1
|
Rangari VA, O'Brien ES, Powers AS, Slivicki RA, Bertels Z, Appourchaux K, Aydin D, Ramos-Gonzalez N, Mwirigi J, Lin L, Mangutov E, Sobecks BL, Awad-Agbaria Y, Uphade MB, Aguilar J, Peddada TN, Shiimura Y, Huang XP, Folarin-Hines J, Payne M, Kalathil A, Varga BR, Kobilka BK, Pradhan AA, Cameron MD, Kumar KK, Dror RO, Gereau RW, Majumdar S. A cryptic pocket in CB1 drives peripheral and functional selectivity. Nature 2025; 640:265-273. [PMID: 40044849 PMCID: PMC11977287 DOI: 10.1038/s41586-025-08618-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 01/09/2025] [Indexed: 03/16/2025]
Abstract
The current opioid overdose epidemic highlights the urgent need to develop safer and more effective treatments for chronic pain1. Cannabinoid receptor type 1 (CB1) is a promising non-opioid target for pain relief, but its clinical use has been limited by centrally mediated psychoactivity and tolerance. We overcame both issues by designing peripherally restricted CB1 agonists that minimize arrestin recruitment. We achieved these goals by computationally designing positively charged derivatives of the potent CB1 agonist MDMB-Fubinaca2. We designed these ligands to occupy a cryptic pocket identified through molecular dynamics simulations-an extended binding pocket that opens rarely and leads to the conserved signalling residue D2.50 (ref. 3). We used structure determination, pharmacological assays and molecular dynamics simulations to verify the binding modes of these ligands and to determine the molecular mechanism by which they achieve this dampening of arrestin recruitment. Our lead ligand, VIP36, is highly peripherally restricted and demonstrates notable efficacy in three mouse pain models, with 100-fold dose separation between analgesic efficacy and centrally mediated side effects. VIP36 exerts analgesic efficacy through peripheral CB1 receptors and shows limited analgesic tolerance. These results show how targeting a cryptic pocket in a G-protein-coupled receptor can lead to enhanced peripheral selectivity, biased signalling, desired in vivo pharmacology and reduced adverse effects. This has substantial implications for chronic pain treatment but could also revolutionize the design of drugs targeting other G-protein-coupled receptors.
Collapse
Affiliation(s)
- Vipin Ashok Rangari
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Evan S O'Brien
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Biophysics and Biophysical Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Alexander S Powers
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Richard A Slivicki
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Zachariah Bertels
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin Appourchaux
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Deniz Aydin
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Nokomis Ramos-Gonzalez
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Juliet Mwirigi
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Li Lin
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, USA
| | - Elizaveta Mangutov
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Briana L Sobecks
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Yaseen Awad-Agbaria
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Manoj B Uphade
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Jhoan Aguilar
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Teja Nikhil Peddada
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Yuki Shiimura
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Division of Molecular Genetics, Institute of Life Science, Kurume University, Fukuoka, Japan
| | - Xi-Ping Huang
- Department of Pharmacology School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jakayla Folarin-Hines
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Maria Payne
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Anirudh Kalathil
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
| | - Balazs R Varga
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian K Kobilka
- Department of Computer Science, Stanford University, Stanford, CA, USA
| | - Amynah A Pradhan
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Michael D Cameron
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL, USA
| | | | - Ron O Dror
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Computer Science, Stanford University, Stanford, CA, USA.
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA.
| | - Robert W Gereau
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA.
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.
| | - Susruta Majumdar
- Center for Clinical Pharmacology, University of Health Sciences and Pharmacy and Washington University School of Medicine, St. Louis, MO, USA.
- Department of Anesthesiology and Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
2
|
Warren WG, Osborn M, David‐Pereira A, Tsantoulas C, Xue W, Yates A, OSullivan SE. ART26.12, a novel fatty acid-binding protein 5 inhibitor, shows efficacy in multiple preclinical neuropathy models. Eur J Pain 2025; 29:e4718. [PMID: 39188040 PMCID: PMC11671339 DOI: 10.1002/ejp.4718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/27/2024] [Accepted: 08/01/2024] [Indexed: 08/28/2024]
Abstract
BACKGROUND Painful neuropathy is a pathological condition caused by numerous factors including diabetes, chemotherapy or cancer. ART26.12 is a novel fatty acid-binding protein 5 inhibitor, which our group showed could prevent and treat persistent pain in a preclinical model of oxaliplatin-induced peripheral neuropathy. METHODS In the current study, the efficacy of orally dosed ART26.12 was tested in multiple neuropathy models of different aetiology. Paw withdrawal threshold to von Frey monofilaments and latency to escape a cold plate were used as measurements of mechanical and cold sensitivity. RESULTS ART26.12 (25 and 50 mg/kg BID), dosed prior to the induction of paclitaxel-induced peripheral neuropathy (PIPN), reversed mechanical allodynia induced by paclitaxel in both male and female rats, and ART26.12 (50 mg/kg BID) prevented the induction of PIPN in female rats. ART26.12 (50 mg/kg BID) also had a protective effect on body weight in the PIPN model. ART26.12 (25 and 100 mg/kg BID) reversed mechanical allodynia when treating established streptozotocin-induced diabetic neuropathy in male rats. In a model of breast cancer-induced bone pain in female rats, ART26.12 (100 mg/kg BID) reversed mechanical allodynia within 1 h of dosing. In the same model, ART26.12 (25 mg/kg BID) reversed mechanical allodynia from day 4 of treatment. CONCLUSION Overall, these preclinical data suggest that ART26.12 is a safe and efficacious therapeutic drug for continued development towards the prevention and treatment of peripheral neuropathy. SIGNIFICANCE STATEMENT This work now shows that ART26.12, a novel and selective inhibitor of FABP5, can prevent and treat multiple preclinical models of peripheral neuropathy. Given its excellent safety profile, further work is warranted to develop ART26.12 as a potential therapeutic tool for pain management.
Collapse
Affiliation(s)
- W. G. Warren
- Artelo Biosciences Ltd., Alderley ParkAlderley EdgeCheshireUK
| | - M. Osborn
- Artelo Biosciences Ltd., Alderley ParkAlderley EdgeCheshireUK
| | - A. David‐Pereira
- Transpharmation Ltd.The London Bioscience Innovation CentreLondonUK
| | - C. Tsantoulas
- Transpharmation Ltd.The London Bioscience Innovation CentreLondonUK
| | - Wenwen Xue
- Pharmaron Inc.BeijingPeople's Republic of China
| | - A. Yates
- Artelo Biosciences Ltd., Alderley ParkAlderley EdgeCheshireUK
| | - S. E. OSullivan
- Artelo Biosciences Ltd., Alderley ParkAlderley EdgeCheshireUK
| |
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
|
Zhang S, Chen L, Ladez SR, Seferge A, Liu J, Feng B. Blocking Aδ- and C-fiber neural transmission by sub-kilohertz peripheral nerve stimulation. Front Neurosci 2024; 18:1404903. [PMID: 39077428 PMCID: PMC11284050 DOI: 10.3389/fnins.2024.1404903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/02/2024] [Indexed: 07/31/2024] Open
Abstract
Introduction We recently showed that sub-kilohertz electrical stimulation of the afferent somata in the dorsal root ganglia (DRG) reversibly blocks afferent transmission. Here, we further investigated whether similar conduction block can be achieved by stimulating the nerve trunk with electrical peripheral nerve stimulation (ePNS). Methods We explored the mechanisms and parameters of conduction block by ePNS via ex vivo single-fiber recordings from two somatic (sciatic and saphenous) and one autonomic (vagal) nerves harvested from mice. Action potentials were evoked on one end of the nerve and recorded on the other end from teased nerve filaments, i.e., single-fiber recordings. ePNS was delivered in the middle of the nerve trunk using a glass suction electrode at frequencies of 5, 10, 50, 100, 500, and 1000 Hz. Results Suprathreshold ePNS reversibly blocks axonal neural transmission of both thinly myelinated Aδ-fiber axons and unmyelinated C-fiber axons. ePNS leads to a progressive decrease in conduction velocity (CV) until transmission blockage, suggesting activity-dependent conduction slowing. The blocking efficiency is dependent on the axonal conduction velocity, with Aδ-fibers efficiently blocked by 50-1000 Hz stimulation and C-fibers blocked by 10-50 Hz. The corresponding NEURON simulation of action potential transmission indicates that the disrupted transmembrane sodium and potassium concentration gradients underly the transmission block by the ePNS. Discussion The current study provides direct evidence of reversible Aδ- and C-fiber transmission blockage by low-frequency (<100 Hz) electrical stimulation of the nerve trunk, a previously overlooked mechanism that can be harnessed to enhance the therapeutic effect of ePNS in treating neurological disorders.
Collapse
Affiliation(s)
| | | | | | | | | | - Bin Feng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
5
|
Hou L, Zhang H, Li Y, Zhu H, Liao K, Guo B, Dong C, Li G, Ye W, Wang L, Xu H. Correlation analysis of positron emission tomography/computed tomography-magnetic resonance imaging of cannabinoid type 1 receptor in the lumbar spine and brain of aged osteoporosis female cynomolgus monkeys. Quant Imaging Med Surg 2023; 13:7924-7935. [PMID: 38106237 PMCID: PMC10722013 DOI: 10.21037/qims-23-118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 09/14/2023] [Indexed: 12/19/2023]
Abstract
Background Although cannabinoid receptor 1 (CB1R) antagonists can inhibit bone loss in osteoporosis mouse models, different strains of mice show different bone mass phenotypes after knock out the CB1R gene. The relationship between CB1R and bone metabolism is complex, and its regulatory role in bone metabolism and as a therapeutic target for osteoporosis requires further investigation. Methods Based on lumbar spine volumetric bone mineral density (vBMD) data of healthy female cynomolgus monkeys aged 1-25 years, naturally aged postmenopausal female osteoporotic monkeys and normal young monkeys were screened by detecting lumbar vertebrae vBMD and estradiol levels in this study. Positron emission tomography-computed tomography (PET/CT) and magnetic resonance imaging (MRI) scans were performed on the lumbar spine and brain of the two groups of monkeys using the probe [11C]OMAR, which specifically targets CB1R, and the difference in the CB1R expression of osteoporotic monkeys was evaluated. Results The vBMD values of two standard deviations (SDs) below the peak bone value (428.1±53.8 g/cm3) were set as the reference standard for osteoporosis vBMD. Of the 49 healthy female cynomolgus monkeys, 4 postmenopausal older osteoporotic monkeys (18-26 years) and 5 young control monkeys (6-7 years) were selected, and the mean vBMD of the lumbar spine of the two groups was 295.07±19.11 and 419.72±16.14 g/cm3, respectively (P<0.0001). Radioactive uptake in the lumbar spine was linearly and negatively correlated with vBMD (r=-0.7977; P=0.01). Dynamic PET/MR imaging of the brains showed that CB1R was upregulated in the osteoporosis group, and there was a negative linear correlation between the vBMD and area under the time-radioactivity curve (AUC) of the thalamus (r=-0.8506; P=0.0153) and prefrontal cortex (r=-0.8306; P=0.0207). Conclusions In this study, PET/CT-MRI molecular imaging technology revealed that CB1R was upregulated in the lumbar spine and brain of the osteoporosis monkeys and that CB1R may be regulated by the brain-bone axis. CB1R antagonist may be a potential drug for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Lu Hou
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Haitong Zhang
- Department of Cardiology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Ying Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Honghao Zhu
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Kai Liao
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bin Guo
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Chenchen Dong
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guocong Li
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weijian Ye
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Lu Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Hao Xu
- Department of Nuclear Medicine, The First Affiliated Hospital of Jinan University, Guangzhou, China
| |
Collapse
|
6
|
Thompson AL, Grenald SA, Ciccone HA, Mohty D, Smith AF, Coleman DL, Bahramnejad E, De Leon E, Kasper-Conella L, Uhrlab JL, Margolis DS, Salvemini D, Largent-Milnes TM, Vanderah TW. Morphine-induced osteolysis and hypersensitivity is mediated through toll-like receptor-4 in a murine model of metastatic breast cancer. Pain 2023; 164:2463-2476. [PMID: 37326644 PMCID: PMC10578422 DOI: 10.1097/j.pain.0000000000002953] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/28/2023] [Accepted: 04/18/2023] [Indexed: 06/17/2023]
Abstract
ABSTRACT The propensity for breast cancer to metastasize to bone is coupled to the most common complaint among breast cancer patients: bone pain. Classically, this type of pain is treated using escalating doses of opioids, which lack long-term efficacy due to analgesic tolerance, opioid-induced hypersensitivity, and have recently been linked to enhanced bone loss. To date, the molecular mechanisms underlying these adverse effects have not been fully explored. Using an immunocompetent murine model of metastatic breast cancer, we demonstrated that sustained morphine infusion induced a significant increase in osteolysis and hypersensitivity within the ipsilateral femur through the activation of toll-like receptor-4 (TLR4). Pharmacological blockade with TAK242 (resatorvid) as well as the use of a TLR4 genetic knockout ameliorated the chronic morphine-induced osteolysis and hypersensitivity. Genetic MOR knockout did not mitigate chronic morphine hypersensitivity or bone loss. In vitro studies using RAW264.7 murine macrophages precursor cells demonstrated morphine-enhanced osteoclastogenesis that was inhibited by the TLR4 antagonist. Together, these data indicate that morphine induces osteolysis and hypersensitivity that are mediated, in part, through a TLR4 receptor mechanism.
Collapse
Affiliation(s)
- Austen L. Thompson
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Shaness A. Grenald
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Haley A. Ciccone
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Dieter Mohty
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Angela F. Smith
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Deziree L. Coleman
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Erfan Bahramnejad
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Erick De Leon
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | - Logan Kasper-Conella
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
| | | | - David S. Margolis
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ, United States
- Orthopaedic Surgery, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Daniela Salvemini
- Department of Pharmacology and Physiology and Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University School of Medicine, St. Louis, MO, United States
| | - Tally M. Largent-Milnes
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| | - Todd W. Vanderah
- Department of Medical Pharmacology, University of Arizona College of Medicine, Tucson, AZ, United States
- Comprehensive Pain and Addiction Center, University of Arizona, Tucson, AZ, United States
| |
Collapse
|
7
|
Takei D, Tagami K. Management of cancer pain due to bone metastasis. J Bone Miner Metab 2022; 41:327-336. [PMID: 36418587 DOI: 10.1007/s00774-022-01382-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/13/2022] [Indexed: 11/26/2022]
Abstract
Bone metastases frequently occur in patients with cancer. Skeletal-related events (SREs), including pain, impaired mobility, hypercalcemia, pathological fracture, spinal cord and nerve root compression, and bone marrow infiltration, can decrease the quality of life of the patients and increase the risk of morbidity. The mechanism of pain due to bone metastasis is complicated and involves various interactions among tumor cells, bone cells, activated inflammatory cells, and bone-innervating neurons. Cancer pain due to bone metastasis can be crippling and a chronic state that causes sarcopenia. For pain management, it is important to diagnose whether the pain is based on background pain or breakthrough pain due to bone metastasis. In addition, the management goal of cancer pain due to bone metastasis is not only to achieve pain relief but also to prevent pain progression and SREs. Pain mechanisms should be applied to achieve optimal management. This review aims to discuss the mechanisms of cancer pain due to bone metastasis and review the recommended drug therapies.
Collapse
Affiliation(s)
- Daisuke Takei
- Department of Pharmacy, Saitama Cancer Center, Saitama, Japan.
| | - Keita Tagami
- Department of Palliative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
8
|
Wiese BM, Liktor-Busa E, Couture SA, Nikas SP, Ji L, Liu Y, Makriyannis A, Spigelman I, Vanderah TW, Largent-Milnes TM. Brain Penetrant, but not Peripherally Restricted, Synthetic Cannabinoid 1 Receptor Agonists Promote Morphine-Mediated Respiratory Depression. Cannabis Cannabinoid Res 2022; 7:621-627. [PMID: 34935460 PMCID: PMC9587769 DOI: 10.1089/can.2021.0090] [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] [Indexed: 11/12/2022] Open
Abstract
Introduction: Cannabis acceptance and use continues to rise despite the gaps in knowledge regarding the mechanisms of cannabinoids and the endocannabinoid system in many physiological functions, including respiratory influence. Methods: With recent evidence of cannabinoid receptor 1 (CB1R) presence in the collection of respiratory neurons in the brainstem, as well as in the peripheral lung tissue, it is vital that the mechanisms involved in central and peripheral CB1R modulation of respiratory function be delineated. In this study we sought to define the roles of central versus peripheral CB1R activation on respiratory depression alone and in combination with morphine using whole body plethysmography. Results: We show that the peripherally restricted CB1 agonist (4-{2-[-(1E)-1[(4-propylnaphthalen-1-yl)methylidene]-1H-inden-3yl]ethyl}morpholine [PrNMI] 0.3, 0.6, and 1 mg/kg) does not induce respiratory depression, while our previous studies showed that a central penetrating synthetic cannabinoid does induce respiratory depression. Significantly, the combination of morphine with the peripheral CB1 agonist, PrNMI, attenuated morphine-induced respiratory depression. Conclusions: These studies support that a peripherally restricted CB1R agonist may be a unique strategy to attenuate the respiratory depression associated with opioid therapy.
Collapse
Affiliation(s)
- Beth M. Wiese
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Erika Liktor-Busa
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Sarah A. Couture
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Spyros P. Nikas
- Chemistry and Chemical Biology, Bouve College Health Sciences—Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| | - Lipin Ji
- Chemistry and Chemical Biology, Bouve College Health Sciences—Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| | - Yingpeng Liu
- Chemistry and Chemical Biology, Bouve College Health Sciences—Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| | - Alexandros Makriyannis
- Chemistry and Chemical Biology, Bouve College Health Sciences—Center for Drug Discovery, Northeastern University, Boston, Massachusetts, USA
| | - Igor Spigelman
- Division of Oral Biology and Medicine, School of Dentistry, University of California, Los Angeles, California, USA
| | - Todd W. Vanderah
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
- Comprehensive Pain and Addiction Center, University of Arizona, Health Sciences, Tucson, Arizona, USA
| | - Tally M. Largent-Milnes
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
- Comprehensive Pain and Addiction Center, University of Arizona, Health Sciences, Tucson, Arizona, USA
| |
Collapse
|
9
|
Slivicki RA, Yi J, Brings VE, Huynh PN, Gereau RW. The cannabinoid agonist CB-13 produces peripherally mediated analgesia in mice but elicits tolerance and signs of central nervous system activity with repeated dosing. Pain 2022; 163:1603-1621. [PMID: 34961756 PMCID: PMC9281468 DOI: 10.1097/j.pain.0000000000002550] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 11/24/2021] [Indexed: 11/25/2022]
Abstract
ABSTRACT Activation of cannabinoid receptor type 1 (CB 1 ) produces analgesia in a variety of preclinical models of pain; however, engagement of central CB 1 receptors is accompanied by unwanted side effects, such as psychoactivity, tolerance, and dependence. Therefore, some efforts to develop novel analgesics have focused on targeting peripheral CB 1 receptors to circumvent central CB 1 -related side effects. In the present study, we evaluated the effects of acute and repeated dosing with the peripherally selective CB 1 -preferring agonist CB-13 on nociception and central CB 1 -related phenotypes in a model of inflammatory pain in mice. We also evaluated cellular mechanisms underlying CB-13-induced antinociception in vitro using cultured mouse dorsal root ganglion neurons. CB-13 reduced inflammation-induced mechanical allodynia in male and female mice in a peripheral CB 1 -receptor-dependent manner and relieved inflammatory thermal hyperalgesia. In cultured mouse dorsal root ganglion neurons, CB-13 reduced TRPV1 sensitization and neuronal hyperexcitability induced by the inflammatory mediator prostaglandin E 2 , providing potential mechanistic explanations for the analgesic actions of peripheral CB 1 receptor activation. With acute dosing, phenotypes associated with central CB 1 receptor activation occurred only at a dose of CB-13 approximately 10-fold the ED 50 for reducing allodynia. Strikingly, repeated dosing resulted in both analgesic tolerance and CB 1 receptor dependence, even at a dose that did not produce central CB 1 -receptor-mediated phenotypes on acute dosing. This suggests that repeated CB-13 dosing leads to increased CNS exposure and unwanted engagement of central CB 1 receptors. Thus, caution is warranted regarding therapeutic use of CB-13 with the goal of avoiding CNS side effects. Nonetheless, the clear analgesic effect of acute peripheral CB 1 receptor activation suggests that peripherally restricted cannabinoids are a viable target for novel analgesic development.
Collapse
Affiliation(s)
- Richard A. Slivicki
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Jiwon Yi
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO
| | - Victoria E. Brings
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Phuong Nhu Huynh
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO
- Department of Neuroscience, Washington University, St. Louis, MO
- Department of Biomedical Engineering, Washington University, St. Louis, MO
| |
Collapse
|
10
|
Mechanisms of bone pain: Progress in research from bench to bedside. Bone Res 2022; 10:44. [PMID: 35668080 PMCID: PMC9170780 DOI: 10.1038/s41413-022-00217-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/29/2022] [Accepted: 05/09/2022] [Indexed: 12/27/2022] Open
Abstract
AbstractThe field of research on pain originating from various bone diseases is expanding rapidly, with new mechanisms and targets asserting both peripheral and central sites of action. The scope of research is broadening from bone biology to neuroscience, neuroendocrinology, and immunology. In particular, the roles of primary sensory neurons and non-neuronal cells in the peripheral tissues as important targets for bone pain treatment are under extensive investigation in both pre-clinical and clinical settings. An understanding of the peripheral mechanisms underlying pain conditions associated with various bone diseases will aid in the appropriate application and development of optimal strategies for not only managing bone pain symptoms but also improving bone repairing and remodeling, which potentially cures the underlying etiology for long-term functional recovery. In this review, we focus on advances in important preclinical studies of significant bone pain conditions in the past 5 years that indicated new peripheral neuronal and non-neuronal mechanisms, novel targets for potential clinical interventions, and future directions of research.
Collapse
|
11
|
The Peripheral Cannabinoid Receptor Type 1 (CB 1) as a Molecular Target for Modulating Body Weight in Man. Molecules 2021; 26:molecules26206178. [PMID: 34684760 PMCID: PMC8538448 DOI: 10.3390/molecules26206178] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/06/2021] [Accepted: 10/09/2021] [Indexed: 01/14/2023] Open
Abstract
The cannabinoid 1 (CB1) receptor regulates appetite and body weight; however, unwanted central side effects of both agonists (in wasting disorders) or antagonists (in obesity and diabetes) have limited their therapeutic utility. At the peripheral level, CB1 receptor activation impacts the energy balance of mammals in a number of different ways: inhibiting satiety and emesis, increasing food intake, altering adipokine and satiety hormone levels, altering taste sensation, decreasing lipolysis (fat break down), and increasing lipogenesis (fat generation). The CB1 receptor also plays an important role in the gut–brain axis control of appetite and satiety. The combined effect of peripheral CB1 activation is to promote appetite, energy storage, and energy preservation (and the opposite is true for CB1 antagonists). Therefore, the next generation of CB1 receptor medicines (agonists and antagonists, and indirect modulators of the endocannabinoid system) have been peripherally restricted to mitigate these issues, and some of these are already in clinical stage development. These compounds also have demonstrated potential in other conditions such as alcoholic steatohepatitis and diabetic nephropathy (peripherally restricted CB1 antagonists) and pain conditions (peripherally restricted CB1 agonists and FAAH inhibitors). This review will discuss the mechanisms by which peripheral CB1 receptors regulate body weight, and the therapeutic utility of peripherally restricted drugs in the management of body weight and beyond.
Collapse
|
12
|
Lowe H, Toyang N, Steele B, Bryant J, Ngwa W. The Endocannabinoid System: A Potential Target for the Treatment of Various Diseases. Int J Mol Sci 2021; 22:9472. [PMID: 34502379 PMCID: PMC8430969 DOI: 10.3390/ijms22179472] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/23/2021] [Accepted: 08/26/2021] [Indexed: 02/06/2023] Open
Abstract
The Endocannabinoid System (ECS) is primarily responsible for maintaining homeostasis, a balance in internal environment (temperature, mood, and immune system) and energy input and output in living, biological systems. In addition to regulating physiological processes, the ECS directly influences anxiety, feeding behaviour/appetite, emotional behaviour, depression, nervous functions, neurogenesis, neuroprotection, reward, cognition, learning, memory, pain sensation, fertility, pregnancy, and pre-and post-natal development. The ECS is also involved in several pathophysiological diseases such as cancer, cardiovascular diseases, and neurodegenerative diseases. In recent years, genetic and pharmacological manipulation of the ECS has gained significant interest in medicine, research, and drug discovery and development. The distribution of the components of the ECS system throughout the body, and the physiological/pathophysiological role of the ECS-signalling pathways in many diseases, all offer promising opportunities for the development of novel cannabinergic, cannabimimetic, and cannabinoid-based therapeutic drugs that genetically or pharmacologically modulate the ECS via inhibition of metabolic pathways and/or agonism or antagonism of the receptors of the ECS. This modulation results in the differential expression/activity of the components of the ECS that may be beneficial in the treatment of a number of diseases. This manuscript in-depth review will investigate the potential of the ECS in the treatment of various diseases, and to put forth the suggestion that many of these secondary metabolites of Cannabis sativa L. (hereafter referred to as "C. sativa L." or "medical cannabis"), may also have potential as lead compounds in the development of cannabinoid-based pharmaceuticals for a variety of diseases.
Collapse
Affiliation(s)
- Henry Lowe
- Biotech R & D Institute, University of the West Indies, Mona 99999, Jamaica; (H.L.); (J.B.)
- Vilotos Pharmaceuticals Inc., Baltimore, MD 21202, USA;
- Flavocure Biotech Inc., Baltimore, MD 21202, USA
- Department of Medicine, University of Maryland Medical School, Baltimore, MD 21202, USA
| | - Ngeh Toyang
- Vilotos Pharmaceuticals Inc., Baltimore, MD 21202, USA;
- Flavocure Biotech Inc., Baltimore, MD 21202, USA
| | - Blair Steele
- Biotech R & D Institute, University of the West Indies, Mona 99999, Jamaica; (H.L.); (J.B.)
| | - Joseph Bryant
- Biotech R & D Institute, University of the West Indies, Mona 99999, Jamaica; (H.L.); (J.B.)
| | - Wilfred Ngwa
- Brigham and Women’s Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02215, USA;
- Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| |
Collapse
|
13
|
Yamamoto T, Mulpuri Y, Izraylev M, Li Q, Simonian M, Kramme C, Schmidt BL, Seltzman HH, Spigelman I. Selective targeting of peripheral cannabinoid receptors prevents behavioral symptoms and sensitization of trigeminal neurons in mouse models of migraine and medication overuse headache. Pain 2021; 162:2246-2262. [PMID: 33534356 PMCID: PMC8277668 DOI: 10.1097/j.pain.0000000000002214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 01/19/2021] [Indexed: 01/03/2023]
Abstract
ABSTRACT Migraine affects ∼15% of the world's population greatly diminishing their quality of life. Current preventative treatments are effective in only a subset of migraine patients, and although cannabinoids seem beneficial in alleviating migraine symptoms, central nervous system side effects limit their widespread use. We developed peripherally restricted cannabinoids (PRCBs) that relieve chronic pain symptoms of cancer and neuropathies, without appreciable central nervous system side effects or tolerance development. Here, we determined PRCB effectiveness in alleviating hypersensitivity symptoms in mouse models of migraine and medication overuse headache. Long-term glyceryl trinitrate (GTN, 10 mg/kg) administration led to increased sensitivity to mechanical stimuli and increased expression of phosphorylated protein kinase A, neuronal nitric oxide synthase, and transient receptor potential ankyrin 1 proteins in trigeminal ganglia. Peripherally restricted cannabinoid pretreatment, but not posttreatment, prevented behavioral and biochemical correlates of GTN-induced sensitization. Low pH-activated and allyl isothiocyanate-activated currents in acutely isolated trigeminal neurons were reversibly attenuated by PRCB application. Long-term GTN treatment significantly enhanced these currents. Long-term sumatriptan treatment also led to the development of allodynia to mechanical and cold stimuli that was slowly reversible after sumatriptan discontinuation. Subsequent challenge with a previously ineffective low-dose GTN (0.1-0.3 mg/kg) revealed latent behavioral sensitization and increased expression of phosphorylated protein kinase A, neuronal nitric oxide synthase, and transient receptor potential ankyrin 1 proteins in trigeminal ganglia. Peripherally restricted cannabinoid pretreatment prevented all behavioral and biochemical correlates of allodynia and latent sensitization. Importantly, long-term PRCB treatment alone did not produce any behavioral or biochemical signs of sensitization. These data validate peripheral cannabinoid receptors as potential therapeutic targets in migraine and medication overuse headache.
Collapse
Affiliation(s)
- Toru Yamamoto
- Division of Oral Biology & Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Yatendra Mulpuri
- Division of Oral Biology & Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Mikhail Izraylev
- Division of Oral Biology & Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Qianyi Li
- Division of Oral Biology & Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Menooa Simonian
- Division of Oral Biology & Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Christian Kramme
- Division of Oral Biology & Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
| | - Brian L. Schmidt
- Department of Oral & Maxillofacial Surgery and Bluestone Center for Clinical Research, New York University College of Dentistry, New York, NY
| | - Herbert H. Seltzman
- Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC
| | - Igor Spigelman
- Division of Oral Biology & Medicine, School of Dentistry, University of California, Los Angeles, Los Angeles, CA
- Brain Research Institute, University of California, Los Angeles, Los Angeles, CA
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW The treatment of cancer-induced bone pain (CIBP) has been proven ineffective and relies heavily on opioids, the target of highly visible criticism for their negative side effects. Alternative therapeutic agents are needed and the last few years have brought promising results, detailed in this review. RECENT FINDINGS Cysteine/glutamate antiporter system, xc, cannabinoids, kappa opioids, and a ceramide axis have all been shown to have potential as novel therapeutic targets without the negative effects of opioids. SUMMARY Review of the most recent and promising studies involving CIBP, specifically within murine models. Cancer pain has been reported by 30-50% of all cancer patients and even more in late stages, however the standard of care is not effective to treat CIBP. The complicated and chronic nature of this type of pain response renders over the counter analgesics and opioids largely ineffective as well as difficult to use due to unwanted side effects. Preclinical studies have been standardized and replicated while novel treatments have been explored utilizing various alternative receptor pathways: cysteine/glutamate antiporter system, xc, cannabinoid type 1 receptor, kappa opioids, and a ceramide axis sphingosine-1-phosphate/sphingosine-1-phosphate receptor 1.
Collapse
|
15
|
Lin X, Chen W, Wei F. Technique Success, Technique Efficacy and Complications of HIFU Ablation for Palliation of Pain in Patients With Bone Lesions: A Meta-Analysis of 28 Feasibility Studies. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1182-1191. [PMID: 33583637 DOI: 10.1016/j.ultrasmedbio.2021.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 01/13/2021] [Accepted: 01/16/2021] [Indexed: 06/12/2023]
Abstract
Several feasibility studies have reported that high-intensity focused ultrasound (HIFU) ablation can be applied to ease patients' bone pain. However, the effect of HIFU ablation to palliate bone pain remains unclear. To evaluate the technique's success, efficacy, minor complication and major complication on patients suffering from bone pain, we searched the PubMed, MEDLINE, EMBASE and Cochrane Library databases from January 1998 to March 2019. Clinical studies that have assessed the association between bone pain and HIFU ablation were involved. We filtered out 28 feasibility studies, which reported the association between HIFU ablation and bone pain, including a total of 717 patients and 736 bone lesions. Overall, our results indicate that the rate of technique success of HIFU ablation was 93% (95% confidence interval [CI] 89%-96%) for patients with bone lesions. The technique efficacy rate of HIFU ablation for palliation of pain from bone lesions was 80% (95% CI 74%-87%) in all the patients, 96% (91%-100%) in the subgroup of retrospective studies and 77% (69%-85%) in the subgroup of prospective studies. In regard to HIFU ablation treatment safety, the hazard ratio for minor complication was 12% (95% CI 7%-17%), and the hazard ratio for major complication was 2% (95% CI 1%-3%). In conclusion, the summary rates for various considerations of using HIFU ablation for the palliation of bone pain are as follows: technique success is 93%, technique efficacy is 77%, minor complication is 12% and major complication is 2%. Our results suggest that extracorporeal HIFU ablation is a promising method for palliation of pain in bone lesions, with high technique success and efficacy, but low adverse events.
Collapse
Affiliation(s)
- Xiaoti Lin
- Department of Breast, Fujian Provincial Maternity and Children's Hospital, Fujian Medical University, Fuzhou, China; Department of Breast Oncology, State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Weiyu Chen
- Department of Physiology, Zhongshan Medical School, Sun Yat-sen University, Guangzhou, China
| | - Fengqin Wei
- Department of Emergency, Fujian Provincial 2nd People's Hospital, Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
16
|
Saldaña-Shumaker SL, Grenning AJ, Cunningham CW. Modern approaches to the development of synthetic cannabinoid receptor probes. Pharmacol Biochem Behav 2021; 203:173119. [PMID: 33508249 DOI: 10.1016/j.pbb.2021.173119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 10/13/2020] [Accepted: 01/18/2021] [Indexed: 10/22/2022]
Abstract
The endocannabinoid system, which spans the central and peripheral nervous systems and regulates many biologic processes, is an important target for probe discovery and medications development. Whereas the earliest endocannabinoid receptor probes were derivatives of the non-selective phytocannabinoids isolated from Cannabis species, modern drug discovery techniques have expanded the definitions of what constitutes a CB1R or CB2R cannabinoid receptor ligand. This review highlights recent advances in synthetic cannabinoid receptor chemistry and pharmacology. We provide examples of new CB1R- and CB2R-selective probes, and discuss rational approaches to the design of peripherally-restricted agents. We also describe structural classes of positive- and negative allosteric modulators (PAMs and NAMs) of CB1R and CB2R. Finally, we introduce new opportunities for cannabinoid receptor probe development that have emerged in recent years, including biased agonists that may lead to medications lacking adverse effects.
Collapse
Affiliation(s)
- Savanah L Saldaña-Shumaker
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N. Lake Shore Drive, Mequon, WI 53097, USA
| | - Alexander J Grenning
- Department of Chemistry, University of Florida, PO Box 117200, Gainesville, FL 32611, USA
| | - Christopher W Cunningham
- Department of Pharmaceutical Sciences, Concordia University Wisconsin, 12800 N. Lake Shore Drive, Mequon, WI 53097, USA.
| |
Collapse
|
17
|
Kim TE, Townsend RK, Branch CL, Romero-Sandoval EA, Hsu W. Cannabinoids in the Treatment of Back Pain. Neurosurgery 2021; 87:166-175. [PMID: 32097466 DOI: 10.1093/neuros/nyz573] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 11/30/2019] [Indexed: 01/17/2023] Open
Abstract
Marijuana is increasingly utilized for the treatment of multiple medical problems, including back pain, in the United States. Although there is strong preclinical evidence supporting the promise of cannabinoids in the treatment of back pain, there is a paucity of clinical data supporting their use in clinical practice. Opioids are an important medication for the treatment of acute and chronic back pain, but utilization of opioid-based regimens have likely contributed to the growing opioid epidemic. The significant risk of morbidity, mortality, and dependence secondary to opioid medications have increased the interest in nonopioid medications, including cannabinoid-based pain regimens, in treating back pain. This review will provide an overview on the pharmacology, drug delivery methods, clinical evidence, and safety considerations critical to understanding the potential role of cannabinoids in the treatment of back pain.
Collapse
Affiliation(s)
- Teddy E Kim
- Department of Neurosurgery, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Robert K Townsend
- Department of Neurosurgery, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Charles L Branch
- Department of Neurosurgery, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Edgar A Romero-Sandoval
- Department of Anesthesiology, Wake Forest University School of Medicine, Winston Salem, North Carolina
| | - Wesley Hsu
- Department of Neurosurgery, Wake Forest University School of Medicine, Winston Salem, North Carolina
| |
Collapse
|
18
|
Pineda-Farias JB, Saloman JL, Scheff NN. Animal Models of Cancer-Related Pain: Current Perspectives in Translation. Front Pharmacol 2021; 11:610894. [PMID: 33381048 PMCID: PMC7768910 DOI: 10.3389/fphar.2020.610894] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 10/30/2020] [Indexed: 01/15/2023] Open
Abstract
The incidence of pain in cancer patients during diagnosis and treatment is exceedingly high. Although advances in cancer detection and therapy have improved patient prognosis, cancer and its treatment-associated pain have gained clinical prominence. The biological mechanisms involved in cancer-related pain are multifactorial; different processes for pain may be responsible depending on the type and anatomic location of cancer. Animal models of cancer-related pain have provided mechanistic insights into the development and process of pain under a dynamic molecular environment. However, while cancer-evoked nociceptive responses in animals reflect some of the patients’ symptoms, the current models have failed to address the complexity of interactions within the natural disease state. Although there has been a recent convergence of the investigation of carcinogenesis and pain neurobiology, identification of new targets for novel therapies to treat cancer-related pain requires standardization of methodologies within the cancer pain field as well as across disciplines. Limited success of translation from preclinical studies to the clinic may be due to our poor understanding of the crosstalk between cancer cells and their microenvironment (e.g., sensory neurons, infiltrating immune cells, stromal cells etc.). This relatively new line of inquiry also highlights the broader limitations in translatability and interpretation of basic cancer pain research. The goal of this review is to summarize recent findings in cancer pain based on preclinical animal models, discuss the translational benefit of these discoveries, and propose considerations for future translational models of cancer pain.
Collapse
Affiliation(s)
- Jorge B Pineda-Farias
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Jami L Saloman
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Nicole N Scheff
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States.,Hillman Cancer Center, University of Pittsburgh Medicine Center, Pittsburgh, PA, United States
| |
Collapse
|
19
|
Takizawa M, Cerneus D, Michon I, Rijnders S, van der Heide D, Meijer J, Stoelzel M, Sato Y. The Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of ASP3652 in First-in-Human and Ascending Multiple Oral Dose Studies in Healthy Subjects. Adv Ther 2020; 37:3878-3900. [PMID: 32681461 DOI: 10.1007/s12325-020-01402-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Inhibitors of fatty acid amide hydrolase (FAAH) increase the levels of endocannabinoids and have shown analgesic and anti-inflammatory activity in animal models. ASP3652 is a peripherally acting FAAH inhibitor in development for the treatment of chronic bladder and pelvic pain disorders. Here we describe the safety, pharmacokinetics, and pharmacodynamics of single and multiple oral doses of ASP3652 administered in healthy non-elderly and elderly male and female volunteers. METHODS Study 1 was a combined single-ascending dose and food-effect study in which ASP3652 was given as single doses (1-600 mg) or matching placebo in healthy subjects. Study 2 was a multiple ascending dose study in which ASP3652 or matching placebo was administered in multiple oral doses (10-300 mg bid and 600 mg qd for 14 days) to healthy subjects. In both studies, the levels of ASP3652, FAAH, endocannabinoids (eCBs) and safety were evaluated. RESULTS ASP3652 was readily absorbed to reach Cmax at 1 h after a single dose. Steady state was reached within 3 days after the start of multiple dosing. The Cmax and AUC of ASP3652 increased in a slightly more than dose-proportional manner after a single dose of ASP3652 at 30-600 mg. There was some accumulation (15-38%) based on Cmax and AUC12h upon multiple doses. Cmax was 47% lower in combination with food. There was no significant effect of gender or age on the pharmacokinetics of ASP3652. FAAH activity was inhibited in a dose-dependent manner in all dose groups after single and multiple doses of ASP3652, paralleled by an increase in plasma levels of anandamide (AEA). The incidence of adverse events following multiple doses was similar across all treatment groups including the placebo group. CONCLUSIONS Single and multiple doses of ASP3652 were safe and well tolerated and increased endogenous cannabinoid plasma levels.
Collapse
Affiliation(s)
| | - Dirk Cerneus
- Astellas Pharma Europe B. V., Leiden, The Netherlands
| | - Ingrid Michon
- Astellas Pharma Europe B. V., Leiden, The Netherlands
| | | | | | - John Meijer
- Astellas Pharma Europe B. V., Leiden, The Netherlands
| | | | | |
Collapse
|
20
|
Morales P, Jagerovic N. Novel approaches and current challenges with targeting the endocannabinoid system. Expert Opin Drug Discov 2020; 15:917-930. [PMID: 32336154 PMCID: PMC7502221 DOI: 10.1080/17460441.2020.1752178] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/02/2020] [Indexed: 12/12/2022]
Abstract
INTRODUCTION The pathophysiological relevance of the endocannabinoid system has been widely demonstrated in a variety of diseases including cancer, neurological disorders, and metabolic issues. Therefore, targeting the receptors and the endogenous machinery involved in this system can provide a successful therapeutic outcome. Ligands targeting the canonical cannabinoid receptors, CB1 and CB2, along with inhibitors of the endocannabinoid enzymes have been thoroughly studied in diverse disease models. In fact, phytocannabinoids such as cannabidiol or Δ9-tetrahydrocannabinol are currently on the market for the management of neuropathic pain due to spasticity in multiple sclerosis or seizures in children epilepsy amongst others. AREAS COVERED Challenges in the pharmacology of cannabinoids arise from its pharmacokinetics, off-target effects, and psychoactive effects. In this context, the current review outlines the novel molecular approaches emerging in the field discussing their clinical potential. EXPERT OPINION Even if orthosteric CB1 and CB2 ligands are on the forefront in cannabinoid clinical research, emerging strategies such as allosteric or biased modulation of these receptors along with controlled off-targets effects may increase the therapeutic potential of cannabinoids.
Collapse
Affiliation(s)
- Paula Morales
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Nadine Jagerovic
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
21
|
Lost in the Haze: The Physician's Role in Cannabinoid Prescribing and Advising. Am J Med 2020; 133:7-8. [PMID: 31251905 DOI: 10.1016/j.amjmed.2019.05.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 11/21/2022]
|
22
|
Dichotomic effects of clinically used drugs on tumor growth, bone remodeling and pain management. Sci Rep 2019; 9:20155. [PMID: 31882872 PMCID: PMC6934511 DOI: 10.1038/s41598-019-56622-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 12/12/2019] [Indexed: 11/08/2022] Open
Abstract
Improvements in the survival of breast cancer patients have led to the emergence of bone health and pain management as key aspects of patient’s quality of life. Here, we used a female rat MRMT-1 model of breast cancer-induced bone pain to compare the effects of three drugs used clinically morphine, nabilone and zoledronate on tumor progression, bone remodeling and pain relief. We found that chronic morphine reduced the mechanical hypersensitivity induced by the proliferation of the luminal B aggressive breast cancer cells in the tumor-bearing femur and prevented spinal neuronal and astrocyte activation. Using MTT cell viability assay and MRI coupled to 18FDG PET imaging followed by ex vivo 3D µCT, we further demonstrated that morphine did not directly exert tumor growth promoting or inhibiting effects on MRMT-1 cancer cells but induced detrimental effects on bone healing by disturbing the balance between bone formation and breakdown. In sharp contrast, both the FDA-approved bisphosphonate zoledronate and the synthetic cannabinoid nabilone prescribed as antiemetics to patients receiving chemotherapy were effective in limiting the osteolytic bone destruction, thus preserving the bone architecture. The protective effect of nabilone on bone metabolism was further accompanied by a direct inhibition of tumor growth. As opposed to zoledronate, nabilone was however not able to manage bone tumor-induced pain and reactive gliosis. Altogether, our results revealed that morphine, nabilone and zoledronate exert disparate effects on tumor growth, bone metabolism and pain control. These findings also support the use of nabilone as an adjuvant therapy for bone metastases.
Collapse
|
23
|
de Oliveira HU, Dos Santos RS, Malta IHS, Pinho JP, Almeida AFS, Sorgi CA, Peti APF, Xavier GS, Reis LMD, Faccioli LH, Cruz JDS, Ferreira E, Galdino G. Investigation of the Involvement of the Endocannabinoid System in TENS-Induced Antinociception. THE JOURNAL OF PAIN 2019; 21:820-835. [PMID: 31785404 DOI: 10.1016/j.jpain.2019.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 08/16/2019] [Accepted: 11/11/2019] [Indexed: 01/14/2023]
Abstract
Transcutaneous electrical nerve stimulation (TENS) promotes antinociception by activating the descending pain modulation pathway and consequently releasing endogenous analgesic substances. In addition, recent studies have shown that the endocannabinoid system controls pain. Thus, the present study investigated the involvement of the endocannabinoid system in TENS-induced antinociception of cancer pain using a cancer pain model induced by intraplantar (i.pl.) injections of Ehrlich tumor cells in male Swiss mice. Low- and high-frequency TENS was applied for 20 minutes to the mice's paws, and to investigate the involvement of the endocannabinoid system were used the N-(peperidin-1-yl)-5-(4-iodophenyl)-1-(2,4-dichlorophenyl)-4-methyl-1H-pitazole-3-carboixamide (AM251), a cannabinoid CB1 receptor antagonist and (5Z,8Z,11Z,14Z)-5,8,11,14-eicosatetraenyl-methylester phosphonofluoridic acid (MAFP), an inhibitor of the endocannabinoid metabolizing enzyme fatty acid amide hydrolase, injected by via i.pl., intrathecal (i.t.), and intradorsolateral periaqueductal gray matter (i.dl.PAG). Furthermore, liquid chromatography-tandem mass spectrometry, western blot, and immunofluorescence assays were used to evaluate the endocannabinoid anandamide levels, cannabinoid CB1 receptor protein levels, and cannabinoid CB1 receptor immunoreactivity, respectively. Low- and high-frequency TENS reduced the mechanical allodynia induced by Ehrlich tumor cells and this effect was reversed by AM251 and potentiated by MAFP at the peripheral and central levels. In addition, TENS increased the endocannabinoid anandamide levels and the cannabinoid CB1 receptor protein levels and immunoreactivity in the paw, spinal cord, and dorsolateral periaqueductal gray matter. These results suggest that low- and high-frequency TENS is effective in controlling cancer pain, and the endocannabinoid system is involved in this effect at both the peripheral and central levels. PERSPECTIVE: TENS is a nonpharmacological strategy that may be used to control cancer pain. Identification of a new mechanism involved in its analgesic effect could lead to the development of clinical studies as well as an increase in its application, lessening the need for pharmacological treatments.
Collapse
Affiliation(s)
| | | | | | - José Phellipe Pinho
- Department of Physiology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Carlos Arterio Sorgi
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, São Paulo Brazil
| | - Ana Paula Ferranti Peti
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, São Paulo Brazil
| | | | | | - Lúcia Helena Faccioli
- Faculty of Pharmaceutical Sciences of Ribeirão Preto, Universidade de São Paulo, São Paulo Brazil
| | - Jader Dos Santos Cruz
- Department of Physiology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Enio Ferreira
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Giovane Galdino
- Sciences of Motricity Institute, Federal University of Alfenas, Alfenas, Brazil.
| |
Collapse
|
24
|
The endocannabinoid system: Novel targets for treating cancer induced bone pain. Biomed Pharmacother 2019; 120:109504. [PMID: 31627091 DOI: 10.1016/j.biopha.2019.109504] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 09/16/2019] [Accepted: 09/26/2019] [Indexed: 02/08/2023] Open
Abstract
Treating Cancer-induced bone pain (CIBP) continues to be a major clinical challenge and underlying mechanisms of CIBP remain unclear. Recently, emerging body of evidence suggested the endocannabinoid system (ECS) may play essential roles in CIBP. Here, we summarized the current understanding of the antinociceptive mechanisms of endocannabinoids in CIBP and discussed the beneficial effects of endocannabinoid for CIBP treatment. Targeting non-selective cannabinoid 1 receptors or selective cannabinoid 2 receptors, and modulation of peripheral AEA and 2-AG, as well as the inhibition the function of fatty acid amide hydrolase (FAAH) and monoacylglycerol lipase (MAGL) have produced analgesic effects in animal models of CIBP. Management of ECS therefore appears to be a promising way for the treatment of CIBP in terms of efficacy and safety. Further clinical studies are encouraged to confirm the possible translation to humans of the very promising results already obtained in the preclinical studies.
Collapse
|
25
|
Dvorácskó S, Keresztes A, Mollica A, Stefanucci A, Macedonio G, Pieretti S, Zádor F, Walter FR, Deli MA, Kékesi G, Bánki L, Tuboly G, Horváth G, Tömböly C. Preparation of bivalent agonists for targeting the mu opioid and cannabinoid receptors. Eur J Med Chem 2019; 178:571-588. [PMID: 31220675 DOI: 10.1016/j.ejmech.2019.05.037] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/30/2019] [Accepted: 05/12/2019] [Indexed: 11/17/2022]
Abstract
In order to obtain novel pharmacological tools and to investigate a multitargeting analgesic strategy, the CB1 and CB2 cannabinoid receptor agonist JWH-018 was conjugated with the opiate analgesic oxycodone or with an enkephalin related tetrapeptide. The opioid and cannabinoid pharmacophores were coupled via spacers of different length and chemical structure. In vitro radioligand binding experiments confirmed that the resulting bivalent compounds bound both to the opioid and to the cannabinoid receptors with moderate to high affinity. The highest affinity bivalent derivatives 11 and 19 exhibited agonist properties in [35S]GTPγS binding assays. These compounds activated MOR and CB (11 mainly CB2, whereas 19 mainly CB1) receptor-mediated signaling, as it was revealed by experiments using receptor specific antagonists. In rats both 11 and 19 exhibited antiallodynic effect similar to the parent drugs in 20 μg dose at spinal level. These results support the strategy of multitargeting G-protein coupled receptors to develop lead compounds with antinociceptive properties.
Collapse
MESH Headings
- Analgesics, Opioid/chemical synthesis
- Analgesics, Opioid/chemistry
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Enkephalins/chemistry
- Enkephalins/pharmacology
- Indoles/chemistry
- Indoles/pharmacology
- Mice
- Molecular Structure
- Naphthalenes/chemistry
- Naphthalenes/pharmacology
- Oxycodone/chemistry
- Oxycodone/pharmacology
- Rats
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/metabolism
- Receptors, Opioid, mu/antagonists & inhibitors
- Receptors, Opioid, mu/metabolism
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Szabolcs Dvorácskó
- A Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62., 6726, Szeged, Hungary
| | - Attila Keresztes
- A Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62., 6726, Szeged, Hungary
| | - Adriano Mollica
- Dipartimento di Farmacia, Università di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100, Chieti, Italy
| | - Azzurra Stefanucci
- Dipartimento di Farmacia, Università di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100, Chieti, Italy
| | - Giorgia Macedonio
- Dipartimento di Farmacia, Università di Chieti-Pescara "G. d'Annunzio", Via dei Vestini 31, 66100, Chieti, Italy
| | - Stefano Pieretti
- Istituto Superiore di Sanità, Centro Nazionale Ricerca e Valutazione Preclinica e Clinica dei Farmaci, Viale Regina Elena 299, 00161, Rome, Italy
| | - Ferenc Zádor
- Laboratory of Opioid Research, Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62., 6726, Szeged, Hungary
| | - Fruzsina R Walter
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62., 6726, Szeged, Hungary
| | - Mária A Deli
- Biological Barriers Research Group, Institute of Biophysics, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62., 6726, Szeged, Hungary
| | - Gabriella Kékesi
- Department of Physiology, Faculty of Medicine, University of Szeged, 6720, Szeged, Dóm tér 10., Hungary
| | - László Bánki
- Department of Traumatology, Faculty of Medicine, University of Szeged, 6725, Szeged, Semmelweis u. 6., Hungary
| | - Gábor Tuboly
- Department of Neurology, Faculty of Medicine, University of Szeged, 6725, Szeged, Semmelweis u. 6., Hungary
| | - Gyöngyi Horváth
- Department of Physiology, Faculty of Medicine, University of Szeged, 6720, Szeged, Dóm tér 10., Hungary
| | - Csaba Tömböly
- A Laboratory of Chemical Biology, Institute of Biochemistry, Biological Research Centre of the Hungarian Academy of Sciences, Temesvári krt. 62., 6726, Szeged, Hungary.
| |
Collapse
|
26
|
Abstract
Extracts from Cannabis species have aided the discovery of the endocannabinoid signaling system (ECSS) and phytocannabinoids that possess broad therapeutic potential. Whereas the reinforcing effects of C. sativa are largely attributed to CB1 receptor agonism by Δ9-tetrahydrocannabinol (Δ9-THC), the observed medicinal effects of Cannabis arise from the combined actions of various compounds. In addition to compounds bearing a classical cannabinoid structure, naturally occurring fatty acid amides and esters resembling anandamide and 2-arachidonoyl glycerol isolated from non- Cannabis species are also valuable tools for studying ECSS function. This review highlights the potential of plant-based secondary metabolites from Cannabis and unrelated species as ECSS modulators.
Collapse
Affiliation(s)
- Christopher W Cunningham
- Department of Pharmaceutical Sciences , Concordia University Wisconsin , Mequon , Wisconsin 53097 , United States
| |
Collapse
|
27
|
Abstract
Cannabis users have long reported therapeutic properties of the plant for a variety of conditions, some of which include nausea, emesis, seizures, cancer, neurogenic diseases and pain control. Research has elucidated many cannabinoid pharmacodynamic and pharmacokinetic properties, expanding the potential use of cannabinoids as a medical therapy. Due to the inconsistent delivery and control of the active components involved with smoking, pharmaceutical companies are investigating and prioritizing routes other than smoke inhalation for therapeutic use of cannabinoids. In this relatively new field of pharmaceutical development, ongoing drug development promises great benefit from targeted endocannabinoid receptor agonism. Available in Canada and Europe, nabiximols, a specific extract from the Cannabis plant, has demonstrated great benefit in the treatment of pain related to spasticity in multiple sclerosis, cancer and otherwise chronic pain conditions. The cannabidiol oral solution Epidiolex®, which is available in the USA, is indicated for management of refractory epilepsy but may offer therapeutic relief to chronic pain conditions as well. Current investigative drugs, such as those developed by Cara Therapeutics and Zynerba Pharmaceuticals, are synthetic cannabinoids which show promise to specifically target neuropsychiatric conditions and chronic pain symptoms such as neuropathy and allodynia. The objective of this review is to provide clinicians with an update of currently available and promising developmental cannabis pharmaceutical derivatives which may stand to greatly benefit patients with otherwise difficult-to-treat chronic conditions.
Collapse
|
28
|
Modulation of Opioid Transport at the Blood-Brain Barrier by Altered ATP-Binding Cassette (ABC) Transporter Expression and Activity. Pharmaceutics 2018; 10:pharmaceutics10040192. [PMID: 30340346 PMCID: PMC6321372 DOI: 10.3390/pharmaceutics10040192] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 10/14/2018] [Accepted: 10/16/2018] [Indexed: 12/23/2022] Open
Abstract
Opioids are highly effective analgesics that have a serious potential for adverse drug reactions and for development of addiction and tolerance. Since the use of opioids has escalated in recent years, it is increasingly important to understand biological mechanisms that can increase the probability of opioid-associated adverse events occurring in patient populations. This is emphasized by the current opioid epidemic in the United States where opioid analgesics are frequently abused and misused. It has been established that the effectiveness of opioids is maximized when these drugs readily access opioid receptors in the central nervous system (CNS). Indeed, opioid delivery to the brain is significantly influenced by the blood-brain barrier (BBB). In particular, ATP-binding cassette (ABC) transporters that are endogenously expressed at the BBB are critical determinants of CNS opioid penetration. In this review, we will discuss current knowledge on the transport of opioid analgesic drugs by ABC transporters at the BBB. We will also examine how expression and trafficking of ABC transporters can be modified by pain and/or opioid pharmacotherapy, a novel mechanism that can promote opioid-associated adverse drug events and development of addiction and tolerance.
Collapse
|
29
|
Mulpuri Y, Marty VN, Munier JJ, Mackie K, Schmidt BL, Seltzman HH, Spigelman I. Synthetic peripherally-restricted cannabinoid suppresses chemotherapy-induced peripheral neuropathy pain symptoms by CB1 receptor activation. Neuropharmacology 2018; 139:85-97. [PMID: 29981335 PMCID: PMC6883926 DOI: 10.1016/j.neuropharm.2018.07.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/04/2018] [Accepted: 07/02/2018] [Indexed: 01/19/2023]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a severe and dose-limiting side effect of cancer treatment that affects millions of cancer survivors throughout the world and current treatment options are extremely limited by their side effects. Cannabinoids are highly effective in suppressing pain symptoms of chemotherapy-induced and other peripheral neuropathies but their widespread use is limited by central nervous system (CNS)-mediated side effects. Here, we tested one compound from a series of recently developed synthetic peripherally restricted cannabinoids (PRCBs) in a rat model of cisplatin-induced peripheral neuropathy. Results show that local or systemic administration of 4-{2-[-(1E)-1[(4-propylnaphthalen-1-yl)methylidene]-1H-inden-3-yl]ethyl}morpholine (PrNMI) dose-dependently suppressed CIPN mechanical and cold allodynia. Orally administered PrNMI also dose-dependently suppressed CIPN allodynia symptoms in both male and female rats without any CNS side effects. Co-administration with selective cannabinoid receptor subtype blockers revealed that PrNMI's anti-allodynic effects are mediated by CB1 receptor (CB1R) activation. Expression of CB2Rs was reduced in dorsal root ganglia from CIPN rats, whereas expression of CB1Rs and various endocannabinoid synthesizing and metabolizing enzymes was unaffected. Daily PrNMI treatment of CIPN rats for two weeks showed a lack of appreciable tolerance to PrNMI's anti-allodynic effects. In an operant task which reflects cerebral processing of pain, PrNMI also dose-dependently suppressed CIPN pain behaviors. Our results demonstrate that PRCBs exemplified by PrNMI may represent a viable option for the treatment of CIPN pain symptoms.
Collapse
MESH Headings
- Analgesics, Non-Narcotic/administration & dosage
- Animals
- Antineoplastic Agents/adverse effects
- Cannabinoid Receptor Modulators/administration & dosage
- Cannabinoids/administration & dosage
- Cisplatin/adverse effects
- Cold Temperature
- Dose-Response Relationship, Drug
- Drug Tolerance
- Female
- Ganglia, Spinal/drug effects
- Ganglia, Spinal/metabolism
- Ganglia, Spinal/pathology
- Gene Expression Regulation/drug effects
- Hyperalgesia/drug therapy
- Hyperalgesia/metabolism
- Hyperalgesia/pathology
- Male
- Peripheral Nervous System Diseases/chemically induced
- Peripheral Nervous System Diseases/metabolism
- Peripheral Nervous System Diseases/pathology
- RNA, Messenger/metabolism
- Rats, Sprague-Dawley
- Receptor, Cannabinoid, CB1/antagonists & inhibitors
- Receptor, Cannabinoid, CB1/metabolism
- Receptor, Cannabinoid, CB2/antagonists & inhibitors
- Receptor, Cannabinoid, CB2/metabolism
- Touch
Collapse
Affiliation(s)
- Yatendra Mulpuri
- Laboratory of Neuropharmacology, Division of Oral Biology & Medicine, University of California, Los Angeles, CA, USA
| | - Vincent N Marty
- Laboratory of Neuropharmacology, Division of Oral Biology & Medicine, University of California, Los Angeles, CA, USA
| | - Joseph J Munier
- Laboratory of Neuropharmacology, Division of Oral Biology & Medicine, University of California, Los Angeles, CA, USA
| | - Ken Mackie
- Department of Psychological & Brain Sciences, Indiana University, Bloomington, IN, USA
| | - Brian L Schmidt
- Department of Oral & Maxillofacial Surgery and Bluestone Center for Clinical Research, New York University College of Dentistry, New York, NY, USA
| | - Herbert H Seltzman
- Organic and Medicinal Chemistry, Research Triangle Institute, Research Triangle Park, NC, USA
| | - Igor Spigelman
- Laboratory of Neuropharmacology, Division of Oral Biology & Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|