Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Jul 8.
Published in final edited form as: Crit Rev Ther Drug Carrier Syst. 2024;41(1):1–33. doi: 10.1615/CritRevTherDrugCarrierSyst.2022038592

Applications of Cannabinoids in Neuropathic Pain: An Updated Review

Peggy Arthur 1, Anil Kumar Kalvala 1, Sunil Kumar Surapaneni 1, Mandip Singh 1,*
PMCID: PMC11228808  NIHMSID: NIHMS1973009  PMID: 37824417

Abstract

Neuropathic pain is experienced due to injury to the nerves, underlying disease conditions or toxicity induced by chemotherapeutics. Multiple factors can contribute to neuropathic pain such as central nervous system (CNS)-related autoimmune and metabolic disorders, nerve injury, multiple sclerosis and diabetes. Hence, development of pharmacological interventions to reduce the drawbacks of existing chemotherapeutics and counter neuropathic pain is an urgent unmet clinical need. Cannabinoid treatment has been reported to be beneficial for several disease conditions including neuropathic pain. Cannabinoids act by inhibiting the release of neurotransmitters from presynaptic nerve endings, modulating the excitation of postsynaptic neurons, activating descending inhibitory pain pathways, reducing neural inflammation and oxidative stress and also correcting autophagy defects. This review provides insights on the various preclinical and clinical therapeutic applications of cannabidiol (CBD), cannabigerol (CBG), and cannabinol (CBN) in various diseases and the ongoing clinical trials for the treatment of chronic and acute pain with cannabinoids. Pharmacological and genetic experimental strategies have well demonstrated the potential neuroprotective effects of cannabinoids and also elaborated their mechanism of action for the therapy of neuropathic pain.

Keywords: cannabinoids, analgesia, pain, neuropathic pain

I. INTRODUCTION

Cannabis, a drug derived from the flowers and buds of the Cannabis sativa plant is used therapeutically for various diseases for centuries.1,2 Since 2700 B.C., it is used by the ancient Chinese people for various medical practices such as rheumatic pain, intestinal constipation, disorders of the female reproductive system and malaria.2 Cannabinoids are basically C21 terpenophenolic group of compounds present in Cannabis sativa L., which include their analogs and transformation products.3 However, cannabinoids in a broader sense refer to all cannabinoid receptor ligands and its related compounds including endogenous cannabinoid receptor ligands and synthetic cannabinoid analogs.4 Based on the source of derivation, these are classified into three types, which include (a) endocannabinoids (eCBs), endogenous animal derivatives of polyunsaturated fatty acids (PUFAs); (b) phytocannabinoids (pCBs), obtained from plants; and (c) synthetic cannabinoids. Majority of the phytocannabinoids are present in female flowers and in most aerial parts of the cannabis plants.5 DeltΔ9-tetrahydrocannabinol (Δ9-THC), deltΔ8-tetrahydrocannabinol (Δ8-THC), and cannabinol (CBN) are examples of psychoactive cannabinoids found in cannabis plants. Cannabichromene (CBC), cannabidiol (CBD), and cannabigerol (CBG) are common non-psychoactive pCBs present in cannabis plants.6 Many subclasses of cannabinoids have been identified, which includes CBG, the first cannabinoid to be known; cannabigerolic acid (CBGA; carboxylic acid form of CBG), the earliest known biosynthetic cannabinoid produced in cannabis plants; CBC: five CBC type cannabinoids are known mostly with C5 and a few with C3 side chains; CBD: seven CBD-type cannabinoids have been identified containing Cl, C3, C4, or C5 side chains among which C5 is prevalent; Δ9-THC and Δ8-THC types. CBG, a non-psychoactive phytocannabinoid is present in high concentrations in Cannabis sativa. CBGA is the precursor of other essential phytocannabinoids.7 CBN and cannabinodiol (CBND) are aromatized derivatives (Fig. 1), which are formed due to air oxidation of THC and CBD, respectively.4 THCV, a naturally occurring analog of THC is non-psychoactive, neutral CB1 antagonist and functions as an agonist/antagonist of CB2 receptors depending on its dose. However, THC is psychoactive and acts as a CB1 and CB2 receptor agonist. THCV unlike THC causes hypophagic effects under fasting and non-fasting in mice.8 Over 150 unique terpenes and 100 different cannabinoids have been identified from various types of cannabis resins.9 Monoterpenes such as myrcene, and sesquiterpenes such as β-caryophyllene, α-humulene, bisabolo, and β-famesene are the common terpenes found in most cannabis strains.10

FIG. 1:

FIG. 1:

Chemical structures of some phytocannabinoids and synthetic cannabinoids

A. The Endocannabinoid System

Each of the numerous active cannabinoids identified in Cannabis sativa can modulate the body’s endocannabinoid system (ECS).11 Endocannabinoids are produced naturally in the human body, which act as lipid mediators by binding to cannabinoid receptors. ECS is made up of endogenous ligands, cannabinoid receptors and the enzymes responsible for their synthesis and degradation.11,12 It plays a vital role in the regulation of numerous cognitive and physiological processes of the CNS such as pain, mood, appetite, stress, and memory.1214 ECS also regulate physiological processes in the bone, reproductive system, heart, gastrointestinal tract and many other systems.1519 Phytocannabinoids are exogenous ligands for numerous receptors of the ECS.19 Many cannabinoids which are structurally and functionally analogous to the endocannabinoids have been synthesized.2022 CB1 and CB2 are the two main isolated endocannabinoid receptors.20 CB121,22 and CB222 receptors belong to the rhodopsin (Rho) family (class A) of G-protein coupled receptors (GPCRs). CB1 receptors are mainly found on the neural tissue consisting of the enteric nervous system and central and peripheral neurons.23 CB2 receptors are mostly present in immune tissues such as T and B cells, neutrophils, epithelial cells, macrophages as well as in the CNS.23,24 Figure 2 shows the distribution of the CB1 and CB2 in the human pain system. Generally, CB1 and CB2 receptors signaling are involved in the regulation of neuromodulatory and immunomodulatory functions respectively. CBG shows low binding affinity for CB1 and CB2 receptors.2527 ECS is affected due to the ability of CBG to inhibit anandamide (AEA) uptake.28 In vitro studies demonstrate that CBD shows weak antagonistic effects towards CB1 and CB2 receptors29,30 despite the findings of several reports showing its lack of affinity for these receptors.31

FIG. 2:

FIG. 2:

Localization of the cannabinoid receptors in the pain perception system of humans. The cannabinoid receptor 1 (CB1) is highly expressed in the peripheral nervous system (PNS) such as the sympathetic nerve terminal and in the brain (CNS). They are also observed in the dorsal root ganglion, dermic nerve endings of the primary sensory neurons. Cannabinoid receptor 2 (CB2) is less abundant in the CNS and PNS as compared to CB1.

B. Applications of Cannabinoids

Figure 3 summarizes the various applications of cannabinoids in medicine. Several studies have shown that cannabinoids have clinical applications in various diseases which are as follows.

FIG. 3:

FIG. 3:

Therapeutic applications of cannabinoids

1. Inflammatory Bowel Disease and Digestive Disorders

Inflammatory bowel diseases (IBD) are chronic immune-mediated diseases which include ulcerative colitis (UC) and Crohn’s disease (CD).32 Symptoms of IBD include periods of inflammatory flares, quiescence, and relapse, which affects the patient both psychologically and emotionally.33 The ECS, present throughout the gastrointestinal tract has been shown to play a role in regulating food intake, emesis, gastric secretion, visceral sensation and intestinal inflammation.34 Animal studies have shown that CB1 receptor agonists reduces gastrointestinal propulsion and transit.35,36 A study by Borrelli et al. has shown the benefits of CBG in the treatment of inflammatory bowel disease using a murine colitis model by the administration of dinitrobenzene sulphonic acid (DNBS) intracolonically. CBG reduced murine colitis, the macrophage production of nitric oxide and ROS production in intestinal epithelial cells.37

2. Cancer and Tumors

Inflammation, apoptosis and antioxidant defensive mechanisms play a pivotal role in progression of various cancers.3842 There are several ongoing research into nanodrug delivery systems (NDDSs) such as liposomes to improve the safety and efficacy of chemotherapeutic agents.4244 The use of immunotoxins for cancer treatment have also been well explored.42,45 CBG has been shown to bind to specific targets, which play a role in the prevention of various cancers. It is reported to be effective in xenograft models of colon cancer by acting as a TRPM8 antagonist, increasing the production of reactive oxygen species (ROS) and activating apoptotic pathways.46 Cannabinoids administered through inhalation route in combination with nanomaterials have shown good efficacy in lung cancer.47 Moreover, intratumoral administration of low doses of cannabinoids can also enhance the effectiveness of chemotherapeutics.4850 Combination of cannabinoids and radiotherapy also showed good anti-cancer efficacy in glioma and pancreatic cancer.51 Common side effects of cytotoxic chemotherapy such as muscle wasting, anorexia, and metabolic dysregulation pose dose-limiting effect on the efficacy of cannabinoids, which affects the quality of life and mortality. Cannabis sativa extracts and analogues of the major phytocannabinoid, Δ9-THC have been used to treat chemotherapy-induced appetite loss and nausea, but psychoactive side effects limit their clinical usage. In a study by Brierley et al., CBG has been shown to stimulate appetite in healthy rats without neuromotor side effects in an acute cachectic phenotype model induced by cisplatin (6 mg/kg i.p.) in male rats.52 CBG (120 mg/kg) modestly increased food intake, mainly at 36–60 h, and strongly attenuated cisplatin-induced weight loss from 6.3% to 2.6% at 72 h, demonstrating the potential of CBG for the treatment of chemotherapy-induced cachexia.52 An in vitro study by Zhang et al. has demonstrated that CBD inhibits the proliferation and colony formation of human gastric cancer (SGC-7901) cells,53 during which an upregulation in the expression of p53 and ataxia telangiectasia-mutated gene (ATM) as well as a downregulation in the expression of p21 protein are observed after CBD treatment. CBD was shown to increase Bax expression and decrease Bcl-2 expression levels, which eventually results in upregulation of cleaved caspase-3 and 9, finally leading to apoptosis.53 CBD increases intracellular reactive oxygen species levels and causes cell cycle arrest at the G0-G1 phase in gastric cancer cells. CBD has been reported to induce apoptosis through various mechanisms including caspase activation and ROS formation in human glioma (U87 and U373) cells.54 The growth of C6 glioma cells was inhibited when co-cultured with CBD and tamoxifen.55 Several reports have shown the ability of CBD to inhibit tumor cell proliferation and metastasis by inducing autophagy or apoptosis.56,57

Non-psychoactive phytocannabinoids (pCBs) derived from Cannabis sativa could be a novel therapeutic option for treatment of cachexia due to their pleiotropic pharmacological activities such as appetite stimulation. A study in rats by Brierley et al. has shown that purified CBG is an appetite stimulant.58 The effects of CBG-rich botanical drug substance (BDS), a potent non-psychoactive extract of Cannabis on feeding behavior was investigated and an increase in the total food intake was observed with CBG-BDS treatment group. CBG-BDS was concluded to be a novel appetite stimulant, which may be more potent than purified CBG and is devoid of psychoactive Δ9-tetrahydrocannabinol.58

3. Ocular

A study by Colansanti et al. has demonstrated that topical chronic administration of CBN/CBG using osmotic minipumps induced reduction of intraocular pressure (IOP) in cats. However, CBN unlike CBG caused conjunctival erythema and hyperemia.59 Different concentrations of CBN and CBG were given as a single drop or chronically using osmotic minipumps for 9 days. A single dose of CBN showed moderate effect on intraocular pressure after a single dose and it caused a more significant reduction in ocular tension during chronic administration. Although CBG had similar effects, the response was greater than CBN. A pilot study by Tomida et al. has assessed the effects of low doses of CBD and Δ−9-THC on intraocular pressure (IOP) when administered through sublingual route. In this study, various concentrations of CBD and Δ−9-THC were given to six patients with ocular hypertension or early primary open angle glaucoma. IOP was temporarily reduced by a single 5 mg sublingual dose of Δ−9-THC. However, 20 mg CBD did not show reduction in IOP, whereas there was a transient increase in IOP when 40 mg CBD was administered sublingually.60

4. Obesity and Metabolic Disorders

Fellous et al. explored the potential therapeutic effects of Cannabis sativa plant (or phytocannabinoids) derived non-euphoric compounds including tetrahydrocannabivarin (THCV), CBD, CBG, cannabidiolic acid (CBDA) and CBGA on the growth of endogenous BM-MSCs and the alterations on cellular differentiation into mature adipocytes.61 In vitro and in vivo experiments revealed that 5 μM of CBD, CBDA, CBGA and THCV increase the viability of BM-MSCs. CBG and CBD either alone or in combination stimulate BM-MSCs to mature into adipocytes via distinct molecular mechanisms. Phytocannabinoids further prevented the palmitate-induced insulin signaling impairment in adipocytes differentiated from BM-MSCs. Phytocannabinoids were suggested as a potential novel pharmacological tool to regain control of the functionality of adipose tissue in metabolic disorders such as type 2 diabetes mellitus (T2DM) and lipodystrophy.61

5. Skin Disorders

All the classes of cannabinoids interact with the skin cannabinoid receptors and regulate the pathways, which affect the homeostasis and metabolism of skin appendages and cutaneous cells.62 Due to the ability of phytocannabinoids to inhibit keratinocytes proliferation and modulate even the associated inflammatory components, phytocannabinoids serve as a potential treatment for psoriasis.63 A patent was filed in 2019 on topical CBD and CBG treatment for psoriasis. CBD and CBG have been shown to be effective in trail subjects in a dose-dependent manner by inhibiting inflammatory cytokines and angiogenic growth factors.64 A study in mice has shown that topical administration of phytocannabinoids effectively alleviate 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced erythema.65 Several in vitro and in vivo experiments have reported that phytocannabinoids inhibit sebocytes proliferation and decrease pro-inflammatory cytokines expression and sebum production. A decrease in erythema and sebum production without side effects were confirmed in a human trial which involved topical application of phytocannabinoids for 12 weeks.6668 On the contrary, CBG and cannabigerovarin (CBGV) increased sebum production in human sebocytel (SZ95) cells.28,69

6. Neurological Disorders and Neurodegenerative Diseases

CBD functions as an anti-inflammatory and neuroprotective agent in neurodegenerative disorders. A study by Rajan et al. has demonstrated that CBD reversed alterations of various genes associated with oxidative stress, mitochondrial dysfunction and pathology of amyotrophic lateral sclerosis (AML) using an in vitro model system of human gingiva-derived mesenchymal stromal cells (hGMSCs).70 Double-blind, randomized cross-over design revealed that CBD was neither effective nor toxic at a dose of 700 mg/kg in neuroleptic-free patients diagnosed with Huntington’s disease (HD).71 A study by Zuardi et al. has demonstrated that CBD decreased stereotypy induced by dopaminergic agonists but did not induce catalepsy, an extrapyramidal side effect of antipsychotics such as haloperidol.72 CBD at higher doses of greater than 120 mg/kg increased prolactin levels, a side effect observed similar to clozapine treatment, indicating that CBD acts as atypical antipsychotic agent. CBD decreases hyperlocomotion and prepulse inhibition (PPI) impairment induced by amphetamine, glutamate NMDA receptor non-competitive antagonists such as ketamine and MK801 in rats and mice.7376 CBD independent of NGF has showed neurorestorative potential in MPP+ model of Parkinson’s disease.77 CBD shows reduced affinity towards endocannabinoid receptors but higher towards Transient Receptor Potential Vanilloid receptor type 1 (TRPV1). TRPV1, a non-selective channel in its active form facilitates glutamate release and Ca2+ permeability, which cause neuronal excitability in epilepsy. Anti-epileptic action of CBD involves desensitization of TRPV1channels and normalization of intracellular Ca2+ levels.78 CBD also shows high affinity towards serotonin receptors such as 5-HT1A and 5-HT2A, which differ in their characteristics and functions. 5HT1 receptors activated in the hippocampus increase neurotransmission whereas 5-HT1A receptor activation in raphe nuclei leads to serotonergic neurons inhibition.79 Δ9-THC and CBD are used therapeutically for central nervous system (CNS) diseases such as epilepsy, anxiety, depression and schizophrenic psychosis because of their neurological actions and lipophilic nature. However, Δ9-THC and CBD differ in their penetrability and disposition in the brain. The positive effects of cannabinoids on cognitive function could be due to their protective effect on blood brain barrier (BBB), cerebrovascular structure and function. CBD will not be converted to THC by the biosynthetic enzymes or pathways present in the human body. However, CBD rapidly gets cyclized to THC in acidic pH conditions of stomach, which suggests that oral administration of CBD can cause some side effects due to its conversion to THC. This study also suggests the need to explore other routes of administration such as transdermal-based for delivery of CBD in order to reduce the formation of psychoactive cannabinoids.80 THC causes psychomotor and cognitive impairments, increased heart rate and dry mouth. However, these effects were not observed when CBD is administered orally at high doses to healthy individuals orally.81,82 In another study, oral CBD (1 mg/kg body weight) administered together with THC (0.5 mg/kg body weight) to health volunteers minimized THC-induced anxiety. This shows that of CBD blocks THC-induced anxiety.83 Oral CBD (600 mg) pretreatment significantly reduced anxiety, cognitive impairment, and discomfort in patients with generalized social anxiety disorder.84 A double-blind study conducted at the University of Cologne revealed that CBD administered orally at a dose of 800 mg daily for 2–4 weeks showed efficacy in 42 patients with schizophrenia or schizophreniform disorder but side effects such as extrapyramidal symptoms, increased prolactin levels, and weight gain were observed.85 CBD enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia by increasing serum anandamide levels, which suggests that anandamide deactivation inhibition may be responsible for the antipsychotic effects of CBD in the treatment of schizophrenia.85

7. Cardiovascular

The cardiovascular effects of both synthetic and endogenous cannabinoids have been well demonstrated in numerous studies.8689 The key component of cannabis, Δ9-THC causes tachycardia in humans. Phytocannabinoids induce cardiovascular effects, which vary from one species to another. Cardiovascular effects observed with Δ9-THC are different in animals when compared to humans.90,91 Δ9-THC causes tachycardia in humans, and conscious monkeys,92 whereas bradycardia in other animal models.9395 The main CYP of cardiomyocytes, CYP2J2, metabolizes AEA into cardioprotective epoxides (EET-EAs). Arnold et al. assessed the CYP2J2 metabolism kinetics with AEA, Δ9-THC, Δ8-THC, CBN, CBD, CBG, and CBC.6 Phytocannabinoids are observed to be highly catalyzed by CYP2J2 when compared to AEA. Moreover, these phytocannabinoids strongly inhibit the metabolism of AEA by CYP2J2 of which Δ9-THC has shown the strongest inhibition.6 A study by Hao et al. has revealed the cardioprotective potential of CBD in doxorubicin-induced cardiotoxicity model.96 Doxorubicin (DOX), a common broad-spectrum chemotherapeutic drug is limited by its dose-dependent cardiotoxicity, which may cause irreversible cardiomyopathy or heart failure.97 DOX induces cardiotoxicity by oxidative stress,98,99 nitrosative and nitrative stress,100,101 dysfunction/toxicity of the mitochondria,97,102104 activation of proteins associated with apoptosis and necrosis,105 as well as metabolic and lipid signaling pathway dysregulation.106108 There was significant improvement in DOX-induced cardiac dysfunction, cell death and oxidative/nitrative stress after CBD treatment. CBD promoted biogenesis and cardiac mitochondrial function, which were impaired by DOX.96

8. Arthritis and Pain

Oral, parental and topical administration of nonsteroidal anti-inflammatory drugs (NSAIDs) such as aspirin, diclofenac, ibuprofen, and naproxen are used as a current treatment for various types of arthritis (rheumatoid arthritis, septic arthritis, juvenile idiopathic arthritis).109 Arthritis requires continuous administration of drugs to sustain the quality of life in patient and the major problem encountered is the side effects of these drugs. Novel strategies and targets are being researched as alternate treatment options to relieve the pain caused by arthritis.110 CBD has been successfully delivered for treatment of inflammation in different species via transdermal route.111113 Despite its hydrophobicity and poor oral bioavailability, CBD has been shown to reduce inflammation and pain without side effects. A study by Hammell et al. demonstrates the efficacy of transdermal delivery of CBD in rats in reducing inflammation and pain in a Freund’s adjuvant-induced monoarthritic knee joint model.114 CBD gels ranging from 0.6 to 62.3 mg/day showed reduction in joint swelling, infiltration of immune cells as well as synovial membrane thickening in a dose-dependent manner. A dose dependent decrease in pro-inflammatory markers were observed in the spinal cord and dorsal root ganglia.114 Pain possesses somatosensory, motivational, cognitive, and affective properties,115 and is an experience that engages various parts of the brain and constitute different neurochemical mechanisms.116 Currently, the use of scaffolds as a drug delivery matrix have been successful in various fields of tissue engineering such as bone formation, periodontal regeneration, cartilage development, for artificial corneas, in tendon repair and ligaments as well as in tumors. They are also used in joint pain inflammation, osteochondrogenesis, and wound dressings.117 Cannabinoids including THC, CBD and synthetic cannabinoids have been reported in rodents to promote eCB action in decreasing pain, inflammation, depression, and anxiety similar to the effects of direct CB1 agonists.118123

B. What is Peripheral Neuropathy?

Peripheral neuropathy (PN) involves damage to multiple parts of the peripheral nerve extending from the cell body (i.e., the dorsal root ganglion or anterior horn cell) to the cell projection itself, with its myelin outer coating and axonal projection.124 Factors responsible for the development of PN include diabetes mellitus, shingles, exposure to toxins such as lead and chemotherapies, vitamin B12 deficiency, alcoholism, autoimmune disorders such as rheumatoid arthritis, Lyme disease, syphilis, HIV, etc.125 It can affect motor, sensory or autonomic fibers depending on the underlying cause.125

II. CHEMOTHERAPY-INDUCED PERIPHERAL NEUROPATHY (CIPN)

Chemotherapy-induced peripheral neuropathy (CIPN) is a serious unwanted side effect associated with the therapeutic usage of some chemotherapeutic agents in cancer patients.126 Substances that can damage the peripheral sensory and motor neurons and cause CIPN are categorized into six groups: (a) platinum based antineoplastic drugs (oxaliplatin and cisplatin); (b) immunomodulatory drugs such as thalidomide; (c) vinca alkaloids (vincristine and vinblastine); (d) epothilones (ixabepilone); (e) proteasome inhibitors (bortezomid); and (f) the taxanes (paclitaxel and docetaxel).127 Therapeutic usage of these drugs induce anti-cancer effects and peripheral neuropathy through different mechanisms, which include mitochondrial dysfunction, oxidative stress, microtubule disruption, neuroinflammation, and ion channel dysregulation.128 These drugs cause peripheral sensory symptoms such as paresthesia, dysesthesia, pain, numbness, tingling, and sensitivity to touch and temperature and motor symptoms such as weakness, gait and balance disturbances.129 CIPN is a common clinical problem that affects about 30–40% of patients receiving neurotoxic chemotherapy and leads to a significant increase in the annual healthcare costs.130 Patients receiving chemotherapy have common side effects of which CIPN is not an exception. CIPN is characterized by symptoms such as nerve pain in the upper and lower extremities, loss of sensory and motor nerves. A systematic review on the assessment of the incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy revealed that the prevalence rate of CIPN is 68.1% and 60.0% during the first and third month after chemotherapy.131

There are varying severities of the side effects of anticancer therapies, and some can persist over several years. A review by Stone and DeAngelis demonstrated that more than 20 chemotherapy treatments and radiotherapies may lead to syndromes, which affect the central and peripheral nervous system (PNS) irrespective of the primary disease.132 PNS complications have been observed with at least 21 distinct chemotherapy agents including cisplatin, carboplatin and oxaliplatin.132 Pain relates to the interruption of sensory function due to large myelinated sensory fibers damage. In CIPN, there is a general damage of PNS which affects motor and autonomic functions. PNS damage can be caused by oxidative stress, calcium homeostasis alterations, axon degeneration and membrane remodeling which results from neuroinflammation and activation of apoptotic pathways.133 Several studies have shown the effect of platinum-based chemotherapy in causing CIPN. A study by Kelly et al. showed that high concentrations of cisplatin (10–50 μM), oxaliplatin or carboplatin (100–500 μM) in rat sensory neurons cause increased cell death and apoptosis,134 which is mostly due to reactive oxygen species production and 8-oxoguanine DNA damage accumulation.134 The bodies of sensory neurons located in DRG are the known targets of Pt based compounds.135 Pt-based compounds lead to morphological changes, usually observed in the nucleoli, whose form is affected by decreased production of ribosomal RNA. The formation of DNA-Pt adducts hinders transcription thereby leading to apoptosis especially in large neuronal cells, which are generally dependent on active transcription and translation.136 A review by Zajaczkowska et al. reveals that altered excitability of peripheral neurons is caused by Pt induced ion channels deregulation and compromised mitochondrial DNA transcription.137 Activation of microglia and astrocytes during the initial stages of neuroinflammation was observed and this study corroborates with similar observations by Colvin et al., which shows the role of cytokines in neuronal toxicity. Generally, the mechanisms which are involved for anticancer effect of various chemotherapeutics are also responsible for the neurotoxic effect they cause.137 The combined effect of procarbazine, CCNU and vincristine (PCV) is utilized as an adjuvant therapy in cerebral glioma recurrence. A study by Postma et al. has showed that out of 26 patients with recurrent gliomas treated with PCV, four of them developed central neurotoxic side effects including focal neurological deficit, cognitive disturbances, slow EEG background activity as well as hematological and hepatic toxicity.138 After months of chemotherapy discontinuation, prolonged myelosuppression and neurological deficits are observed in three of these four patients. The central neurotoxic effects were hypothesized to be resulting from the intensive PCV although all four patients have previously been treated with radiotherapy and anticonvulsants.138 Breast cancer patients treated with paclitaxel who developed neuropathy showed differential expression in several pathways involved in mitochondrial dysfunction such as oxidative stress139 in a study by Kober et al.140 Bioenergetic deficits in peripheral nerves have also been observed in CIPN.141 A study by Flatters and Benner revealed that rats treated with paclitaxel had swollen vacuolated mitochondria that is usual of about 3 months of pain-like behaviors.142 Several animal studies have shown that chemotherapy increases oxidative stress.143,144

III. MECHANISM OF CIPN INDUCTION BY VINCA ALKALOIDS

Vincristine is an antimitotic drug utilized for the treatment of numerous tumors and hematologic malignancies such as breast cancer, leukemia and non-Hodgkin’s lymphomas. Microtubules are essential for nerve axons function in vesicle mediated transport and interference of these by vincristine causes axonopathy, which manifests gradually.145 Vincristine as a chemotherapy is essential for the treatment of pediatric malignancies including medulloblastomas and neuroblastomas. Vincristine works by binding to β-tubulin leading to the inhibition of microtubule formation, which subsequently stops cell division and interrupts cancer cell growth.146 A dose-dependent peripheral neuropathy involving sensory, autonomic and motoric interruptions are observed in most of the patients during vincristine treatment.147150 Vincristine-induced motor neuropathy is characterized by foot drop, cramps and abnormalities in gait.151 Loss of sensory discrimination, tingling and numbness are sensory symptoms of vincristine-induced neuropathic pain.152 Vincristine deregulates and modifies neuronal mitochondria structurally, which leads to activation of apoptotic pathway, neuronal excitability and glial cells dysfunction.146

Vincristine induced autonomic dysfunctions are characterized by symptoms such as constipation, urinary retention, orthostatic hypotension and paralytic ileus.153,154 Although vincristine has poor blood-brain barrier penetration,155 numerous reports show that vincristine treatment cause cranial nerve palsies and CNS toxicity.156158

IV. DIABETIC PERIPHERAL NEUROPATHY (DPN)

Diabetic peripheral neuropathy refers to diabetes mellitus induced neuropathic conditions, which vary from one another.159 It is also commonly termed as distal symmetric poly neuropathy or diabetic sensorimotor neuropathy. It mainly damages the nerves present in the arms and legs. More than 80% of the people with clinical diabetic neuropathy are known to be suffering from this form of neuropathies.160 DPN is defined as the presence of symptoms and/or signs in the peripheral nerves, predominantly affecting the lower extremities in the absence of other causes of neuropathy.161 Clinical symptoms of DPN include tingling, burning and painful sensation, sharp pin prick sensations and muscle cramps, sensitivity to touch, loss of balance and in coordination, numbness or loss of sensitivity to thermal and mechanical stimuli and insensitivity to trauma, which leads to foot ulcerations and amputations.162 DPN, a multifactorial disorder which arises due to hyperglycemia and/or insulin deficiency, is characterized by symptoms and/or signs in the peripheral nerves, which predominantly affects the lower extremities in the absence of other factors contributing neuropathy.163 These interrelated defects lead to a progressive unmyelinated and myelinated peripheral nerve fiber loss or damage. The pathophysiology of DPN is associated with multiple factors which makes it challenging to identify targeted therapeutic regimen for this condition. Reports have shown that metabolic excess mediated damage to the endothelial cells of blood vessels supplying blood flow initiates the pathological events in the neurons and glial cells.164 Conversely, metabolic excess inside the dorsal root ganglions or sciatic nerves activates many alternative metabolic pathways such as the polyol pathway, hexosamine pathway, protein kinase C pathway, AGE’s formation, etc., thereby leading to redox imbalance which initiates apoptotic pathways to kill these cells.165 Accumulating literature extensively explains the involvement of autophagic deficiency, mitochondrial dysfunction, disabled mitochondrial biogenesis, inflammation, oxidative stress, endoplasmic stress, etc., in the progression of DPN.165 Oxidative stress has been reported recently to play key factor in advancement of DPN.163,166,167 Oxidative stress is promoted by hyperglycemia by both non-enzymatic and enzymatic mechanisms such as non-enzymatic protein glycation and glucose reduction to sorbitol by nicotinamide dinucleotide-linked enzyme aldose reductase (AR).163,166 Sorbitol accumulation in the body results in the reduction of taurine, which is an endogenous antioxidant,168 and osmolytes, thus leading to oxidative stress.163,166,169

V. APPLICATION OF CBD, CBN, AND CBG IN NEUROPATHIC PAIN

Pain is classified into two main types: chronic and acute, as shown in Fig. 4. Cannabinoids are one of the potential candidates for the therapy of neuropathic, inflammatory, and cancer associated pain.170172 These cannabinoids have been studied for different neuropathy conditions in animals such as DPN, CIPN, chronic construction traumatic injury (CCI), trigeminal neuralgia, etc. A study by Lazic et al. revealed that CBD was highly effective with minimal side effects when compared to cannabis and Δ9-THC, the efficacies of which are hindered due to side effects.173 THC and CBD reduce CCI induced mechanical allodynia in a dose dependent manner. However, side effects are less with CBD when compared to THC. Moreover, co-administration of low doses of THC and CBD significantly reduced allodynia in a biphasic dose-dependent manner with minimal side effects in an animal model of neuropathic pain.173 Another study by Ward et al. shows the protective action of CBD in combating paclitaxel (PAC) induced neuropathic pain partly through mediation by the 5-HT1A receptor system.174 The combination of CBD and PAC chemotherapy was suggested because of its minimal side effects as well as its ability to attenuate CIPN.174 Recently, CBD was recruited in phase 2 clinical trials for studying its pain-relieving effects against DPN (NCT04679545). This study intends to evaluate the impact of CBD on neuropathic pain, anxiety, and sleep quality compared to a placebo control. University of Copenhagen registered in clinicaltrials.gov to study the effects of CBD in attenuating oxaliplatin or paclitaxel induced neuropathy in a phase 2 clinical trials (NCT04582591). Meta-analysis of 16 studies with 1750 participants showed that cannabis-based medicines (oral mucosal spray of TCH + CBD, nabilone and dronabinol) may increase the number of people achieving 50% or greater pain relief compared with placebo.175 In another systematic review and meta-analysis of randomized controlled trials articulated cannabis-based medicines might be effective for chronic pain treatment, based on limited evidence, primarily for neuropathic pain (NP) patients.176 Figure 5 summarizes possible pathways by which cannabinoids attenuates neuropathic pain and other disease conditions.

FIG. 4:

FIG. 4:

Types of pain. Neuropathic pain is sub-type of chronic pain and could be central/peripheral. Peripheral neuropathic pain includes diabetic neuropathy, CIPN and HIV related neuropathic pain.

FIG. 5:

FIG. 5:

Plausible mechanism of action of cannabinoids in protecting peripheral neurons against various disease conditions. Cannabinoids have been found to activate TRPV1 ion channels, GPR55 and 5HT1A receptors including CB1 and CB2 receptors. By activation of these receptors or by regulating the downstream AMPK, Nrf2, NF-κB and BDNF cell signalling pathways, Cannabinoids improved neurogenesis, increased autophagosomes formation, increased mitochondrial biogenesis and improved mitochondrial function and enhanced antioxidant response with decreasing inflammation in different conditions of neuropathy. AMPK, adenosine monophosphate-activated protein kinase; Atg, anti-thymocyte globulin; BDNF, brain-derived neurotrophic factor; FIP200, FAK family kinase-interacting protein of 200 kDa; GSH, Glutathione; HO1, heme oxygenase 1; mTOR, mechanistic target of rapamycin; SIRT1, silent mating type information regulation 2 homolog 1; NRF, nuclear respiratory factor; Nrf2, nuclear factor erythroid 2 (NFE2)-related factor 2; NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells; PGC-lα, peroxisome proliferator-activated receptor gamma coactivator 1-alpha; TFAM, mitochondrial transcription factor; Ulk1, unc-51 like autophagy activating kinase; VPS34, vacuolar protein sorting 34.

A. US Food and Drug Administration (FDA)-Approved Products and Clinical Trials

Two synthetic cannabis-related drug products, Dronabinol and Nabilone containing Δ9-THC are approved by the FDA for the treatment of anorexia associated with weight loss in AIDS patients and chemotherapy-induced nausea and vomiting. Another FDA approved product is CBD, a cannabis-derived compound177 for the treatment of seizures associated with Lennox-Gastaut syndrome and Dravet syndrome in children who are 2 years and above.178 Lennox-Gastaut syndrome, a disease characterized by multiple types of seizures is usually first diagnosed in children between 3 to 5 years and usually persists into adulthood. Various anti-epileptic drugs are used as first-line treatment and CBD is used as an adjuvant therapy.179 CBD stands as the only FDA approved treatment for the patients with Dravet syndrome.180 Epidiolex, a pure CBD oral solution manufactured by GW pharmaceuticals received approval from the FDA when it significantly decreased the frequency of seizures in cases of treatment-resistant epilepsy from several trials.181183 CBD has been explored for potential therapeutic use in cancers, chronic pain, neurodegenerative diseases such as Alzheimer and Parkinson disease, inflammatory and psychiatric diseases.184,185 However none of these trials (as shown in Tables 1 and 2) have been approved by the FDA for the treatment of neurodegenerative disease, chronic pain, cancers and psychiatric diseases with CBD oil.186,187

TABLE 1:

Clinical trials of cannabinoids in neuropathic pain

Study Title ID and year (estimated ) Status Condition Intervention Phase
Cannabinoids and an Anti-inflammatory Diet for the Treatment of Neuropathic Pain After Spinal Cord Injury NCT04057456 2020–2022 Not yet Recruiting Spinal Cord Injuries Neuropathic Pain Drug: TCH/CBD Capsules Other: Anti-inflammatory diet Phase 2188
Cannabinoids Effects on the Pain Modulation System NCT02560545 2015–2019 Unknown Neuropathic Pain Drug: Cannabis oil N/A189
Effect of Cannabinoids on Spasticity and Neuropathic Pain in Spinal Cord Injured Persons NCT01222468 2012–2015 Completed Muscle Spasticity as a Result of Spinal Cord Injury Drug: Nabilone 0.5 mg Drug: Placebo Phase 2190
Nabilone for the Treatment of Phantom Limb Pain NCT00699634 2009–2011 Completed Phantom Limb Pain Neuropathic Pain Drug: Nabilone N/A191
Sativex for Treatment of Chemotherapy Induced Neuropathic Pain NCT00872144 2009–2014 Completed Neuropathic Pain Drug: Sativex Phase 3192
Efficacy Study of Nabilone in the Treatment of Diabetic Peripheral Neuropathic Pain NCT01035281 2008–2011 Diabetic Neuropathies Drug: Nabilone, flexible dosing Phase 3193
Effect of Cannabis and Endocannabinoids on HIV Neuropathic Pain NCT03099005 2018–2020 Recruiting Cannabis HIV Neuropathy Pain Syndrome Drug: Cannabis Phase 2194
An Observational Post-Marketing Safety Registry of Sativex® NCT02073474 2011–2015 Completed Multiple Sclerosis Diabetes Cancer Neuropathic Pain Drug: Sativex N/A195
Efficacy and Safety of the Pain Relieving Effect of Dronabinol in Central Neuropathic Pain Related to Multiple Sclerosis NCT00959218 2007–2010 Completed Central Neuropathic Pain in Multiple Sclerosis Drug: Dronabinol Drug: Placebo N/A196
Trial of Dronabinol and Vaporized Cannabis in Neuropathic Low Back Pain NCT02460692 2016–2020 Recruiting Cannabis Low-Back Pain Neuropathic Pain Drug: Placebos Drug: Dronabinol Drug: Vaporized Cannabis 3.7% THC/5.6% CBD N/A197
Effect of Delta-9-Tetrahydrocannabinol on the Prevention of Chronic Pain in Patients with Acute CRPS (ETIC-Study) NCT00377468 2006–2008 Unknown Complex Regionl Pain Syndromes CRPS Drug: Delta9-Tetrahydrocannabinol Phase 2198

TABLE 2:

Clinical trials of cannabinoids in chronic and acute pain

Study title ID and year (estimated ) Status Condition Intervention Phase
Study to Evaluate the Efficacy of Dronabinol (Marinol) as Add-On Therapy for Patients on Opioids for Chronic Pain NCT00153192 2001–2006 Completed Chronic Pain Drug: Marinol (dronabinol) Phase 2 and Phase 3199
Vaporized Cannabis for Chronic Pain Associated with Sickle Cell Disease (Cannabis-SCD) NCT01771731 2014–2017 Completed Sickle Cell Disease (Chronic Pain) Drug: Cannabis Phase 1 and Phase 2200
A Study to Compare Sublingual Cannabis Based Medicine Extracts with Placebo to Treat Brachial Plexus Injury Pain NCT01606189 2001–2002 Completed Pain Drug: Immediate-release Oral Morphine Sulfate Tablets Other: Thermal and Pressure Nociceptive Sensitivity Drug: GW-1000–02(Sativex) Drug: GW-2000–02(THC (25 mg/ml) Drug: Placebo Phase 3201
Pain Research: Innovative Strategies with Marijuana (PRISM) NCT03522324 2018–2022 Recruiting Chronic Pain Chronic Low Back Pain Cannabis Use Drug: Cannabis Edible N/A202
Opioid and Cannabinoid Pharmacokinetic Interactions NCT00308555 2006–2009 Completed Pain Drug: Cannabis Phase l203
Safety and Efficacy of Medical Cannabis Oil in the Treatment of Patients with Chronic Pain NCT03337503 Jan 2018-Dec 2018 Unknown Chronic Pain Drug: THC and CDB in a 1 to 1 ratio Drug: Experimental: THC and CBD in a 1 to 2 ratio Drug: High CBD with trace THC oil Other: Placebo Phase 4204
Efficacy of Ultra-Micronized Palmitoylethanolamide (Um-PEA) in Geriatric Patients with Chronic Pain NCT02699281 2015–2016 Completed Chronic Pain Drug: Ultramicronized palmitoylethanolamide Drug: Placebo Phase 4205
Treatment of Chronic Pain with Cannabidiol (CBD) and Delta-9-tetrahydrocannabinol (THC) NCT03215940 2018–2020 Recruiting Chronic Pain Drug: Dronabinol Early Phase 2 206
Dronabinol Opioid Sparing Evaluation (DOSE) Trial NCT03766269 2018–2019 Recruiting Chronic Pain Drug: Dronabinol Phase 2207
Effect of Delta-9-Tetrahydrocannabinol on the Prevention of Chronic Pain in Patients with Acute CRPS (ETIC-Study) NCT00377468 2006–2008 Unknown Complex Regional Pain Syndromes (CRPS) Drug: Delta9-Tetrahydrocannabinol Phase 2198
Namisol®) in Patients with Persistent Postsurgical Abdominal Pain NCT01562483 2012–2014 Completed Postsurgical Pain Abdominal Pain Chronic Pain Drug: Tetrahydrocannabinol Drug: Placebo Phase 2208
Δ9-THC (Namisol®) m Chronic Pancreatitis Patients Suffering From Persistent Abdominal Pain NCT01551511 2012–2014 Completed Pancreatitis, Chronic Abdominal Pain Chronic Pain Drug: Tetrahydrocannabinol Drug: Placebo Phase 2209
Outcomes Mandate National Integration With Cannabis as Medicine (OMNI-Can) NCT03944447 2018–2025 Chronic Pain Chronic Pain Syndrome Chronic Pain Due to Injury Chronic Pain Due to Trauma Fibromyalgia Seizures Hepatitis C Cancer Drug: Cannabis, Medical Phase 2210
Use of the Cannabinoid Nabilone for the Promotion of Sleep in Chronic, Non-Malignant Pain Patients NCT00384410 2005 Unknown Pain Drug: Nabilone Phase 2211
Cannabis Oil for Chronic Non-Cancer Pain Treatment NCT03635593 2019–2022 Not yet recruiting Chronic Non-cancer Pain Drug: CBD Drug: CBD+THC Other: Placebo Phase 2212
Efficacy Study of Δ9-THC to Treat Chronic Abdominal Pain (Delta-pain) NCT01318369 2011–2013 Completed Cannabinoids Tetrahydrocannabinol Chronic Pancreatitis Abdominal Pain Drug: Namisol Drug: Diazepam Phase 2213
Supporting Effect of Dronabinol on Behavioral Therapy in Fibromyalgia and Chronic Back Pain NCT00176163 2005–2009 Completed Behavioral: Operant behavioral treatment; Drug: THC Fibromyalgia Back Pain Phase 2214
Palmitoylethanolamide for Post-operative Pain Prevention (PEA for CPSP) NCT01491191 2012–2013 Unknown Dietary Supplement: Palmitoylethanolamide Dietary Supplement: Placebo Chronic Post-operative Pain N/A215
Nabilone Versus Amitriptyline in Improving Quality of Sleep in Patients With Fibromyalgia NCT00381199 2006–2007 Completed Drug: Amitriptyline Hydrochloride Drug: Nabilone Fibromyalgia Sleep Initiation and Maintenance Disorders N/A216
Efficacy of Palmitoylethanolamide-polydatin, Combination on Chronic Pelvic Pain in Patients With Endometriosis NCT02372903 2013–2015 Completed Drug: Administration of micronized Palmitoylethanolamide (PEA)-Transpolydatin Endometriosis Chronic Pelvic Pain N/A217
Nabilone Use For Acute Pain in Inflammatory Bowel Disease Patients NCT03422861 2020–2021 Not yet recruiting Drug: Nabilone Drug: Placebos Inflammatory Bowel Diseases N/A218
Evaluation of Dronabinol For Acute Pain Following Traumatic Injury NCT03928015 2019–2021 Enrolling by invitation Drug: Adjunctive dronabinol Drug: Systemic analgesics Traumatic Injury Pain, Acute Phase 2219
Cannabidiol, Morphine, Pain (CMP) NCT04030442 2019–2021 Recruiting CBD Chronic Pain Drug: Immediate release oral morphine sulfate tablets Other: Thermal and Pressure nociceptive sensitivity Phase 1220

VI. PHARMACOKINETICS OF CANNABINOIDS

The route of administration of cannabinoids has considerable effects on their pharmacokinetics, bioactivity, bioavailability, and effectiveness. Cannabinoids cannot be administered intravenously due to their poor water solubility. In cancer therapy, oral administration of cannabinoids may not be the most effective route due to partial degradation by the acidic environment present in stomach.221 A study by Deiana et al. has assessed the pharmacokinetic profiles of CBD, cannabidivarine (CBDV), Δ9-tetrahydrocannabivarin (THCV), and CBG after an acute single-dose intraperitoneal and oral administration in mice and rats.222 A marble burying test was used to assess the pharmacodynamic-pharmacokinetic relationship of CBD when administered at a dose of 120 mg/kg through oral and intraperitoneal route in mice. Phytocannabinoids dispersed in solutol and administered orally have easily penetrated the blood-brain barrier when compared to cremophor based phytocannabinoids. However, cremophor-based phytocannabinoids administered intraperitoneally showed higher plasma and brain concentrations. CBD and CBDV show higher brain concentrations when administered orally. However, THCV and CBG were more effective when administered through intraperitoneal route.222 CBD administered orally has shown low bioavailability due to its first-pass metabolism in both dogs and humans.223,224 The pharmacokinetics of CBD were assessed in healthy dogs in a study by Bartner et al., when administered in the form of oral microencapsulated oil beads, oral CBD-infused oil or CBD-infused transdermal cream.225 This study demonstrates that oral CBD-infused oil showed good pharmacokinetic profile.223,225

Oral or intranasal administration of CBD have shown poor bioavailability. CBD has been shown in several studies in rodents to possess antidepressant-like effects,226229 but bioavailability is a major challenge in the use of CBD in clinical trials despite its significant effects in suppressing depression.222,230 The administration of medical cannabis to patients is usually via inhalation,231,232 spraying,233 or oral in the form of oil,234 capsules,235 and cookies.236 Oral administration of CBD or via inhalation has poor bioavailability. Xu et al. explored the pharmacokinetics and the efficacy of CBD administration over a long period via oral and intravenous routes for treating chronic mild stress (CMS).237

VIII. FUTURE DIRECTIONS

Although cannabinoids have great therapeutic potential in neuropathic pain, there is a need to scale up its production. The production of cannabinoids is limited to the cannabis plant, which may lead to insufficient supply of quality cannabinoids for therapeutic uses. Cannabis plant as a sole source of cannabinoids faces several challenges including climatic, disease and pest control, land/agricultural space, legal issues because of abuse of plant, low quantities of minor phytocannabinoids and chemical procedures for cannabinoid isolation. A solution to this problem could be the use of biotechnological approaches such as microbes and yeast biosynthesis. This will enhance the production of cannabinoids for therapeutic use as well as increase the accessibility of rare cannabinoids for clinical use and drug testing. Biotechnological approaches could lead to the discovery and production of novel cannabinoids, which are more effective or could be used in targeted therapy. Microbial synthesis of cannabinoids can be employed to increase the yield of cannabinoids which are produced in low quantities in the cannabis plant. The selection of the microbes for the synthesis/production of cannabinoids requires certain considerations to be taken into account, such as genetic factors, genetic engineering procedures or biological tools and the pathways for the synthesis of these compounds.

ACKNOWLEDGMENTS

The authors are thankful to the Consortium for Medical Marijuana Clinical Outcomes Research (Grant No. SUB00002097), the National Institute on Minority Health and Health Disparities of National Institutes of Health (Grant No. U54 MD007582), and the NSF-CREST Center for Complex Materials Design for Multidimensional Additive Processing (CoManD) (Grant No. 1735968). The authors declare no competing interests.

REFERENCES

  • 1.Friedman D, Sirven JI. Historical perspective on the medical use of cannabis for epilepsy: Ancient times to the 1980s. Epilepsy Behav. 2017;70:298–301. [DOI] [PubMed] [Google Scholar]
  • 2.Zuardi AW. History of cannabis as a medicine: A review. Braz J Psychiatry. 2006;28(2):153–7. [DOI] [PubMed] [Google Scholar]
  • 3.ElSohly MA, Slade D. Chemical constituents of marijuana: The complex mixture of natural cannabinoids. Life Sciences. 2005;78(5):539–48. [DOI] [PubMed] [Google Scholar]
  • 4.Grotenhermen F, Russo E. Cannabis and cannabinoids: Pharmacology, toxicology, and therapeutic potential. Boca Raton, FL: Psychology Press; 2002. [Google Scholar]
  • 5.Andre CM, Hausman JF, Guerriero G. Cannabis sativa: The plant of the thousand and one molecules. Front Plant Sci. 2016;7:19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Arnold WR, Weigle AT, Das A. Cross-talk of cannabinoid and endocannabinoid metabolism is mediated via human cardiac CYP2J2. J Inorg Biochem. 2018;184:88–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Morales P, Hurst DP, Reggio PH. Molecular targets of the phytocannabinoids: A complex picture. In: Kinghorn DA, Falk H, Gibbons S, Kobayashi J, editors. Phytocannabinoids. Cham, Switzerland: Springer; 2017. p. 103–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Riedel G, Fadda P, McKillop SS, Pertwee RG, Platt B, Robinson L. Synthetic and plant derived cannabinoid receptor antagonists show hypophagic properties in fasted and non-fasted mice. Br J Pharmacol. 2009;156(7):1154–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hanuš LO, Meyer SM, Muñoz E, Taglialatela-Scafati O, Appendino G. Phytocannabinoids: A unified critical inventory. Nat Prod Rep. 2016;33(12):1357–92. [DOI] [PubMed] [Google Scholar]
  • 10.Booth JK, Bohlmann J. Terpenes in Cannabis sativa from plant genome to humans. Plant Science. 2019;284:67–72. [DOI] [PubMed] [Google Scholar]
  • 11.Mechoulam R, Hanuš LO, Pertwee R, Howlett AC. Early phytocannabinoid chemistry to endocannabinoids and beyond. Nat Rev Neurosci. 2014;15(11):757–64. [DOI] [PubMed] [Google Scholar]
  • 12.Battista N, Di Tommaso M, Bari M, Maccarrone M. The endocannabinoid system: An overview. Front Behav Neurosci. 2012;6:9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Pertwee RG, Ross RA. Cannabinoid receptors and their ligands. Prostaglandins Leukot Essent Fatty Acids. 2002;66(2–3):101–21. [DOI] [PubMed] [Google Scholar]
  • 14.Rodriguez de Fonseca F, Del Arco I, Bermudez-Silva FJ, Bilbao A, Cippitelli A, Navarro M. The endocannabinoid system: Physiology and pharmacology. Alcohol Alcoholism. 2005;40(1):2–14. [DOI] [PubMed] [Google Scholar]
  • 15.Brents LK. Focus: Sex and gender health: Marijuana, the endocannabinoid system and the female reproductive system. Yale J Biol Med. 2016;89(2):175. [PMC free article] [PubMed] [Google Scholar]
  • 16.Du Plessis SS, Agarwal A, Syriac A. Marijuana, phytocannabinoids, the endocannabinoid system, and male fertility. J Assist Reprod Genet. 2015;32(11): 1575–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Alfulaij N, Meiners F, Michalek J, Small-Howard AL, Turner HC, Stokes AJ. Cannabinoids, the heart of the matter. J Am Heart Assoc. 2018;7(14):e009099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Bab I, Ofek O, Tam J, Rehnelt J, Zimmer A. Endocannabinoids and the regulation of bone metabolism. J Endocrinol. 2008;20:69–74. [DOI] [PubMed] [Google Scholar]
  • 19.Pertwee RG. Cannabinoids and the gastrointestinal tract. Gut. 2001;48(6):859–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Duncan M, Davison JS, Sharkey KA. Endocannabinoids and their receptors in the enteric nervous system. Aliment Pharmacol Ther. 2005;22(8):667–83. [DOI] [PubMed] [Google Scholar]
  • 21.Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bonner TI. Structure of a cannabinoid receptor and functional expression of the cloned cDNA. Nature. 1990;346(6284):561–4. [DOI] [PubMed] [Google Scholar]
  • 22.Munro S, Thomas KL, Abu-Shaar M. Molecular characterization of a peripheral receptor for cannabinoids. Nature. 1993;365(6441):61. [DOI] [PubMed] [Google Scholar]
  • 23.Massa F, Storr M, Lutz B. The endocannabinoid system in the physiology and pathophysiology of the gastrointestinal tract. J Mol Med. 2005;83(12):944–54. [DOI] [PubMed] [Google Scholar]
  • 24.Wright K, Rooney N, Feeney M, Tate J, Robertson D, Welham M, Ward S. Differential expression of cannabinoid receptors in the human colon: Cannabinoids promote epithelial wound healing. Gastroenterology. 2005;129(2):437–53. [DOI] [PubMed] [Google Scholar]
  • 25.Rosenthaler S, Pohn B, Kolmanz C, Huu CN, Krewenka C, Huber A, Kranner B, Rausch W-D, Moldzio R. Differences in receptor binding affinity of several phytocannabinoids do not explain their effects on neural cell cultures. Neurotoxicol Teratol. 2014;46:49–56. [DOI] [PubMed] [Google Scholar]
  • 26.Izzo AA, Borrelli F, Capasso R, Di Marzo V, Mechoulam R. Non-psychotropic plant cannabinoids: New therapeutic opportunities from an ancient herb. Trends Pharmacol Sci. 2009;30(10):515–27. [DOI] [PubMed] [Google Scholar]
  • 27.Pertwee RG. The diverse CB1 and CB2 receptor pharmacology of three plant cannabinoids: DeltΔ9-tetrahydrocannabinol, cannabidiol and deltΔ9-tetrahydrocannabivarin. Br J Pharmacol. 2008;153(2): 199–215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.De Petrocellis L, Ligresti A, Moriello AS, Allar ÃM, Bisogno T, Petrosino S, Stott CG, Di Marzo V Effects of cannabinoids and cannabinoid-enriched Cannabis extracts on TRP channels and endocannabinoid metabolic enzymes. Br J Pharmacol.163(7):1479–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Pertwee RG, Ross RA, Craib SJ, Thomas A. (−)-Cannabidiol antagonizes cannabinoid receptor agonists and noradrenaline in the mouse vas deferens. Eur J Pharmacol. 2002;456(1–3):99–106. [DOI] [PubMed] [Google Scholar]
  • 30.Thomas A, Baillie GL, Phillips AM, Razdan RK, Ross RA, Pertwee RG. Cannabidiol displays unexpectedly high potency as an antagonist of CB1 and CB2 receptor agonists in vitro. Br J Pharmacol. 2007;150(5):613–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.McPartland JM, Glass M, Pertwee RG. Meta analysis of cannabinoid ligand binding affinity and receptor distribution: Interspecies differences. Br J Pharmacol. 2007;152(5):583–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mowat C, Cole A, Windsor AL, Ahmad T, Arnott I, Driscoll R, Mitton S, Orchard T, Rutter M, Younge L. Guidelines for the management of inflammatory bowel disease in adults. Gut. 2011;60(5):571–607. [DOI] [PubMed] [Google Scholar]
  • 33.Devlen J, Beusterien K, Yen L, Ahmed A, Cheifetz AS, Moss AC. The burden of inflammatory bowel disease: A patient-reported qualitative analysis and development of a conceptual model. Inflamm Bowel Dis. 2014;20(3):545–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Izzo AA, Sharkey KA. Cannabinoids and the gut: New developments and emerging concepts. Pharmacol Ther. 2010;126(1):21–38. [DOI] [PubMed] [Google Scholar]
  • 35.Mancinelli R, Fabrizi A, Del Monaco S, Azzena GB, Vargiu R, Colombo GC, Gessa GL. Inhibition of peristaltic activity by cannabinoids in the isolated distal colon of mouse. Life Sci. 2001;69(1): 101–11. [DOI] [PubMed] [Google Scholar]
  • 36.Coutts AA, Irving AJ, Mackie K, Pertwee RG, Anavi Goffer S. Localisation of cannabinoid CB1 receptor immunoreactivity in the guinea pig and rat myenteric plexus. J Comp Neurol. 2002;448(4):410–22. [DOI] [PubMed] [Google Scholar]
  • 37.Borrelli F, Fasolino I, Romano B, Capasso R, Maiello F, Coppola D, Orlando P, Battista G, Pagano E, Di Marzo V. Beneficial effect of the non-psychotropic plant cannabinoid cannabigerol on experimental inflammatory bowel disease. Biochem Pharmacol. 2013;85(9):1306–16. [DOI] [PubMed] [Google Scholar]
  • 38.Bagde A, Patel K, Mondal A, Kutlehria S, Chowdhury N, Gebeyehu A, Patel N, Kumar N, Singh M. Combination of UVB absorbing titanium dioxide and quercetin nanogel for skin cancer chemoprevention. AAPS PharmSciTech. 2019;20(6): 1–12. [DOI] [PubMed] [Google Scholar]
  • 39.Arthur P, Patel N, Surapaneni SK, Mondal A, Gebeyehu A, Bagde A, Kutlehria S, Nottingham E, Singh M. Targeting lung cancer stem cells using combination of Tel and Docetaxel liposomes in 3D cultures and tumor xenografts. Toxicol Appl Pharmacol. 2020;401:115112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Miranda MA, Marcato PD, Mondal A, Chowdhury N, Gebeyehu A, Surapaneni SK, Bentley MVLB, Amaral R, Pan C-X, Singh M. Cytotoxic and chemosensitizing effects of glycoalkaloidic extract on 2D and 3D models using RT4 and patient derived xenografts bladder cancer cells. Mater Sci Eng C Mater Biol Appl. 2021;119:111460. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Patel K, Doddapaneni R, Patki M, Sekar V, Bagde A, Singh M. Erlotinib-valproic acid liquisolid formulation: Evaluating oral bioavailability and cytotoxicity in erlotinib-resistant non-small cell lung cancer cells. AAPS PharmSciTech. 2019;20(3):1–11. [DOI] [PubMed] [Google Scholar]
  • 42.Shaik MS, Chatteijee A, Singh M. Effects of monensin liposomes on the cytotoxicity, apoptosis and expression of multidrug resistance genes in doxorubicin-resistant human breast tumour (MCF-7/dox) cell-line. J Pharm Pharmacol. 2004;56(7):899–907. [DOI] [PubMed] [Google Scholar]
  • 43.Jain A, Tiwari A, Verma A, Saraf S, Jain SK. Combination cancer therapy using multifunctional liposomes. Crit Rev Ther Drug Carrier Syst. 2020;37(2):105–34. [DOI] [PubMed] [Google Scholar]
  • 44.Patel AR, Chougule MB, Townley I, Patlolla R, Wang G, Singh M. Efficacy of aerosolized celecoxib encapsulated nanostructured lipid carrier in non-small cell lung cancer in combination with docetaxel. Pharm Res. 2013;30(5):1435–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Ferdous AJ, Stembridge NY, Singh M. Role of monensin PLGA polymer nanoparticles and liposomes as potentiator of ricin A immunotoxins in vitro. J Control Release. 1998;50(1):71–8. [DOI] [PubMed] [Google Scholar]
  • 46.Borrelli F, Pagano E, Romano B, Panzera S, Maiello F, Coppola D, De Petrocellis L, Buono L, Orlando P, Izzo AA. Colon carcinogenesis is inhibited by the TRPM8 antagonist cannabigerol, a Cannabis-derived non-psychotropic cannabinoid. Carcinogenesis. 2014;35(12):2787–97. [DOI] [PubMed] [Google Scholar]
  • 47.Ngwa W, Kumar R, Moreau M, Dabney R, Herman A. Nanoparticle drones to target lung cancer with radiosensitizers and cannabinoids. Front Oncol. 2017;7:208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Guzman M. Cannabinoids: Potential anticancer agents. Nat Rev Cancer. 2003;3(10):745–55. [DOI] [PubMed] [Google Scholar]
  • 49.Ngwa W, Boateng F, Kumar R, Irvine DJ, Formenti S, Ngoma T, Herskind C, Veldwijk MR, Hildenbrand GL, Hausmann M. Smart radiation therapy biomaterials. Int J Radiat Oncol Biol Phys. 2017;97(3):624–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Ngwa W, Irabor OC, Schoenfeld JD, Hesser J, Demaria S, Formenti SC. Using immunotherapy to boost the abscopal effect. Nat Rev Cancer. 2018;18(5):313–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Scott KA, Dalgleish AG, Liu WM. The combination of cannabidiol and Δ9-tetrahydrocannabinol enhances the anticancer effects of radiation in an orthotopic murine glioma model. Mol Cancer Ther. 2014;13(12):2955–67. [DOI] [PubMed] [Google Scholar]
  • 52.Brierley DI, Harman JR, Giallourou N, Leishman E, Roashan AE, Mellows BAD, Bradshaw HB, Swann JR, Patel K, Whalley BJ. Chemotherapy induced cachexia dysregulates hypothalamic and systemic lipoamines and is attenuated by cannabigerol. J Cachexia Sarcopenia Muscle. 2019;10(4):844–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang X, Qin Y, Pan Z, Li M, Liu X, Chen X, Qu G, Zhou L, Xu M, Zheng Q. Cannabidiol induces cell cycle arrest and cell apoptosis in human gastric cancer SGC-7901 cells. Biomolecules. 2019;9(8):302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jacobsson SOP, Rongärd E, Stridh M, Tiger G, Fowler CJ. Serum-dependent effects of tamoxifen and cannabinoids upon C6 glioma cell viability. Biochem Pharmacol. 2000;60(12):1807–13. [DOI] [PubMed] [Google Scholar]
  • 55.Elmes MW, Kaczocha M, Berger WT, Leung K, Ralph BP, Wang L, Sweeney JM, Miyauchi JT, Tsirka SE, Ojima I. Fatty acid-binding proteins (FABPs) are intracellular carriers for Δ9-tetrahydrocannabinol (THC) and cannabidiol (CBD). J Biol Chem. 2015;290(14):8711–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.O’Sullivan SE, Sun Y, Bennett AJ, Randall MD, Kendall DA. Time-dependent vascular actions of cannabidiol in the rat aorta. Eur J Pharmacol. 2009;612(1–3):61–8. [DOI] [PubMed] [Google Scholar]
  • 57.Ramer R, Hinz B. Cannabinoids as anticancer drugs. Adv Pharmacol. 2017;80:397–436. [DOI] [PubMed] [Google Scholar]
  • 58.Brierley DI, Samuels J, Duncan M, Whalley BJ, Williams CM. A cannabigerol-rich Cannabis sativa extract, devoid of [INCREMENT]9-tetrahydrocannabinol, elicits hyperphagia in rats. Behav Pharmacol. 2017;28(4):280–4. [DOI] [PubMed] [Google Scholar]
  • 59.Colasanti BK, Craig CR, Allara RD. Intraocular pressure, ocular toxicity and neurotoxicity after administration of cannabinol or cannabigerol. Exp Eye Res. 1984;39(3):251–9. [DOI] [PubMed] [Google Scholar]
  • 60.Tomida I, Azuara-Blanco A, House H, Flint M Pertwee RG, Robson PJ. Effect of sublingual application of cannabinoids on intraocular pressure: A pilot study. J Glaucoma. 2006;15(5):349–53. [DOI] [PubMed] [Google Scholar]
  • 61.Fellous T, De Maio F, Kalkan H, Carannante B, Boccella S, Petrosino S, Maione S, Di Marzo V, Iannotti FA. Phytocannabinoids promote viability and functional adipogenesis of bone marrow-derived mesenchymal stem cells through different molecular targets. Biochem Pharmacol. 2020;175:113859. [DOI] [PubMed] [Google Scholar]
  • 62.Tóth KF, Ádám D, Bíró T, Oláh A. Cannabinoid signaling in the skin: Therapeutic potential of the c(ut)annabinoid system. Molecules. 2019;24(5):918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sheriff T, Lin MJ, Dubin D, Khorasani H. The potential role of cannabinoids in dermatology. J Dermatolog Treat. 2020;31(8):839–45. [DOI] [PubMed] [Google Scholar]
  • 64.Method to Treat Psoriasis. 2019. [cited 2021 Jan 1]. Available from: http://www.freepatentsonline.com/y2019/0060250.html.
  • 65.Formukong EA, Evans AT, Evans FJ. Analgesic and antiinflammatory activity of constituents of Cannabis sativaL. Inflammation. 1988;12(4):361–71. [DOI] [PubMed] [Google Scholar]
  • 66.Ali A, Akhtar N. The safety and efficacy of 3% Cannabis seeds extract cream for reduction of human cheek skin sebum and erythema content. Pak J Pharm Sci. 2015;28(4):1389–95. [PubMed] [Google Scholar]
  • 67.Dobrosi N, Tóth BI, Nagy G, Dózsa A, Géczy T, Nagy L, Zouboulis CC, Paus R, Kovács L, Bíró T. Endocannabinoids enhance lipid synthesis and apoptosis of human sebocytes via cannabinoid receptor-2-mediated signaling. FASEB J. 2008;22(10):3685–95. [DOI] [PubMed] [Google Scholar]
  • 68.Oláh A, Markovics A Szabó-Papp J, Szabó PT, Stott C, Zouboulis CC, Bíró T. Differential effectiveness of selected non-psychotropic phytocannabinoids on human sebocyte functions implicates their introduction in dry/seborrhoeic skin and acne treatment. Exp Dermatol. 2016;25(9):701–7. [DOI] [PubMed] [Google Scholar]
  • 69.Oláh A, Tóth BA, Borbíró I, Sugawara K, Szöllõsi AG, Czifra G, Pál B, Ambrus L, Kloepper J, Camera E, Ludovici M, Picardo M, Voets T, Zouboulis CC, Paus R, Bíró T. Cannabidiol exerts sebostatic and antiinflammatory effects on human sebocytes. J Clin Invest. 2014;124(9):3713–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Rajan TS, Scionti D, Diomede F, Grassi G, Pollastro F, Piattelli A, Cocco L, Bramanti P, Mazzon E, Trubiani O. Gingival stromal cells as an in vitro model: Cannabidiol modulates genes linked with amyotrophic lateral sclerosis. J Cell Biochem. 2017;118(4):819–28. [DOI] [PubMed] [Google Scholar]
  • 71.Consroe P, Laguna J, Allender J, Snider S, Stern L, Sandyk R, Kennedy K, Schram K. Controlled clinical trial of cannabidiol in Huntington’s disease. Pharmacol Biochem Behav. 1991;40(3):701–8. [DOI] [PubMed] [Google Scholar]
  • 72.Zuardi AW, Rodrigues JA, Cunha JM. Effects of cannabidiol in animal models predictive of antipsychotic activity. Psychopharmacology. 1991;104(2):260–4. [DOI] [PubMed] [Google Scholar]
  • 73.Moreira FA, Guimaraes FS. Cannabidiol inhibits the hyperlocomotion induced by psychotomimetic drugs in mice. Eur J Pharmacol. 2005;512(2–3):199–205. [DOI] [PubMed] [Google Scholar]
  • 74.Pedrazzi JFC, Issy AC, Gomes FV, Guimarães FS, Del-Bel EA. Cannabidiol effects in the prepulse inhibition disruption induced by amphetamine. Psychopharmacology. 2015;232(16):3057–65. [DOI] [PubMed] [Google Scholar]
  • 75.Gomes FV, Issy AC, Ferreira FR, Viveros MP, Del Bel EA, Guimarães FS. Cannabidiol attenuates sensorimotor gating disruption and molecular changes induced by chronic antagonism of NMDA receptors in mice. Int J Neuropsychopharmacol. 2014;18(5):pyu041. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Gomes FV, Llorente R, Del Bel EA, Viveros MP, Lãpez-Gallardo M, Guimaraes FS. Decreased glial reactivity could be involved in the antipsychotic-like effect of cannabidiol. Shizophr Res. 2015;164(1–3):155–63. [DOI] [PubMed] [Google Scholar]
  • 77.Santos NAG, Martins NM, Sisti FM, Fernandes LS, Ferreira RS, Queiroz RHC, Santos AC. The neuroprotection of cannabidiol against MPP+-induced toxicity in PC12 cells involves trkA receptors, upregulation of axonal and synaptic proteins, neuritogenesis, and might be relevant to Parkinson’s disease. Toxicol In Vitro. 2015;30(1 Part B):231–40. [DOI] [PubMed] [Google Scholar]
  • 78.Nazäroglu M. TRPV1 channel: A potential drug target for treating epilepsy. Curr Neuropharmacol. 2015;13(2):239–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Theodore WH. Does serotonin play a role in epilepsy? Epilepsy Curr. 2003;3(5):173–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Calapai F, Cardia L, Sorbara EE, Navarra M, Gangemi S, Calapai G, Mannucci C. Cannabinoids, blood brain barrier, and brain disposition. Pharmaceutics. 2020;12(3):265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Consroe P, Carlini EA, Zwicker AP, Lacerda LA. Interaction of cannabidiol and alcohol in humans. Psychopharmacology. 1979;66(1):45–50. [DOI] [PubMed] [Google Scholar]
  • 82.Bhattacharyya S, Morrison PD, Fusar-Poli P, Martin-Santos R, Borgwardt S, Winton-Brown T, Nosarti C, O’Carroll CM, Seal M, Allen P. Opposite effects of Δ−9-tetrahydrocannabinol and cannabidiol on human brain function and psychopathology. Neuropsychopharmacology. 2010;35(3):764–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zuardi AW, Shirakawa I, Finkelfarb E, Karniol IG. Action of cannabidiol on the anxiety and other effects produced by delta 9-THC in normal subjects. Psychopharmacology. 1982;76(3):245–50. [DOI] [PubMed] [Google Scholar]
  • 84.Bergamaschi MM, Queiroz RHC, Chagas MHN, De Oliveira DCG, De Martinis BS, Kapczinski Fv, Quevedo Jo, Roesler R, Schrãder N, Nardi AE. Cannabidiol reduces the anxiety induced by simulated public speaking in treatment-naive social phobia patients. Neuropsychopharmacology. 2011;36(6):1219–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Leweke FM, Piomelli D, Pahlisch F, Muhl D, Gerth CW, Hoyer C. Cannabidiol enhances anandamide signaling and alleviates psychotic symptoms of schizophrenia. Transl Psychiatry. 2012;2(3):e94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Randall MD, Hams D, Kendall DA, Ralevic V. Cardiovascular effects of cannabinoids. Pharmacol Ther. 2002;95(2):191–202. [DOI] [PubMed] [Google Scholar]
  • 87.Hillard CJ. Endocannabinoids and vascular function. J Pharmacol Exp Ther. 2000;294(1):27–32. [PubMed] [Google Scholar]
  • 88.Kunos G, Jãrai Z, Bätkai S, Goparaju SK, Ishac EJN, Liu J, Wang L, Wagner JA. Endocannabinoids as cardiovascular modulators. Chem Phys Lipids. 2000;108(1–2):159–68. [DOI] [PubMed] [Google Scholar]
  • 89.Ralevic V, Kendall DA, Randall MD, Smart D. Cannabinoid modulation of sensory neurotransmission via cannabinoid and vanilloid receptors: Roles in regulation of cardiovascular function. Life Sci. 2002;71(22):2577–94. [DOI] [PubMed] [Google Scholar]
  • 90.Mendizäbal VE, Adler-Graschinsky E. Cannabinoids as therapeutic agents in cardiovascular disease: A tale of passions and illusions. Br J Pharmacol. 2007;151(4):427–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Randall MD, Kendall DA, O’Sullivan S. The complexities of the cardiovascular actions of cannabinoids. Br J Pharmacol. 2004;142(1):20–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Fredericks AB, Benowitz NL, Savanapridi CY. The cardiovascular and autonomic effects of repeated administration of delta-9-tetrahydrocannabinol to rhesus monkeys. J Pharmacol Exp Ther. 1981;216(2) : 247–53. [PubMed] [Google Scholar]
  • 93.Dewey WL. Cannabinoid pharmacology. Pharm Rev. 1986;38(2):151–78. [PubMed] [Google Scholar]
  • 94.Adams MD, Chait LD, Earnhardt JT. Tolerance to the cardiovascular effects of deltΔ9-tetrahydrocannabinol in the rat. Br J Pharmacol. 1976;56(1):43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Jones RT. Cardiovascular system effects of marijuana. J Clin Pharmacol. 2002;42(S1):58S–63S. [DOI] [PubMed] [Google Scholar]
  • 96.Hao E, Mukhopadhyay P, Cao Z, Erdälyi K, Holovac E, Liaudet L, Lee W-S, Haskã G, Mechoulam R, Pacher P. Cannabidiol protects against doxorubicin-induced cardiomyopathy by modulating mitochondrial function and biogenesis. Mol Med. 2015;21(1):38–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Carvalho FS, Burgeiro A, Garcia R, Moreno AnJ, Carvalho RA, Oliveira PJ. Doxorubicin induced cardiotoxicity: From bioenergetic failure and cell death to cardiomyopathy. Med Res Rev. 2014;34(1):106–35. [DOI] [PubMed] [Google Scholar]
  • 98.Rajagopalan S, Politi PM, Sinha BK, Myers CE. Adriamycin-induced free radical formation in the perfused rat heart: Implications for cardiotoxicity. Cancer Res. 1988;48(17):4766–9. [PubMed] [Google Scholar]
  • 99.Sterba M, Popelovã O, Vãvrovã A, Jirkovskã E, Kavaräkova P, Gersl V, Simune T. Oxidative stress, redox signaling, and metal chelation in anthracycline cardiotoxicity and pharmacological cardioprotection. Antioxid Redox Signal. 2013;18:899–929. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Pacher P, Liaudet L, Bai P, Mabley JG, Kaminski PM, Virãg L, Deb A, Szabó E, Ungvári Z, Wolin MS, Groves JT, Szabó C. Potent metalloporphyrin peroxynitrite decomposition catalyst protects against the development of doxorubicin-induced cardiac dysfunction. Circulation. 2003;107(6):896–904. [DOI] [PubMed] [Google Scholar]
  • 101.Mukhopadhyay P, Rajesh M, Bátkai S, Kashiwaya Y, Hasko G, Liaudet L, Szabó C, Pacher P. Role of superoxide, nitric oxide, and peroxynitrite in doxorubicin-induced cell death in vivo and in vitro. Am J Physiol Heart Circ Physiol. 2009;296(5):H1466–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Doroshow JH, Davies KJ. Redox cycling of anthracyclines by cardiac mitochondria. II. Formation of superoxide anion, hydrogen peroxide, and hydroxyl radical. J Biol Chem. 1986;261(7):3068–74. [PubMed] [Google Scholar]
  • 103.Berthiaume JM, Wallace KB. Adriamycin-induced oxidative mitochondrial cardiotoxicity. Cell Biol Toxicol. 2007;23(1):15–25. [DOI] [PubMed] [Google Scholar]
  • 104.Ichikawa Y, Ghanefar M, Bayeva M, Wu R, Khechaduri A, Prasad SVN, Mutharasan RK, Naik TJ, Ardehali H. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J Clin Invest. 2014;124(2):617–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Pacher P, Liaudet L, Bai P, Virag L, Mabley JG, Hasko G, Szabo C. Activation of poly (ADP-ribose) polymerase contributes to development of doxorubicin-induced heart failure. J Pharmacol Exp Ther. 2002;300(3):862–7. [DOI] [PubMed] [Google Scholar]
  • 106.Mukhopadhyay P, Batkai Sn, Rajesh M, Czifra N, Harvey-White J, Haská G, Zsengeller Z, Gerard NP Liaudet L, Kunos G. Pharmacological inhibition of CB cannabinoid receptor protects against doxorubicin-induced cardiotoxicity. J Am Coll Cardiol. 2007;50(6):528–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Mukhopadhyay P, Rajesh M, Batkai S, Patel V, Kashiwaya Y, Liaudet L, Evgenov OV, Mackie K, Haská G, Pacher P. CB cannabinoid receptors promote oxidative stress and cell death in murine models of doxorubicin-induced cardiomyopathy and in human cardiomyocytes. Cardiovasc Res. 2010;85(4):773–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mukhopadhyay P, Horvath B, Rajesh M, Matsumoto S, Saito K, Batkai S, Patel V, Tanchian G, Gao RY, Cravatt BF. Fatty acid amide hydrolase is a key regulator of endocannabinoid-induced myocardial tissue injury. Free Radic Biol Med. 2011;50(1):179–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Manjanna K, Shivakumar B, Kumar TP. Microencapsulation: An acclaimed novel drug-delivery system for NSAIDs in arthritis. Crit Rev Ther Drug Carrier Syst. 2010;27(6):509–45. [DOI] [PubMed] [Google Scholar]
  • 110.Srivastava S, Patel S, Singh D, Singh MR. Rationalized insights on causes of rheumatoid arthritis in the elderly and women: Special emphasis on treatment strategies. Crit Rev Ther Drug Carrier Syst. 2017;34(2):97–147. [DOI] [PubMed] [Google Scholar]
  • 111.Lodzki M, Godin B, Rakou L, Mechoulam R, Gallily R, Touitou E. Cannabidiol transdermal delivery and anti-inflammatory effect in a murine model. J Control Release. 2003;93(3):377–87. [DOI] [PubMed] [Google Scholar]
  • 112.Stinchcomb AL, Valiveti S, Hammell DC, Ramsey DR. Human skin permeation of DeltΔ8-tetrahydro-cannabinol, cannabidiol and cannabinol. J Pharm Pharmacol. 2004;56(3):291–7. [DOI] [PubMed] [Google Scholar]
  • 113.Paudel KS, Milewski M, Swadley CL, Brogden NK, Ghosh P, Stinchcomb AL. Challenges and opportunities in dermal/transdermal delivery. Ther Deliv. 2010; 1(1): 109–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Hammell DC, Zhang LP, Ma F, Abshire SM, McIlwrath SL, Stinchcomb AL, Westlund KN. Transdermal cannabidiol reduces inflammation and pain-related behaviours in a rat model of arthritis. Eur J Pain. 2016;20(6):936–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Melzack R, Casey KL. Sensory, motivational, and central control determinants of pain: A new conceptual model. The Skin Senses. 1968;1:423–43. [Google Scholar]
  • 116.Bushnell MC, Ceko M, Low LA. Cognitive and emotional control of pain and its disruption in chronic pain. Nat Rev Neurosci. 2013;14(7):502–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Garg T, Singh O, Arora S, Murthy R. Scaffold: A novel carrier for cell and drug delivery. Crit Rev Ther Drug Carrier Syst. 2012;29(1):1–63. [DOI] [PubMed] [Google Scholar]
  • 118.Piomelli D, Tarzia G, Duranti A, Tontini A, Mor M, Compton TR, Dasse O, Monaghan EP, Parrott JA, Putman D. Pharmacological profile of the selective FAAH inhibitor KDS-4103 (URB597). CNS Drug Rev. 2006;12(1):21–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Russo EB, Guy GW, Robson PJ. Cannabis, pain, and sleep: Lessons from therapeutic clinical trials of Sativex®, a cannabis-based medicine. Chem Biodivers. 2007;4(8):1729–43. [DOI] [PubMed] [Google Scholar]
  • 120.Ahn K, McKinney MK, Cravatt BF. Enzymatic pathways that regulate endocannabinoid signaling in the nervous system. Chem Rev. 2008;108(5):1687–707. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Petrosino S, Di Marzo V. FAAH and MAGL inhibitors: Therapeutic opportunities from regulating endocannabinoid levels. Curr Opin Investig Drugs. 2010;11(1):51–62. [PubMed] [Google Scholar]
  • 122.Jensen B, Chen J, Furnish T, Wallace M. Medical marijuana and chronic pain: A review of basic science and clinical evidence. Curr Pain Headache Rep. 2015;19(10):50. [DOI] [PubMed] [Google Scholar]
  • 123.Kramer JL. Medical marijuana for cancer. CA Cancer J Clin. 2015;65(2):109–22. [DOI] [PubMed] [Google Scholar]
  • 124.Stino AM, Smith AG. Peripheral neuropathy in prediabetes and the metabolic syndrome. J Diabetes Investig. 2017;8(5):646–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Boland EG, Ahmedzai SH. Persistent pain in cancer survivors. Curr Opin Support Palliat Care. 2017;11(3): 181–90. [DOI] [PubMed] [Google Scholar]
  • 126.Pain PP. Persistent pain in cancer survivors: Pathogenesis and treatment options. Pain.2. [Google Scholar]
  • 127.Starobova H, Vetter I. Pathophysiology of chemotherapy-induced peripheral neuropathy. Front Mol Neurosci. 2017;10:174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Gordon-Williams R, Farquhar-Smith P. Recent advances in understanding chemotherapy-induced peripheral neuropathy. F1000Res. 2020;9:F1000 Faculty Rev-177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Visovsky C, Collins M, Abbott L, Aschenbrenner J, Hart C. Putting evidence into practice: Evidence-based interventions for chemotherapy-induced peripheral neuropathy. Clin J Oncol Nursing. 2007;11(6):901–13. [DOI] [PubMed] [Google Scholar]
  • 130.Staff NP, Grisold A, Grisold W, Windebank AJ. Chemotherapy-induced peripheral neuropathy: A current review. Ann Neurol. 2017;81(6):772–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, Colvin LA, Fallon M. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain. 2014;155(12):2461–70. [DOI] [PubMed] [Google Scholar]
  • 132.Stone JB, DeAngelis LM. Cancer-treatment-induced neurotoxicity focus on newer treatments. Nat Rev Clin Oncol. 2016;13(2):92–105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chiorazzi A, Semperboni S, Marmiroli P. Current view in platinum drug mechanisms of peripheral neurotoxicity. Toxics. 2015;3(3):304–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Kelley MR, Jiang Y, Guo C, Reed A, Meng H, Vasko MR. Role of the DNA base excision repair protein, APE1 in cisplatin, oxaliplatin, or carboplatin induced sensory neuropathy. PLoS One. 2014;9(9):e106485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Ta LE, Espeset L, Podratz J, Windebank AJ. Neurotoxicity of oxaliplatin and cisplatin for dorsal root ganglion neurons correlates with platinum DNA binding. Neurotoxicology. 2006;27(6):992–1002. [DOI] [PubMed] [Google Scholar]
  • 136.McWhinney SR, Goldberg RM, McLeod HL. Platinum neurotoxicity pharmacogenetics. Mol Cancer Therap. 2009;8(1):10–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Zajączkowska R, Kocot-Kępska M, Leppert W, Wrzosek A, Mika J, Wordliczek J. Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci. 2019;20(6):1451. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Postma TJ, Van Groeningen CJ, Witjes R, Weerts JGE, Kralendonk JH, Heimans JJ. Neurotoxicity of combination chemotherapy with procarbazine, CCNU and vincristine (PCV) for recurrent glioma. J Neurooncol. 1998;38(1):69–75. [DOI] [PubMed] [Google Scholar]
  • 139.Ziegler D, Hanefeld M, Ruhnau KJ, Meissner HP, Lobisch M, Schütte K, Gries FA. Treatment of symptomatic diabetic peripheral neuropathy with the anti-oxidant alpha-lipoic acid. A 3-week multicentre randomized controlled trial (ALADIN study). Diabetologia. 1995;38(12):1425–33. [DOI] [PubMed] [Google Scholar]
  • 140.Kober KM, Olshen A, Conley YP, Schumacher M, Topp K, Smoot B, Mazor M, Chesney M, Hammer M, Paul SM. Expression of mitochondrial dysfunction-related genes and pathways in paclitaxel-induced peripheral neuropathy in breast cancer survivors. Mol Pain. 2018;14: 1744806918816462. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Krukowski K, Ma J, Golonzhka O, Laumet GO, Gutti T, Van Duzer JH, Mazitschek R, Jarpe MB, Heijnen CJ, Kavelaars A. HDAC6 inhibition effectively reverses chemotherapy-induced peripheral neuropathy. Pain. 2017;158(6):1126–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Flatters SJL, Bennett GJ. Studies of peripheral sensory nerves in paclitaxel-induced painful peripheral neuropathy: Evidence for mitochondrial dysfunction. Pain. 2006;122(3):245–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Ma J, Kavelaars A, Dougherty PM, Heijnen CJ. Beyond symptomatic relief for chemotherapy-induced peripheral neuropathy: Targeting the source. Cancer. 2018;124(11):2289–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Shim HS, Bae C, Wang J, Lee K-H, Hankerd KM, Kim HK, Chung JM, La J-H. Peripheral and central oxidative stress in chemotherapy-induced neuropathic pain. Mol Pain. 2019;15:1744806919840098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Chan SY, Worth R, Ochs S. Block of axoplasmic transport in vitro by vinca alkaloids. J Neurobiol. 1980;11(3):251–64. [DOI] [PubMed] [Google Scholar]
  • 146.Starobova H, Mueller A, Allavena R, Lohman R-J, Sweet MJ, Vetter I. Minocycline prevents the development of mechanical allodynia in mouse models of vincristine induced peripheral neuropathy. Front Neurosci. 2019;13:653. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Ramchandren S, Leonard M, Mody RJ, Donohue JE, Moyer J, Hutchinson R, Gurney JG. Peripheral neuropathy in survivors of childhood acute lymphoblastic leukemia. J Peripher Nerv Syst. 2009;14(3):184–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Toopchizadeh V, Barzegar M, Rezamand A, Feiz AH. Electrophysiological consequences of vincristine contained chemotherapy in children: A cohort study. J Pediatr Neurol. 2009;7(4):351–6. [Google Scholar]
  • 149.Seretny M, Currie GL, Sena ES, Ramnarine S, Grant R, MacLeod MR, Colvin LA, Fallon M. Incidence, prevalence, and predictors of chemotherapy-induced peripheral neuropathy: A systematic review and meta-analysis. Pain. 2014;155(12):2461–70. [DOI] [PubMed] [Google Scholar]
  • 150.Lavoie Smith EM, Li L, Chiang C, Thomas K, Hutchinson RJ, Wells EM, Ho RH, Skiles J, Chakraborty A, Bridges CM. Patterns and severity of vincristine-induced peripheral neuropathy in children with acute lymphoblastic leukemia. J Peripher Nerv Syst. 2015;20(1):37–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Mora E, Smith EML, Donohoe C, Hertz DL. Vincristine-induced peripheral neuropathy in pediatric cancer patients. Am J Cancer Res. 2016;6(11):2416. [PMC free article] [PubMed] [Google Scholar]
  • 152.Barton DL, Wos EJ, Qin R, Mattar BI, Green NB, Lanier KS, Bearden JD, Kugler JW, Hoff KL, Reddy PS. A double-blind, placebo-controlled trial of a topical treatment for chemotherapy-induced peripheral neuropathy: NCCTG trial N06CA. Support Care Cancer. 2011;19(6):833–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Mokhtar GM, Shaaban SY, Elbarbary NS, Fayed WA. Atrial to assess the efficacy of glutamic acid in prevention of vincristine-induced neurotoxicity in pediatric malignancies: A pilot study. J Pediatr Hematol Oncol. 2010;32(8):594–600. [DOI] [PubMed] [Google Scholar]
  • 154.Hayek ME, Trad O, Jamil A. Vincristine-induced urinary bladder paralysis. J Pediatr Hematol Oncol. 2005;27(5):286–7. [DOI] [PubMed] [Google Scholar]
  • 155.Kellie SJ, Barbaric D, Koopmans P, Earl J, Carr DJ, de Graaf SSN. Cerebrospinal fluid concentrations of vincristine after bolus intravenous dosing: A surrogate marker of brain penetration. Cancer. 2002;94(6):1815–20. [DOI] [PubMed] [Google Scholar]
  • 156.Adhikari S, Dongol RM, Hewett Y, Shah BK. Vincristine-induced blindness: A case report and review of literature. Anticancer Res. 2014;34(11):6731–3. [PubMed] [Google Scholar]
  • 157.Sanderson PA, Kuwabara T, Cogan DG. Optic neuropathy presumably caused by vincristine therapy. Am J Ophthalmol. 1976;81(2):146–50. [DOI] [PubMed] [Google Scholar]
  • 158.Robertson GL, Bhoopalam N, Zelkowitz LJ. Vincristine neurotoxicity and abnormal secretion of antidiuretic hormone. Arch Intern Med. 1973;132(5):717–20. [PubMed] [Google Scholar]
  • 159.Watterworth B, Wright TB. Diabetic peripheral neuropathy. In: Abd-Elsayed A, editor. Pain. Cham, Switzerland: Springer; 2019. p. 911–3. [Google Scholar]
  • 160.Bheereddy P, Yerra VG, Kalvala AK, Sherkhane B, Kumar A. SIRT1 activation by polydatin alleviates oxidative damage and elevates mitochondrial biogenesis in experimental diabetic neuropathy. Cell Mol Neurobiol. 2021;41(7):1563–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Shakher J, Stevens MJ. Update on the management of diabetic polyneuropathies. Diabetes Metab Syndr Obes. 2011;4:289–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Tesfaye S, Selvarajah D, Gandhi R, Greig M, Shillo P, Fang F, Wilkinson ID. Diabetic peripheral neuropathy may not be as its name suggests: Evidence from magnetic resonance imaging. Pain. 2016;157(Suppl 1):S72–80. [DOI] [PubMed] [Google Scholar]
  • 163.Greene DA, Stevens MJ, Obrosova I, Feldman EL. Glucose-induced oxidative stress and programmed cell death in diabetic neuropathy. Eur J Pharmacol. 1999;375(1–3):217–23. [DOI] [PubMed] [Google Scholar]
  • 164.Kalvala AK, Kumar R, Sherkhane B, Gundu C, Arruri VK, Kumar A. Bardoxolone methyl ameliorates hyperglycemia induced mitochondrial dysfunction by activating the keap1-Nrf2-ARE pathway in experimental diabetic neuropathy. Mol Neurobiol. 2020;57(8):3616–31. [DOI] [PubMed] [Google Scholar]
  • 165.Kalvala AK, Yerra VG, Kumar A. LONP1 induction by SRT1720 attenuates mitochondrial dysfunction against high glucose induced neurotoxicity in PC12 cells. Toxicol In Vitro. 2020;62: 104695. [DOI] [PubMed] [Google Scholar]
  • 166.Nagamatsu M, Nickander KK, Schmelzer JD, Raya A, Wittrock DA, Tritschler H, Low PA. Lipoic acid improves nerve blood flow, reduces oxidative stress, and improves distal nerve conduction in experimental diabetic neuropathy. Diabetes Care. 1995;18(8): 1160–7. [DOI] [PubMed] [Google Scholar]
  • 167.Garrett NE, Malcangio M, Dewhurst M, Tomlinson DR. Alpha-lipoic acid corrects neuropeptide deficits in diabetic rats via induction of trophic support. Neurosci Lett. 1997;222(3):191–4. [DOI] [PubMed] [Google Scholar]
  • 168.Aruoma OI, Halliwell B, Hoey BM, Butler J. The antioxidant action of taurine, hypotaurine and their metabolic precursors. Biochem J. 1988;256(1):251–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Cameron NE, Cotter MA, Maxfield EK. Anti-oxidant treatment prevents the development of peripheral nerve dysfunction in streptozotocin-diabetic rats. Diabetologia. 1993;36(4):299–304. [DOI] [PubMed] [Google Scholar]
  • 170.Vučković S, Srebro D, Vujović KS, Vučetić Č, Prostran M. Cannabinoids and pain: New insights from old molecules. Front Pharmacol. 2018;9:1259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.Rock EM, Limebeer CL, Parker LA. Effect of cannabidiolic acid and Δ9-tetrahydrocannabinol on carrageenan—induced hyperalgesia and edema in a rodent model of inflammatory pain. Psychopharmacology. 2018;235(11):3259–71. [DOI] [PubMed] [Google Scholar]
  • 172.Xiong W, Cheng K, Cui T, Godlewski G, Rice KC, Xu Y, Zhang L. Cannabinoid potentiation of glycine receptors contributes to cannabis-induced analgesia. Nat Chem Biol. 2011;7(5):296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Lazic A, Popović J, Paunesku T, Woloschak GE, Stevanović M. Insights into platinum-induced peripheral neuropathy-current perspective. Neural Regen Res. 2020;15(9):1623–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Ward SJ, McAllister SD, Kawamura R, Murase R, Neelakantan H, Walker EA. Cannabidiol inhibits paclitaxel-induced neuropathic pain through 5-HT(1A) receptors without diminishing nervous system function or chemotherapy efficacy. Br J Pharmacol. 2014;171(3):636–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Mücke M, Phillips T, Radbruch L, Petzke F, Häuser W. Cannabis-based medicines for chronic neuropathic pain in adults. Cochrane Databsae Syst Rev. 2018;3(3):CD012182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Aviram J, Samuelly-Leichtag G. Efficacy of cannabis-based medicines for pain management: A systematic review and meta-analysis of randomized controlled trials. Pain Physician. 2017;20(6):E755–96. [PubMed] [Google Scholar]
  • 177.Meissner H, Cascella M. Cannabidiol (CBD). In: StatPearls. Treasure Island, FL: StatPearls Publishing; 2022. [PubMed] [Google Scholar]
  • 178.Silvestro S, Mammana S, Cavalli E, Bramanti P, Mazzon E. Use of cannabidiol in the treatment of epilepsy: Efficacy and security in clinical trials. Molecules. 2019;24(8):1459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Asadi-Pooya AA. Lennox-Gastaut syndrome: A comprehensive review. Neurol Sci. 2018;39(3):403–14. [DOI] [PubMed] [Google Scholar]
  • 180.Lattanzi S, Brigo F, Trinka E, Zaccara G, Striano P, Del Giovane C, Silvestrini M. Adjunctive cannabidiol in patients with dravet syndrome: A systematic review and meta—analysis of efficacy and safety. CNS Drugs. 2020;34(3):229–41. [DOI] [PubMed] [Google Scholar]
  • 181.Devinsky O, Cross JH, Laux L, Marsh E, Miller I, Nabbout R Scheffer IE, Thiele EA, Wright S. Trial of cannabidiol for drug-resistant seizures in the Dravet syndrome. N Engl J Med. 2017;376(21): 2011–20. [DOI] [PubMed] [Google Scholar]
  • 182.Devinsky O, Patel AD, Cross JH, Villanueva V, Wirrell EC, Privitera M, Greenwood SM, Roberts C, Checketts D, VanLandingham KE. Effect of cannabidiol on drop seizures in the Lennox-Gastaut syndrome. N Engl J Med. 2018;378(20):1888–97. [DOI] [PubMed] [Google Scholar]
  • 183.Thiele EA, Marsh ED, French JA, Mazurkiewicz-Beldzinska M, Benbadis SR, Joshi C, Lyons PD, Taylor A, Roberts C, Sommerville K. Cannabidiol in patients with seizures associated with LennoxGastaut syndrome (GWPCARE4): A randomised, double-blind, placebo-controlled phase 3 trial. Lancet. 2018;391(10125):1085–96. [DOI] [PubMed] [Google Scholar]
  • 184.Laczkovics C, Kothgassner OD, Felnhofer A, Klier CM. Cannabidiol treatment in an adolescent with multiple substance abuse, social anxiety and depression. Neuropsychiatr. 2021;35(1):31–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Levinsohn EA, Hill KP. Clinical uses of cannabis and cannabinoids in the United States. J Neurol Sci. 2020;411:116717. [DOI] [PubMed] [Google Scholar]
  • 186.Crippa JA, Guimaraes FS, Campos AC, Zuardi AW. Translational investigation of the therapeutic potential of cannabidiol (CBD): Toward a new age. Front Immunol. 2018;8:2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Watt G, Karl T. In vivo evidence for therapeutic properties of cannabidiol (CBD) for Alzheimer’s disease. Front Pharmacol. 2017;8:20. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Cannabinoids and an Anti-inflammatory diet for the treatment of neuropathic pain after spinal cord injury. [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT04057456?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=1. [DOI] [PubMed]
  • 189.Cannabinoids effects on the pain modulation system, [cited 2020 Aug 4], Available from: https://clinicaltrials.gov/ct2/show/NCT02560545?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=2.
  • 190.Effect of cannabinoids on spasticity and neuropathic pain in spinal cord injured persons, [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT01222468?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=3
  • 191.Nabilone for the treatment of phantom limb pain. [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT00699634?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=4.
  • 192.Sativex for treatment of chemotherapy induced neuropathic pain. [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT00872144?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=5.
  • 193.Efficacy study of nabilone in the treatment of diabetic peripheral neuropathic pain, [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT01035281?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=6.
  • 194.Effect of cannabis and endocannabinoids on HTV neuropathic pain, [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT03099005?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=7.
  • 195.An observational post-marketing safety registry of Sativex®. [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT02073474?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=8.
  • 196.Efficacy and safety of the pain relieving effect of dronabinol in central neuropathic pain related to multiple sclerosis. [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT00959218?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=9.
  • 197.Trial of dronabinol and vaporized cannabis in neuropathic low back pain. [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT02460692?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=1&rank=10.
  • 198.Effect of delta-9-tetrahydrocannabinol on the prevention of chronic pain in patients with acute CRPS (ETIC-study). [cited 2020 Aug 4]. Available from: https://clinicaltrials.gov/ct2/show/NCT00377468?term=Cannabinoids&cond=Pain%2C+Neuropathic&draw=3&rank=11.
  • 199.Study to evaluate the efficacy of dronabinol (Marinol) as add-on therapy for patients on opioids for chronic pain. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT00153192?term=cannabinoids&cond=Pain%2C+Chronic&draw=2&rank=4.
  • 200.Vaporized cannabis for chronic pain associated with sickle cell disease (Cannabis-SCD). [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT01771731?term=cannabinoids&cond=Pain%2C+Chronic&draw=2&rank=5.
  • 201.A study to compare sublingual cannabis based medicine extracts with placebo to treat brachial plexus injury pain. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT01606189?term=Cannabinoids&cond=Pain%2C+Chronic&draw=2&rank=8.
  • 202.Pain research: Innovative strategies with marijuana (PRISM). [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT03522324?term=cannabinoids&cond=Pain%2C+Chronic&draw=2&rank=1.
  • 203.Opioid and cannabinoid pharmacokinetic interactions. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT00308555?term=Cannabinoids&cond=Pain%2C+Chronic&draw=2&rank=9.
  • 204.Safety and efficacy of medical cannabis oil in the treatment of patients with chronic pain, [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT03337503?term=Cannabinoids&cond=Pain%2C+Chronic&draw=2&rank=10.
  • 205.Efficacy of ultra-micronized palmitoylethanolamide (Um-PEA) in geriatric patients with chronic pain. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT02699281?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=11. [DOI] [PMC free article] [PubMed]
  • 206.Treatment of chronic pain with cannabidiol (CBD) and delta-9-tetrahydrocannabinol (THC). [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT03215940?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=12.
  • 207.Dronabinol opioid sparing evaluation (DOSE) trial. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT03766269?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=13.
  • 208.(Namisol®) in patients with persistent postsurgical abdominal pain. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT01562483?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=15.
  • 209.Δ9-THC (Namisol®) in chronic pancreatitis patients suffering from persistent abdominal pain, [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT01551511?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=16.
  • 210.Outcomes mandate national integration with cannabis as medicine (OMNI-Can). [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT03944447?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=17.
  • 211.Use of the cannabinoid nabilone for the promotion of sleep in chronic, non-malignant pain patients. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT00384410?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=18.
  • 212.Cannabis oil for chronic non-cancer pain treatment. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT03635593?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=19.
  • 213.Efficacy study of Δ9-THC to treat chronic abdominal pain (delta-pain). [cited 2020 Aug 22], Available from: https://clinicaltrials.gov/ct2/show/NCT01318369?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=20.
  • 214.Supporting effect of dronabinol on behavioral therapy in fibromyalgia and chronic back pain, [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT00176163?term=Cannabinoids&cond=Pain%2C+Chronic&draw=1&rank=21.
  • 215.Palmitoylethanolamide for post-operative pain prevention (PEAforCPSP). [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT01491191?term=Cannabinoids&cond=Pain%2C+Chronic&draw=3&rank=22.
  • 216.Nabilone versus amitriptyline in improving quality of sleep in patients with fibromyalgia, [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT00381199?term=Cannabinoids&cond=Pain%2C+Chronic&draw=1&rank=23.
  • 217.Efficacy of palmitoylethanolamide-polydatin combination on chronic pelvic pain in patients with endometriosis. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT02372903?term=Cannabinoids&cond=Pain%2C+Chronic&draw=1&rank=24.
  • 218.Nabilone use for acute pain in inflammatory bowel disease patients, [cited 2020 Aug 22], Available from: https://clinicaltrials.gov/ct2/show/NCT03422861?term=Cannabinoids&cond=Pain%2C+Acute&draw=2&rank=1.
  • 219.Evaluation of dronabinol for acute pain following traumatic injury. [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT03928015?term=Cannabinoids&cond=Pain%2C+Acute&draw=1&rank=2.
  • 220.Cannabidiol, morphine, pain (CMP). [cited 2020 Aug 22]. Available from: https://clinicaltrials.gov/ct2/show/NCT04030442?term=cannabinoids&cond=Pain%2C+Chronic&draw=2&rank=6.
  • 221.Grotenhermen F. Pharmacokinetics and pharmacodynamics of cannabinoids. Clin Pharmacokinet. 2003;42(4):327–60. [DOI] [PubMed] [Google Scholar]
  • 222.Deiana S, Watanabe A, Yamasaki Y, Amada N, Arthur M, Fleming S, Woodcock H, Dorward P. Pigliacampo B, Close S. Plasma and brain pharmacokinetic profile of cannabidiol (CBD), cannabidivarine (CBDV), Δ9-tetrahydrocannabivarin (THCV) and cannabigerol (CBG) in rats and mice following oral and intraperitoneal administration and CBD action on obsessive-compulsive behaviour. Psychopharmacology. 2012;219(3): 859–73. [DOI] [PubMed] [Google Scholar]
  • 223.Devinsky O, Cilio MR, Cross H, Fernandez-Ruiz J, French J, Hill C, Katz R, Di Marzo V, Jutrasâ Aswad D, Notcutt WG. Cannabidiol: Pharmacology and potential therapeutic role in epilepsy and other neuropsychiatric disorders. Epilepsia. 2014;55(6):791–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Samara E, Bialer M, Mechoulam R. Pharmacokinetics of cannabidiol in dogs. Drug Metab Dispos. 1988;16(3):469–72. [PubMed] [Google Scholar]
  • 225.Bartner LR, McGrath S, Rao S, Hyatt LK, Wittenburg LA. Pharmacokinetics of cannabidiol administered by 3 delivery methods at 2 different dosages to healthy dogs. Can J Vet Res. 2018;82(3): 178–83. [PMC free article] [PubMed] [Google Scholar]
  • 226.El-Alfy AT, Ivey K, Robinson K, Ahmed S, Radwan M, Slade D, Khan I, ElSohly M, Ross S. Antidepressant-like effect of Δ9-tetrahydrocannabinol and other cannabinoids isolated from Cannabis sativa L. Pharmacol Biochem Behav. 2010;95(4):434–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 227.Sales AJ, Fogafa MV, Sartim AG, Pereira VS, Wegener G, Guimarães FS, Joca SMR. Cannabidiol induces rapid and sustained antidepressant-like effects through increased BDNF signaling and synaptogenesis in the prefrontal cortex. Mol Neurobiol. 2019;56(2):1070–81. [DOI] [PubMed] [Google Scholar]
  • 228.de Mello Schier AR, de Oliveira Ribeiro NP, Coutinho DS, Machado S, Arias-Carrión O, Crippa JA, Zuardi AW, Nardi AE, Silva AC. Antidepressant-like and anxiolytic-like effects of cannabidiol: A chemical compound of Cannabis sativa. CNS Neurol Disord Drug Targets. 2014;13(6):953–60.24923339 [Google Scholar]
  • 229.Zanelati TV, Biojone C, Moreira FA, Guimarães FS, Joca SRL. Antidepressant-like effects of cannabidiol in mice: Possible involvement of 5-HT1A receptors. Br J Pharmacol. 2010;159(1): 122–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Schonhofen P, Bristot IJ, Crippa JA, Hallak JEC, Zuardi AW, Parsons RB, Klamt F. Cannabinoid-based therapies and brain development: Potentid harmful effect of early modulation of the endocannabinoid system. CNS Drugs. 2018;32(8):697–712. [DOI] [PubMed] [Google Scholar]
  • 231.Ravula A, Chandasana H, Setlow B, Febo M, Bruijnzeel AW, Derendorf H. Simultaneous quantification of cannabinoids tetrahydrocannabinol, cannabidiol and CB1 receptor antagonist in rat plasma: An application to characterize pharmacokinetics after passive cannabis smoke inhalation and co-administration of rimonabant. J Pharm Biomed Anal. 2018;160:119–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 232.Schlienz NJ, Lee DC, Stitzer ML, Vandrey R. The effect of high-dose dronabinol (oral THC) maintenance on cannabis self-administration. Drug Alcohol Depend. 2018;187:254–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Scully C. Cannabis; adverse effects from an oromucosal spray. Br Dental J. 2007;203(6):E12. [DOI] [PubMed] [Google Scholar]
  • 234.Pellesi L, Licata M, Verri P, Vandelli D, Palazzoli F, Marchesi F, Cainazzo MM, Pini LA, Guerzoni S. Pharmacokinetics and tolerability of oral cannabis preparations in patients with medication overuse headache MOH-a pilot study. Eur J Clin Pharmacol. 2018;74(11):1427–36. [DOI] [PubMed] [Google Scholar]
  • 235.Badowski ME. A review of oral cannabinoids and medical marijuana for the treatment of chemotherapy-induced nausea and vomiting: A focus on pharmacokinetic variability and pharmacodynamics. Cancer Chemother Pharmacol. 2017;80(3):441–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Cao D, Srisuma S, Bronstein AC, Hoyte CO. Characterization of edible marijuana product exposures reported to United States poison centers. Clin Toxicol. 2016;54(9):840–6. [DOI] [PubMed] [Google Scholar]
  • 237.Xu C, Chang T, Du Y, Yu C, Tan X, Li X. Pharmacokinetics of oral and intravenous cannabidiol and its antidepressant-like effects in chronic mild stress mouse model. Environ Toxicol Pharmacol. 2019;70:103202. [DOI] [PubMed] [Google Scholar]

RESOURCES