recent legalization of cannabis will likely promote use of cannabinoids as an adjunct or alternative to opioids, despite a lack of evidence. We aimed to investigate whether cannabinoids inhibit mouse colonic nociception, alone or in combination with opioids at low doses. Experiments were performed on C57BL/6 male and female mice. Visceral nociception was evaluated by measuring visceromotor responses (VMR), afferent nerve mechanosensitivity in flat-sheet colon preparations, and excitability of isolated DRG neurons. Blood oxygen saturation, locomotion, and defecation were measured to evaluate side effects. An agonist of cannabinoid 1 receptor (CB1R), arachidonyl-2'-chloroethylamide (ACEA), dose-dependently decreased VMR. ACEA and HU-210 (another CB1R agonist) also attenuated colonic afferent nerve mechanosensitivity. Additionally, HU-210 concentration-dependently decreased DRG neuron excitability, which was reversed by the CB1R antagonist AM-251. Conversely, cannabinoid 2 receptor (CB2R) agonists did not attenuate VMR, afferent nerve mechanosensitivity, or DRG neuron excitability. Combination of subanalgesic doses of CB1R and µ-opioid receptor agonists decreased VMR; importantly, this analgesic effect was preserved after 6 d of twice daily treatment. This combination also attenuated afferent nerve mechanosensitivity and DRG neuron excitability, which was inhibited by neuronal nitric oxide synthase and guanylate cyclase inhibitors. This combination avoided side effects (decreased oxygen saturation and colonic transit) caused by analgesic dose of morphine. Activation of CB1R, but not CB2R, decreased colonic nociception both alone and in synergy with µ-opioid receptor. Thus, CB1R agonists may enable opioid dose reduction and avoid opioid-related side effects.SIGNIFICANCE STATEMENT One of the most cited needs for patients with abdominal pain are safe and effective treatment options. The effectiveness of opioids in the management of abdominal pain is undermined by severe adverse side effects. Therefore, strategies to replace opioids or reduce the doses of opioids to suppress abdominal pain is needed. This study in mice demonstrates that cannabinoid 1 receptor (CB1R) agonists inhibit visceral sensation. Furthermore, a combination of subanalgesic doses of µ-opioid receptor agonist and CB1R agonist markedly reduce abdominal pain without causing the side effects of high-dose opioids. Thus, CB1R agonists, alone or in combination with low-dose opioids, may be a novel and safe treatment strategy for abdominal pain.
Collapse
Affiliation(s)
- Yang Yu
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Quentin K Tsang
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Josue Jaramillo-Polanco
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario K7L 2V7, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario K7L 2V7, Canada
| |
Collapse
|
17
|
Whiting ZM, Yin J, de la Harpe SM, Vernall AJ, Grimsey NL. Developing the Cannabinoid Receptor 2 (CB2) pharmacopoeia: past, present, and future. Trends Pharmacol Sci 2022; 43:754-771. [PMID: 35906103 DOI: 10.1016/j.tips.2022.06.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 06/23/2022] [Accepted: 06/27/2022] [Indexed: 12/28/2022]
Abstract
Cannabinoid Receptor 2 (CB2) is a G protein-coupled receptor (GPCR) with considerable, though as yet unrealised, therapeutic potential. Promising preclinical data supports the applicability of CB2 activation in autoimmune and inflammatory diseases, pain, neurodegeneration, and osteoporosis. A diverse pharmacopoeia of cannabinoid ligands is available, which has led to considerable advancements in the understanding of CB2 function and extensive preclinical evaluation. However, until recently, most CB2 ligands were highly lipophilic and as such not optimal for clinical application due to unfavourable physicochemical properties. A number of strategies have been applied to develop CB2 ligands to achieve closer to 'drug-like' properties and a few such compounds have now undergone clinical trial. We review the current state of CB2 ligand development and progress in optimising physicochemical properties, understanding advanced molecular pharmacology such as functional selectivity, and clinical evaluation of CB2-targeting compounds.
Collapse
Affiliation(s)
- Zak M Whiting
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Jiazhen Yin
- Department of Chemistry, Division of Sciences, University of Otago, Dunedin, New Zealand
| | - Sara M de la Harpe
- Department of Chemistry, Division of Sciences, University of Otago, Dunedin, New Zealand
| | - Andrea J Vernall
- Department of Chemistry, Division of Sciences, University of Otago, Dunedin, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand
| | - Natasha L Grimsey
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, New Zealand.
| |
Collapse
|
18
|
Singh R, Zogg H, Ghoshal UC, Ro S. Current Treatment Options and Therapeutic Insights for Gastrointestinal Dysmotility and Functional Gastrointestinal Disorders. Front Pharmacol 2022; 13:808195. [PMID: 35145413 PMCID: PMC8822166 DOI: 10.3389/fphar.2022.808195] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Functional gastrointestinal disorders (FGIDs) have been re-named as disorders of gut-brain interactions. These conditions are not only common in clinical practice, but also in the community. In reference to the Rome IV criteria, the most common FGIDs, include functional dyspepsia (FD) and irritable bowel syndrome (IBS). Additionally, there is substantial overlap of these disorders and other specific gastrointestinal motility disorders, such as gastroparesis. These disorders are heterogeneous and are intertwined with several proposed pathophysiological mechanisms, such as altered gut motility, intestinal barrier dysfunction, gut immune dysfunction, visceral hypersensitivity, altered GI secretion, presence and degree of bile acid malabsorption, microbial dysbiosis, and alterations to the gut-brain axis. The treatment options currently available include lifestyle modifications, dietary and gut microbiota manipulation interventions including fecal microbiota transplantation, prokinetics, antispasmodics, laxatives, and centrally and peripherally acting neuromodulators. However, treatment that targets the pathophysiological mechanisms underlying the symptoms are scanty. Pharmacological agents that are developed based on the cellular and molecular mechanisms underlying pathologies of these disorders might provide the best avenue for future pharmaceutical development. The currently available therapies lack long-term effectiveness and safety for their use to treat motility disorders and FGIDs. Furthermore, the fundamental challenges in treating these disorders should be defined; for instance, 1. Cause and effect cannot be disentangled between symptoms and pathophysiological mechanisms due to current therapies that entail the off-label use of medications to treat symptoms. 2. Despite the knowledge that the microbiota in our gut plays an essential part in maintaining gut health, their exact functions in gut homeostasis are still unclear. What constitutes a healthy microbiome and further, the precise definition of gut microbial dysbiosis is lacking. More comprehensive, large-scale, and longitudinal studies utilizing multi-omics data are needed to dissect the exact contribution of gut microbial alterations in disease pathogenesis. Accordingly, we review the current treatment options, clinical insight on pathophysiology, therapeutic modalities, current challenges, and therapeutic clues for the clinical care and management of functional dyspepsia, gastroparesis, irritable bowel syndrome, functional constipation, and functional diarrhea.
Collapse
Affiliation(s)
- Rajan Singh
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| | - Hannah Zogg
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| | - Uday C Ghoshal
- Department of Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Seungil Ro
- Department of Physiology and Cell Biology, Reno School of Medicine, University of Nevada, Reno, NV, United States
| |
Collapse
|
19
|
Camilleri M. New Drugs on the Horizon for Functional and Motility Gastrointestinal Disorders. Gastroenterology 2021; 161:761-764. [PMID: 33989661 PMCID: PMC8380736 DOI: 10.1053/j.gastro.2021.04.079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/16/2022]
Affiliation(s)
- Michael Camilleri
- Clinical Enteric Neuroscience Translational and Epidemiological Research, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
20
|
Cabañero D, Martín-García E, Maldonado R. The CB2 cannabinoid receptor as a therapeutic target in the central nervous system. Expert Opin Ther Targets 2021; 25:659-676. [PMID: 34424117 DOI: 10.1080/14728222.2021.1971196] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Targeting CB2 cannabinoid receptor (CB2r) represents a promising approach for the treatment of central nervous system disorders. These receptors were identified in peripheral tissues, but also in neurons in the central nervous system. New findings have highlighted the interest to target these central receptors to obtain therapeutic effects devoid of the classical cannabinoid side-effects. AREAS COVERED In this review, we searched PubMed (January 1991-May 2021), ClinicalTrials.gov and Cochrane Library databases for articles, reviews and clinical trials. We first introduce the relevance of CB2r as a key component of the endocannabinoid system. We discuss CB2r interest as a possible novel target in the treatment of pain. This receptor has raised interest as a potential target for neurodegenerative disorders treatment, as we then discussed. Finally, we underline studies revealing a novel potential CB2r interest in mental disorders treatment. EXPERT OPINION In spite of the interest of targeting CB2r for pain, clinical trials evaluating CB2r agonist analgesic efficacy have currently failed. The preferential involvement of CB2r in preventing the development of chronic pain could influence the failure of clinical trials designed for the treatment of already established pain syndromes. Specific trials should be designed to target the prevention of chronic pain development.
Collapse
Affiliation(s)
- David Cabañero
- Institute of Research, Development and Innovation in Healthcare Biotechnology of Elche (IDiBE), Universidad Miguel Hernández. Elche, Alicante, Spain
| | - Elena Martín-García
- Neuropharmacology Laboratory, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain.,IMIM (Hospital Del Mar Medical Research Institute), Barcelona, Spain
| | - Rafael Maldonado
- Neuropharmacology Laboratory, Department of Experimental and Health Sciences, Pompeu Fabra University, Barcelona, Spain.,IMIM (Hospital Del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
21
|
Najjar SA, Albers KM. Pain in Inflammatory Bowel Disease: Optogenetic Strategies for Study of Neural-Epithelial Signaling. CROHN'S & COLITIS 360 2021; 3:otab040. [PMID: 34805983 PMCID: PMC8600958 DOI: 10.1093/crocol/otab040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
Abdominal pain is common in patients with active inflammation of the colon but can persist even in its absence, suggesting other mechanisms of pain signaling. Recent findings suggest colon epithelial cells are direct regulators of pain-sensing neurons. Optogenetic activation of epithelial cells evoked nerve firing and pain-like behaviors. Inhibition of epithelial cells caused the opposite effect, reducing responses to colon distension and inflammatory hypersensitivity. Thus, epithelial cells alone can regulate the activation of pain circuits. Future goals are to define the anatomical and cellular mechanisms that underlie epithelial-neural pain signaling and how it is altered in response to colon inflammation.
Collapse
Affiliation(s)
- Sarah A Najjar
- Department of Neurobiology and Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Present address: Department of Pediatrics, Columbia University Medical Center, New York, New York, USA
| | - Kathryn M Albers
- Department of Neurobiology and Pittsburgh Center for Pain Research, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA,Address correspondence to: Kathryn M. Albers, PhD, Department of Neurobiology, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15216, USA ()
| |
Collapse
|