Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2024 Mar 18.
Published in final edited form as: Annu Rev Med. 2023 Aug 15;75:353–367. doi: 10.1146/annurev-med-052422-020627

Adverse Impact of Marijuana on Human Health

Mark Chandy 1,2,3,4, Masataka Nishiga 1,2,3, Thomas Wei 1,2,3,5, Kari Nadeau 6, Joseph C Wu 1,2,3,*
PMCID: PMC10947506  NIHMSID: NIHMS1971329  PMID: 37582489

Abstract

Cannabis, the most commonly used recreational drug, is illicit in many areas of the world. With increasing decriminalization and legalization of cannabis, its use is increasing in the United States and other countries. The adverse effects of cannabis are unclear because its status as a Schedule 1 drug in the United States restricts research on the drug. Despite a paucity of data, cannabis is commonly perceived as a benign drug or a panacea. However, recent studies have shown that cannabis has adverse cardiovascular and pulmonary effects and is linked with malignancy. Moreover, case reports have shown an association between cannabis use and neuropsychiatric disorders. With increasing availability, cannabis misuse by minors has led to increasing incidences of overdose and toxicity, and cannabis intoxication is difficult to detect and may be linked to impaired driving and motor vehicle accidents. Overall, cannabis use is on the rise, and adverse effects are becoming apparent in clinical datasets.

Keywords: Cannabis, Cardiovascular disease, Pulmonary disease, Cannabis induced psychosis, Cannabis use disorder

Introduction

Cannabis has been cultivated by humans for thousands of years for recreational and medicinal uses (1). Consumed in various modalities, including smoking, hookah, vaping, or edibles, cannabis is the most commonly used recreational drug and is illicit in many areas of the world (2). With decriminalization and legalization, cannabis use has increased worldwide (3, 4).

There are over 500 chemicals in cannabis, and the chief psychoactive component, delta-9-tetrahydrocannabinol, Δ9-THC, is the most abundant cannabinoid (5). However, there are over 100 other cannabinoids that are structurally similar to Δ9-THC, including cannabidiol (CBD), the second most abundant component, which is non-psychotropic. The effects of cannabinoids are mediated by two cannabinoid receptors, cannabinoid receptor 1 (CB1) and cannabinoid receptor 2 (CB2), which belong to the G protein-coupled receptor (GPCR) superfamily (6). CB1, the most abundant GPCR in the mammalian brain, regulates the psychoactive effects of Δ9-THC (Figure 1). The CB1 receptor is also expressed in other peripheral cells, tissues, and organs such as the heart, vasculature, and smooth muscle. The CB2 receptor is expressed in immune cells, particularly in macrophages. The cannabinoid receptors modulate adenylyl cyclase, ion channels, mitogen-activated protein kinases (p44/42 MAPKs, p38, ERK, and JNK), and ceramide signaling (7).

Figure 1. Cannabinoids and cognate receptors.

Figure 1.

A) Cannabinoid receptor 1 (CB1) agonists. The molecular structure of three representative CB1 agonists that cause psychoactive effects: delta-9-tetrahydrocannabinol (Δ9-THC), hexahydrocannabinol (HHC), and delta-8-tetrahydrocannabinol (Δ8-THC). B) Cannabinoid receptor 2 (CB2) agonists. The molecular structure of three CB2 agonists: including beta-caryophyllene (BCP), 2-iodo-5-nitrophenyl)(1-((1-methylpiperidin-2-yl)methyl)-1H-indol-3-yl)methanone (AM 1261), and (6AR,10AR)-3-(1,1-Dimethylbutyl)-6A,7,10,10A-tetrahydro-6,6,9-trimethyl-6H-dibenzo[B,D]pyran (JWH 133). C) CB1 activation causes inflammation and oxidative stress via multiple signaling pathways. Δ9-THC binding to the CB1 receptor causes activation of the G-protein coupled receptor (GPCR). The Gi/o complex dissociated into the αi, βγ, and β arrestin subunits. The αi subunit inhibits adenylyl cyclase causing decreased cyclic AMP production (cAMP) and prevents protein kinase A (PKA) phosphorylation, which modulates transcription. The βγ subunit activates the mitogen activated pathway (MAP) and p38, which activates NF-κβ thus leading to activation of inflammatory gene and down regulation of oxidative-stress protective genes. The βγ subunit simultaneously affects voltage gated calcium channels (CGGC), G-protein gated potassium channels (GIRK), and PI3K/AKT pathway. (D) CB2 activation prevents inflammation and oxidative stress. BCP binding to the CB2 receptor causes activation of the GPCR. The Gai/o complex dissociates into the αi, βγ, and β arrestin subunits. Similar to CB1, the αi subunit inhibits adenylyl cyclase causing decreased cyclic AMP production (cAMP) and prevents protein kinase A (PKA) phosphorylation, which modulates transcription. The βγ subunit activates the mitogen activated pathway (MAP) and p38. However, CB2 activation prevents NF-κβ translocation to the nucleus, thus blocking the expression of inflammatory genes and down regulation of oxidative-stress protective genes. The βγ subunit simultaneously affects voltage gated calcium channels (CGGC), G-protein gated potassium channels (GIRK), and the PI3K/AKT pathway. Created with BioRender.com.

Beneficial Effects of Cannabis

A recent systematic review and metanalysis found that cannabinoids such as Δ9-THC and CBD might be of benefit for chronic pain, spasticity, epilepsy, cancer and neurological disorders (8). There are several reports describing the anti-inflammatory effects of cannabis and Δ9-THC (9). Δ9-THC has been reported to have apoptotic effects on immune cells (10, 11). Inflammatory cytokine production is also reduced after treatment with Δ9-THC (12, 13). In neurological disorders such as multiple sclerosis, Δ9-THC is also thought to exert beneficial effects due to immunosuppression (14). Indeed, several reports show that Δ9-THC can induce cytotoxicity in cancer, including acute lymphoblastic leukemia (15, 16).

CBD is reported to have anti-inflammatory properties (17) and might ameliorate inflammatory arthropathies (18). CBD is also described as having a neuroprotective effect (19) and is used for neurodegenerative disorders such as multiple sclerosis, Parkinson’s disease, and Alzheimer’s disease (20, 21). Cannabinoids are known to induce sedation and analgesia and could be an alternative to opioids (22, 23). The adverse effects of opioids include constipation, overdose causing bradypnea, bradycardia, and death, as well as cardiovascular disease. Opioid misuse is exacerbated by the propensity for addiction and highlighted by the ongoing and unrelenting opioid crisis.

Medicinal marijuana has been used in HIV and cancer patients (24). Synthetic Δ9-THC in several forms has been approved by the U.S. Food and Drug Administration (FDA) for treating chemotherapy-induced nausea and vomiting, as well as for HIV and anorexia: Marinol® (dronabinol), Syndros® (dronabinol), and Cesamet® (nabilone). More recently, synthetic cannabinol, Epidiolex®, has been approved for treating epilepsy in children (25). However, while medicinal cannabis might be beneficial, its adverse effects are becoming evident with increased usage (24; 26).

The assumption that marijuana has pleiotropic benefits without any adverse consequences lacks supporting evidence in clinical practice. Some cannabinoids, such as cannabinol (CBD), might have uses such as its potential anti-inflammatory effects. However, the overwhelming evidence from in vitro, in vivo, and clinical studies indicated that Δ9-THC is pro-inflammatory concerning overall cardiovascular disease risk and likely a precipitant for myocardial infarction and increased mortality.

Adverse Effects of Cannabis:

Heart:

Epidemiological studies support the link between recreational cannabis use and heart disease, generally finding that cannabis increases the risk of cardiovascular disease, cardiomyopathy, and arrhythmias (2, 27, 28). Medical marijuana and synthetic cannabinoids are also associated with adverse cardiovascular effects (24, 26).

Cardiovascular:

Inflammation plays a central role in the pathophysiology of atherosclerosis (29, 30). Chronic inflammation is speculated to accelerate atherosclerosis in diseases such as HIV, rheumatoid arthritis, and systemic lupus erythematosus (31-33). Δ9-THC has been described as promoting inflammation and oxidative stress via the CB1 receptor, MAP kinase activation, and NF-κβ pathways.

As the most abundantly expressed GPCR in the central nervous system, CB1 is also expressed in peripheral cells, tissues, and organs such as in the heart and vasculature, and has been implicated in atherosclerosis (34). CB1 activation occurs via the MAP kinase pathway, which causes oxidative stress, inflammation, and cell death in human coronary artery endothelial cells (35, 36). In contrast, cannabinoid receptor 2 (CB2) while also expressed in the vasculature is anti-atherogenic (7). Preclinical studies have found that CB1 activation mediates increased oxidative stress and inflammation, which are implicated in diabetic retinopathy, cardiomyopathy, and endothelial dysfunction (36-38). Animal studies have shown a causal link between Δ9-THC via the atherogenic CB1 receptor (28, 39). The loss of the atheroprotective CB2 receptor exacerbates the progression of atherosclerosis (40, 41).

Historically, clinical associations between marijuana and cardiovascular disease were limited to case reports or case series (28), and these studies showed that even low doses of Δ9-THC are associated with adverse cardiovascular events (42, 43). A meta-analysis found an association between marijuana and cardiovascular disease. Frost et al. followed patients who are chronic marijuana users and found increased mortality (44). The landmark study by DeFilippis et al. showed a significant association between marijuana and cardiovascular disease in patients with index myocardial infarction (27). Those using cannabis had a lower incidence of cardiovascular risk factors such as hypertension, diabetes, and hyperlipidemia when compared to non-users. However, after adjusting for age, sex, diabetes, hypertension, peripheral vascular disease, smoking, HDL-C, triglycerides, revascularization, creatinine, medications at discharge, and length of stay, the adjusted hazard ratio was 2.09 (95% CI: 1.25 to 3.50; p = 0.005) for marijuana. The confounder of cigarette smoking is also noted in the DeFilippis et al. study, with participants significantly more likely to use tobacco (70.3% vs. 49.1%; p < 0.001). However, when adjusted for baseline characteristics, including cigarette smoking, the DeFilippis et al. study showed that marijuana had a hazard ratio of 2.09 (95% CI: 1.25 to 3.50; p = 0.005). While cigarette smoking was strongly linked to marijuana use, the hazard ratio for marijuana and cardiovascular events was higher than smoking tobacco cigarettes alone. Compared to a healthy non-smoker, the adjusted hazard ratio in a current smoker was 1.7 (95% CI: 1.3–2.2) for all-cause cardiovascular disease (45). The study was conducted between 2000 and 2016 when recreational cannabis use was illicit in Massachusetts. Despite marijuana being illegal and concerns about participants not disclosing cannabis use, an increased incidence of cardiovascular events with a worse prognosis was observed among cannabis users.

In a recent study, Wei et al. described the adverse effect of Δ9-THC on the cardiovascular system using data from the UK biobank, as well as from in vitro and in vivo models (46). Previous studies that showed a link between cannabis and cardiovascular disease were small and retrospective (27). The UK Biobank analysis found an association between cannabis use and myocardial infarction. The largest prospective cohort study to date, the UK Biobank, has genetic and phenotypic data on 500,000 individuals ages 40 to 69 (47), thus providing a unique opportunity to characterize relationships among clinical phenotypes. The UK Biobank analysis revealed an increased incidence of myocardial infarction for cannabis users under age 50 (0.53% vs. 0.45%). Prior studies demonstrating an association between cannabis use and myocardial infarction were conducted on individuals who developed myocardial infarction before age 50 (27). When controlling for age, body mass index (BMI), and sex, a logistic regression model developed to determine if cannabis use is a predictive factor for the development of myocardial infarction, demonstrated that cannabis use was a statistically significant positive predictor for myocardial infarction. O-link analysis of blood from acute marijuana smokers found that 13 inflammatory cytokines associated with cardiovascular disease were elevated (46).

Using induced pluripotent stem cell (iPSC) disease modeling (48), Wei et al. discovered that Δ9-THC caused toxicity, inflammation, and oxidative stress in iPSC-derived endothelial cells. Using siRNA and CRISPR-Cas9 knock down of CB1 expression, the effects of Δ9-THC were shown to be dependent on the CB1 receptor. The adverse effects of Δ9-THC were attenuated by CB1 antagonists such as rimonabant, which is associated with psychiatric side effects. However, virtual ligand modeling discovered that genistein is also a CB1 antagonist and could attenuate Δ9-THC mediated inflammation and oxidative stress in iPSC-derived endothelial cells. Previous studies by Steffens et al. showed that low dose Δ9-THC at (1 mg/kg P.O.) was associated with reduced progression of atherosclerosis in LDLR−/− mice (49). At low doses, the cmax of Δ9-THC was 6 ng/ml and was an agonist of the CB2 receptor; CB2 antagonists abrogated these effects. However, in a 3% marijuana joint, the cmax concentration of Δ9-THC was 130 ng/ml. Moreover, marijuana is now cultivated to produce higher concentrations of Δ9-THC, and e-cigarettes allow for vaping up to 85% Δ9-THC. Using the LDLR−/− and ApoE−/− mouse model, Wei et al. showed that Δ9-THC at 1 mg/kg I.P. achieved a cmax dose of 130 ng/ml caused endothelial dysfunction and atherosclerosis. More importantly, genistein ameliorated Δ9-THC-mediated inflammation, oxidative stress, and inflammation. This study was significant as it provided a mechanistic link between Δ9-THC and atherosclerotic cardiovascular disease.

Cardiomyopathy:

There are case reports and case series describing cannabis-induced cardiomyopathy (50). The link between cardiovascular disease and cannabis suggests that ischemic cardiomyopathy will increase with cannabis use. However, preclinical evidence that cannabinoids cause cardiotoxicity is conflicting. Rajesh et al. found that CB1 activation promotes the pathogenesis of diabetic cardiomyopathy in a mouse model of diabetic cardiomyopathy (37). However, Wei et al. found Δ9-THC had no toxicity in either primary cardiomyocytes, iPSC-derived cardiomyocytes, or in a mouse model (51). Δ9-THC had minimal or no effect on contractility or beat rate in engineered heart cells composed of iPSC-derived cardiomyocytes and endothelial cells. RNA sequencing data sets found no significant effects on iPSC-derived cardiomyocytes or endothelial cells. Moreover, C57BL/6J mice exposed to 1 mg/kg Δ9-THC via intraperitoneal delivery for 1 month showed no evidence of cardiovascular dysfunction as assessed by echocardiography that interrogated systolic function and diastolic function using speckle tracking strain imaging.

Arrhythmias:

Cannabis is associated with both supraventricular and ventricular arrhythmia. From multiple animal models and clinical data, the immediate effect of cannabis is tachycardia and hypertension, followed by bradycardia and hypotension (52). Despite lower blood pressure, cannabis users are at risk for dysrhythmias, including atrial fibrillation, atrial flutter, atrioventricular block, premature ventricular contractions, premature atrial contractions, ventricular tachycardia, and ventricular fibrillation (53). With legalization and increased use of cannabis, cannabis use disorder is now associated with more arrhythmias, such as atrial fibrillation, requiring hospitalization and placing patients at a higher risk of stroke and embolic events (54, 55). Interestingly, the arrhythmias are more common in younger patients with cannabis use disorder suggesting the effects are due to the toxicity of cannabis. The precise mechanisms of the cannabis-induced arrhythmias are unclear and therefore merit further investigation given the severe consequences of dysthymias, such as atrial fibrillation causing stroke with potentially significant changes in quality of life for younger users.

Pulmonary:

Widespread use of marijuana has highlighted the need to learn more about its potential deleterious side effects, particularly those affecting the respiratory system. Smoking is a major cause of cardiopulmonary disease, the leading cause of death worldwide (56). The harm from conventional cigarettes is from the combustion of tobacco products, exacerbated by long-term use due to nicotine addiction, which can lead to acute and long-term adverse changes in the lung, including chronic obstructive lung disease, emphysema, idiopathic pulmonary fibrosis, and pulmonary hypertension (57). Moreover, smoking cannabis increases the respiratory burden of carbon monoxide and tar compared to traditional cigarettes (58). Cannabis and tobacco are often consumed together (59). Wu et al. speculate that by mixing tobacco and cannabis, smokers can inhale more deeply and experience enhanced psychoactive effects and increased acute exposure to toxins (60). With deeper inhalation, cannabis users are at a higher risk of developing COPD and experiencing a more rapid decline in pulmonary function (61). The combined or synergistic effects of nicotine and Δ9-THC on the cardiopulmonary system are unclear and need further investigation. While the detrimental effects of conventional cigarette smoke are now well documented, the toxicities of e-cigarettes remain poorly understood (62).

E-cigarettes and other electronic nicotine dispenser systems (ENDS) are popular particularly among the younger generation and are considered by some to be useful smoking cessation aids (62). E-cigarettes do not produce carbon monoxide and toxins associated with conventional cigarettes. Because e-cigarettes do not involve combustion, they are speculated to aid in smoking cessation without some of the adverse effects of conventional cigarettes. However, the debate about whether e-cigarettes will provide long-term benefits or harm is ongoing, with little data on the pulmonary effects (63). Although the components are toxic, the potentially adverse effects of e-cigarette liquids on the cardiopulmonary system are largely unknown. Given the increasing popularity of flavored e-cigarettes, their components and potential health risks must be studied systematically. E-cigarette and vaping-related acute lung injury (EVALI) have recently emerged as an urgent crisis (64). The U.S. Center for Disease Control (CDC) estimates that over 2,500 patients developed EVALI, with over 60 deaths from this mysterious illness (65). A breakthrough by the CDC eventually revealed the components associated with EVALI. While studying the bronchoalveolar lavage specimens, investigators discovered nicotine, Δ9-THC, and vitamin E acetate as the most common components of e-cigarettes in EVALI patients (66) However, the mechanism by which these components and others affect the pulmonary tissue is unclear.

Neuropsychiatric:

Cannabinoids are used to treat various neurological disorders, including chronic pain, neurodegenerative diseases, and epilepsy. However, cannabinoids can also cause adverse neuropsychiatric effects. The effects are both acute and chronic and the incidence is increasing as a consequence of legalization.

Acute cannabis use causes sedation, analgesia, hypothermia, and hypomobility (67). Cannabis intoxication can cause impaired motor coordination and judgement while producing euphoria and anxiety. Heavy cannabis use may alter neurological pathways and lead to addiction via the psychoactive component of cannabis, Δ9-THC (68). Withdrawal symptoms from cannabis is also reported in a subset of patients (69).

Cannabis misuse has led to an increase in intoxication and cannabis hyperemesis syndrome (70). Chronic cannabis use causes dysregulation of the neuronal pathways in the central and enteric nervous system, resulting in cyclical nausea, vomiting, and abdominal pain after cannabis use (71). The presentation resembles cyclical vomiting and can resolve with cannabis cessation.

Cannabis use disorder is the continued use of cannabis despite causing harm to the user or impaired social functioning (72). Cerda et al. found an increase in cannabis use disorder with legalization (73). There was an increase in respondents aged 12 to 17 who reported increased cannabis use disorder (2.18% to 2.72%). In comparison, the proportion of respondents over 26 years of age who reported an increase in frequent cannabis use and cannabis abuse disorder increased from 2.13% to 2.62% and from 0.9% to 1.23%, respectively.

Cannabinoids are associated with psychiatric conditions, including psychosis and mood disorders. Patients who use cannabis are more likely to have anxiety, depression, and bipolar disorder (74). Acutely, cannabis causes euphoria, but chronic use might perturb neurological pathways and cause mood disorders. However, cannabis use might be confounded with bipolar disorder and mitigate impulsive behavior (75), although cannabis use is associated with a higher rate of suicide in bipolar disorder.

Cannabis is also linked with psychosis. Using logistic regression analysis, Di Forti et al. found a link between high-potency cannabis use and the development of psychotic disorders (76). Exogenous cannabinoids affect the development of the central nervous system in rats, cause cognitive deficits, and alters cortical gene expression (77). A genetic variation of the AKT1 gene that affects dopamine signaling increases the risk of developing psychosis with chronic cannabis use (78).

Trauma and Overdose: In the United States, motor vehicle accidents are a leading cause of mortality. Alcohol intoxication and, more recently, distracted driving with cellular phones and other personal devices are associated with increased traffic accidents and fatalities (79). Cannabis intoxication is also associated with motor vehicle accidents (80). While acute alcohol intoxication can be detected with a breathalyzer or blood test, detecting cannabis intoxication is more difficult and requires novel methods (81, 82). Cannabis can remain detectable in blood, saliva, and urine for hours to days, making it difficult to distinguish between acute intoxication and chronic use.

Misadventure with cannabis is now being reported with cannabis edibles. Unlike smoking or vaping, edibles appear innocuous and are easy to ingest by anyone regardless of experience or age. A recent report suggests that pediatric emergency department visits have increased in Canada since the legalization of cannabis due to unintentional ingestion of cannabis (83). Pediatric patients present with analgesia and sedation, which can be fatal.

Reproduction and Fertility:

Cannabis affects male fertility and conception. A systematic review found that chronic marijuana exposure reduced male fertility with lower sperm count, abnormal morphology, mobility, and viability (84). Harlow et al. report that male preconception use of cannabis is associated with spontaneous abortion (85). Compared to non-users, male cannabis use increased the hazard ratio of spontaneous abortion by 2.0 (95% CI: 1.2-3.1) when using cannabis more than once per week.

Prenatal exposure to cannabinoids can affect fetal development. Pregnant women frequently experience nausea and vomiting. Hyperemesis gravidarum is protracted nausea and vomiting that is only partly responsive to antiemetic medications. Cannabis is considered a benign herbal antiemetic and was recently studied for hyperemesis gravidarum and found to significantly improve symptoms (86). However, the potential effects on the fetus are concerning. The Centers for Disease Control recommends against using marijuana during pregnancy because it is associated with abnormal neurological development and low birth weight (87). In a systematic review, Gunn et al. found that prenatal exposure to cannabis is associated with maternal anemia, low birth weight, low neonatal length, and premature gestational age (88). Furthermore, Roncero et al. found a link between prenatal cannabis exposure and developmental and mental disorders in children, such as attention deficit hyperactivity disorder and depression (89).

Malignancy:

Cannabinoids are approved for treating cancer symptoms, including pain, nausea, and vomiting (90). Preclinical evidence suggests that cannabinoids might be effective therapy for glioblastoma multiforme, colon cancer, skin cancer, prostate cancer, and breast cancer (90). However, the clinical data on the efficacy of cannabinoids in cancer is unclear. There is a paucity of evidence-based clinical therapies showing cannabis improves cancer survival. After tumor debulking, only a single study in glioblastoma multiforme showed that Δ9-THC treatment caused a reduction in angiogenesis and tumor growth when assessed with MRI on post-treatment biopsies (91).

Recent evidence suggests a link between marijuana use and cancer. Ghasemiesfe et al. conducted a systematic review and metanalysis and found an association between marijuana use and cancer (92). While there was no association with head and neck cancer or oral cancer, testicular germ cell tumor was linked to marijuana use (OR, 1.36; 95% CI, 1.03–1.81; P = .03; I2 = 0%). The study was underpowered to link marijuana use with lung cancer. However, Addington et al. found in case-control study that cannabis was associated with lung cancer (93). After adjusting for tobacco smoking, the risk of lung cancer increased with higher cannabis use. With legalization and increased use, the link between cancer and marijuana is expected to become more pronounced in the future and warrants further investigation.

Conclusion

Cannabis has been cultivated for thousands of years for recreation and medical therapy. In the early 20th century, restrictions limited its widespread consumption. Over the past twenty years, decriminalization and legalization have increased the availability of cannabis. Medicinal marijuana may benefit patients with chemotherapy-induced nausea and vomiting, HIV-induced cachexia, and neurodegenerative disorders. However, restrictions continue to limit research into the efficacy of medical marijuana. Preclinical studies and emerging epidemiological data suggest cannabis is expected to affect human health adversely in multiple ways. At the beginning of the twentieth century, tobacco cigarettes were incorporated into the American lifestyle and spread worldwide. We now know this led to dramatic increases in respiratory disease, cancer, and cardiovascular disease that caused millions of deaths. Cannabis might have medicinal benefits, but no medication is without side effects or adverse effects. Growing evidence suggests that growing cannabis use will exacerbate cardiopulmonary disease and malignancy in the future. However, CB1 antagonists and CB2 agonists might provide a novel class of medical therapies (Figure 2).

Figure 2. Modulation of cannabinoid receptor activity.

Figure 2.

A) Cannabinoid receptor-1 (CB1) agonists activate the CB1 receptor causing inflammation, oxidative stress, and atherosclerosis. A CB1 agonist such as THC (red) binds to the CB1 receptor and activates the G-protein coupled receptor (GPCR) causing inhibition of adenylyl cyclase, mitogen activated pathway (MAP) kinase phosphorylation, NF-κβ translocation to the nucleus, and activation of B-arrestin pathways. CB1 activation ultimately causes increased expression of inflammatory cytokines and oxidative stress that promote atherosclerosis. B) A CB1 receptor inverse agonist binds and prevents the agonists from binding the CB1 receptor, modulates receptor function, and attenuates CB1 mediated atherosclerosis. An inverse agonist such as rimonabant (blue) blocks CB1 agonist binding (red) and downregulates CB1 receptor activity via the GPCR thus preventing inhibition of adenylyl cyclase, MAP kinase phosphorylation, NF-κβ translocation to the nucleus, and B-arrestin pathway activation. Consequently, inverse agonists abrogate inflammation, oxidative stress, and atherosclerosis that are mediated by CB1-receptor agonist interactions. C) A neutral antagonist blocks CB1 agonist binding and attenuates CB1 mediated atherosclerosis. A neutral antagonist such as genistein (green) blocks CB1 agonist (red) and does not affect CB1 receptor or downstream pathways. Neutral antagonists also abrogate CB1-mediated inflammation, oxidative stress, and atherosclerosis. D) CB2 agonists attenuate the adverse effects of CB1 activation by modulating the immune system, inflammation and oxidative stress, and thus preventing atherosclerosis. A CB2 agonist such as JWH133 (yellow) binds the CB2 receptor and counteracts the effects of a CB1 agonist (red) binding the CB1 receptor by decreasing MAP kinase phosporylation, preventing NF-κβ translocation, and modulating B-arrestin pathways. Because CB2 receptors are expressed on immune cells and the vasculature, CB2 agonists prevent transformation of monocytes into macrophages in addition to suppressing vascular inflammation and oxidative stress which promotes vascular quiescence and prevents atherosclerosis. Created with BioRender.com.

The CB2 receptor is expressed mainly in immune and hematopoietic cells but also in peripheral cells, tissues, and organs including the central nervous system, enteric nervous system, heart, vasculature, liver, and pancreas (94). CB2 agonists modulate immune function and attenuate vascular inflammation (95). Thus, CB2 agonists might be beneficial for treating inflammatory diseases such as inflammatory bowel disease, atherosclerosis, sepsis, traumatic brain injury, pain, and neurodegenerative diseases (96).

Novel CB1 antagonist therapies might mitigate the adverse effects of cannabis caused by Δ9-THC. CB1 signaling involves various pathophysiological processes, including obesity, smoking cessation, diabetes, coronary artery disease, atherosclerosis, liver cirrhosis, and cancer (34). CB1 antagonists are capable of attenuating Δ9-THC-mediated inflammation and oxidative stress. Experimental and clinical evidence supports the therapeutic potential of CB1 antagonists. In 2006, Acomplia® (rimonabant) became the first approved CB1 antagonist in Europe that is approved for treating obesity (97). However, rimonabant was later withdrawn in 2008 due to severe psychiatric side effects (98). Second and third-generation compounds derived from rimonabant thus far have failed to translate to the clinic because of concerns for psychiatric effects and efficacy (99). To reduce the psychiatric side effects, pharmaceutical companies developed peripherally restricted CB1 antagonists with limited blood-brain barrier (BBB) permeability and, in theory, less central nervous system exposure (100). Despite the restricted bioavailability, psychiatric side effects could not be completely excluded. Therefore, continuing efforts to discover novel CB1 antagonists with therapeutic potential that lack psychiatric side effects would be clinically important with the rapid growth of cannabis use worldwide.

Acknowledgements:

We thank Blake Wu for his assistance with manuscript preparation. Owing to space limitation, we are unable to include all the important papers relevant to iPSC and environmental exposure research, and we apologize to those investigators who have otherwise contributed substantially to this field. This work was supported by the Stanford Cardiovascular Institute, Tobacco-Related Disease Research Program (TRDRP) 27IR-0012, Steven M. Gootter Foundation, P01 HL152953, and AHA 20YVNR3500014 (JCW).

Footnotes

Disclosure Statement & Competing Interests

J.C.W. is a co-founder and SAB of Greenstone Biosciences, and M.C. is a consultant for Greenstone Biosciences, but this manuscript was written independently.

For more information:

For additional information on iPSC Biobanking, cell culture protocols, publications, and contact information to request iPSC lines. Please visit the Stanford Cardiovascular Institute (SCVI) Biobank website: https://med.stanford.edu/scvibiobank.html

References:

  • 1.Bridgeman MB, Abazia DT. 2017. Medicinal Cannabis: History, Pharmacology, And Implications for the Acute Care Setting. P T 42:180–8 [PMC free article] [PubMed] [Google Scholar]
  • 2.Page RL 2nd, Allen LA, Kloner RA, Carriker CR, Martel C, et al. 2020. Medical Marijuana, Recreational Cannabis, and Cardiovascular Health: A Scientific Statement From the American Heart Association. Circulation 142:e131–e52 [DOI] [PubMed] [Google Scholar]
  • 3.Rotermann M. 2020. What has changed since cannabis was legalized? Health Rep 31:11–20 [DOI] [PubMed] [Google Scholar]
  • 4.Martins SS, Segura LE, Levy NS, Mauro PM, Mauro CM, et al. 2021. Racial and Ethnic Differences in Cannabis Use Following Legalization in US States With Medical Cannabis Laws. JAMA Network Open 4:e2127002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Atakan Z. 2012. Cannabis, a complex plant: different compounds and different effects on individuals. Therapeutic Advances in Psychopharmacology 2:241–54 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Hoffman AF, Lupica CR. 2013. Synaptic targets of Delta9-tetrahydrocannabinol in the central nervous system. Cold Spring Harb Perspect Med 3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Ibsen MS, Connor M, Glass M. 2017. Cannabinoid CB1 and CB2 Receptor Signaling and Bias. Cannabis and Cannabinoid Research 2:48–60 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bilbao A, Spanagel R. 2022. Medical cannabinoids: a pharmacology-based systematic review and meta-analysis for all relevant medical indications. BMC Med 20:259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Nagarkatti P, Pandey R, Rieder SA, Hegde VL, Nagarkatti M. 2009. Cannabinoids as novel anti-inflammatory drugs. Future Med Chem 1:1333–49 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Do Y, McKallip RJ, Nagarkatti M, Nagarkatti PS. 2004. Activation through cannabinoid receptors 1 and 2 on dendritic cells triggers NF-kappaB-dependent apoptosis: novel role for endogenous and exogenous cannabinoids in immunoregulation. J Immunol 173:2373–82 [DOI] [PubMed] [Google Scholar]
  • 11.Zhu W, Friedman H, Klein TW. 1998. Delta9-tetrahydrocannabinol induces apoptosis in macrophages and lymphocytes: involvement of Bcl-2 and caspase-1. The Journal of Pharmacology and Experimental Therapeutics 286:1103–9 [PubMed] [Google Scholar]
  • 12.Srivastava MD, Srivastava BI, Brouhard B. 1998. Delta9 tetrahydrocannabinol and cannabidiol alter cytokine production by human immune cells. Immunopharmacology 40:179–85 [DOI] [PubMed] [Google Scholar]
  • 13.Smith SR, Terminelli C, Denhardt G. 2000. Effects of cannabinoid receptor agonist and antagonist ligands on production of inflammatory cytokines and anti-inflammatory interleukin-10 in endotoxemic mice. The Journal of Pharmacology and Experimental Therapeutics 293:136–50 [PubMed] [Google Scholar]
  • 14.Pertwee RG. 2002. Cannabinoids and multiple sclerosis. Pharmacol Ther 95:165–74 [DOI] [PubMed] [Google Scholar]
  • 15.Jia W, Hegde VL, Singh NP, Sisco D, Grant S, et al. 2006. Delta9-tetrahydrocannabinol-induced apoptosis in Jurkat leukemia T cells is regulated by translocation of Bad to mitochondria. Mol Cancer Res 4:549–62 [DOI] [PubMed] [Google Scholar]
  • 16.McKallip RJ, Lombard C, Fisher M, Martin BR, Ryu S, et al. 2002. Targeting CB2 cannabinoid receptors as a novel therapy to treat malignant lymphoblastic disease. Blood 100:627–34 [DOI] [PubMed] [Google Scholar]
  • 17.Soares RZ, Vuolo F, Dall'Igna DM, Michels M, Crippa JA, et al. 2015. Evaluation of the role of the cannabidiol system in an animal model of ischemia/reperfusion kidney injury. Rev Bras Ter Intensiva 27:383–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lowin T, Tingting R, Zurmahr J, Classen T, Schneider M, Pongratz G. 2020. Cannabidiol (CBD): a killer for inflammatory rheumatoid arthritis synovial fibroblasts. Cell Death Dis 11:714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Sánchez AJ, García-Merino A. 2012. Neuroprotective agents: cannabinoids. Clin Immunol 142:57–67 [DOI] [PubMed] [Google Scholar]
  • 20.Vallée A, Lecarpentier Y, Guillevin R, Vallée JN. 2017. Effects of cannabidiol interactions with Wnt/β-catenin pathway and PPARγ on oxidative stress and neuroinflammation in Alzheimer's disease. Acta Biochim Biophys Sin (Shanghai) 49:853–66 [DOI] [PubMed] [Google Scholar]
  • 21.Santos NA, Martins NM, Sisti FM, Fernandes LS, Ferreira RS, et al. 2015. 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 30:231–40 [DOI] [PubMed] [Google Scholar]
  • 22.Chandy M, Obal D, Wu JC. 2022. Elucidating effects of environmental exposure using human-induced pluripotent stem cell disease modeling. EMBO Mol Med 14:e13260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Obal D, Wu JC. 2020. Induced pluripotent stem cells as a platform to understand patient-specific responses to opioids and anaesthetics. British journal of pharmacology 177:4581–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Whiting PF, Wolff RF, Deshpande S, Di Nisio M, Duffy S, et al. 2015. Cannabinoids for Medical Use: A Systematic Review and Meta-analysis. JAMA 313:2456–73 [DOI] [PubMed] [Google Scholar]
  • 25.Hussain SA, Zhou R, Jacobson C, Weng J, Cheng E, et al. 2015. Perceived efficacy of cannabidiol-enriched cannabis extracts for treatment of pediatric epilepsy: A potential role for infantile spasms and Lennox-Gastaut syndrome. Epilepsy Behav 47:138–41 [DOI] [PubMed] [Google Scholar]
  • 26.Volkow ND, Baler RD, Compton WM, Weiss SR. 2014. Adverse health effects of marijuana use. N Engl J Med 370:2219–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.DeFilippis EM, Singh A, Divakaran S, Gupta A, Collins BL, et al. 2018. Cocaine and Marijuana Use Among Young Adults With Myocardial Infarction. Journal of the American College of Cardiology 71:2540–51 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Pacher P, Steffens S, Hasko G, Schindler TH, Kunos G. 2018. Cardiovascular effects of marijuana and synthetic cannabinoids: the good, the bad, and the ugly. Nature reviews. Cardiology 15:151–66 [DOI] [PubMed] [Google Scholar]
  • 29.Libby P. 2002. Inflammation in atherosclerosis. Nature 420:868–74 [DOI] [PubMed] [Google Scholar]
  • 30.Everett BM, MacFadyen JG, Thuren T, Libby P, Glynn RJ, Ridker PM. 2020. Inhibition of Interleukin-1beta and Reduction in Atherothrombotic Cardiovascular Events in the CANTOS Trial. Journal of the American College of Cardiology 76:1660–70 [DOI] [PubMed] [Google Scholar]
  • 31.Shrestha S, Irvin MR, Grunfeld C, Arnett DK. 2014. HIV, inflammation, and calcium in atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology 34:244–50 [DOI] [PubMed] [Google Scholar]
  • 32.Skeoch S, Bruce IN. 2015. Atherosclerosis in rheumatoid arthritis: is it all about inflammation? Nat Rev Rheumatol 11:390–400 [DOI] [PubMed] [Google Scholar]
  • 33.Frostegard J. 2005. SLE, atherosclerosis and cardiovascular disease. J Intern Med 257:485–95 [DOI] [PubMed] [Google Scholar]
  • 34.Sugamura K, Sugiyama S, Nozaki T, Matsuzawa Y, Izumiya Y, et al. 2009. Activated endocannabinoid system in coronary artery disease and antiinflammatory effects of cannabinoid 1 receptor blockade on macrophages. Circulation 119:28–36 [DOI] [PubMed] [Google Scholar]
  • 35.Liu J, Gao B, Mirshahi F, Sanyal AJ, Khanolkar AD, et al. 2000. Functional CB1 cannabinoid receptors in human vascular endothelial cells. Biochem J 346 Pt 3:835–40 [PMC free article] [PubMed] [Google Scholar]
  • 36.Rajesh M, Mukhopadhyay P, Hasko G, Liaudet L, Mackie K, Pacher P. 2010. Cannabinoid-1 receptor activation induces reactive oxygen species-dependent and -independent mitogen-activated protein kinase activation and cell death in human coronary artery endothelial cells. British Journal of Pharmacology 160:688–700 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Rajesh M, Batkai S, Kechrid M, Mukhopadhyay P, Lee WS, et al. 2012. Cannabinoid 1 receptor promotes cardiac dysfunction, oxidative stress, inflammation, and fibrosis in diabetic cardiomyopathy. Diabetes 61:716–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.El-Remessy AB, Rajesh M, Mukhopadhyay P, Horvath B, Patel V, et al. 2011. Cannabinoid 1 receptor activation contributes to vascular inflammation and cell death in a mouse model of diabetic retinopathy and a human retinal cell line. Diabetologia 54:1567–78 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Dol-Gleizes F, Paumelle R, Visentin V, Mares AM, Desitter P, et al. 2009. Rimonabant, a selective cannabinoid CB1 receptor antagonist, inhibits atherosclerosis in LDL receptor-deficient mice. Arteriosclerosis, Thrombosis, and Vascular Biology 29:12–8 [DOI] [PubMed] [Google Scholar]
  • 40.Netherland CD, Pickle TG, Bales A, Thewke DP. 2010. Cannabinoid receptor type 2 (CB2) deficiency alters atherosclerotic lesion formation in hyperlipidemic Ldlr-null mice. Atherosclerosis 213:102–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hoyer FF, Steinmetz M, Zimmer S, Becker A, Lutjohann D, et al. 2011. Atheroprotection via cannabinoid receptor-2 is mediated by circulating and vascular cells in vivo. J Mol Cell Cardiol 51:1007–14 [DOI] [PubMed] [Google Scholar]
  • 42.Draz EI, Oreby MM, Elsheikh EA, Khedr LA, Atlam SA. 2017. Marijuana use in acute coronary syndromes. Am J Drug Alcohol Abuse 43:576–82 [DOI] [PubMed] [Google Scholar]
  • 43.Patel RS, Katta SR, Patel R, Ravat V, Gudipalli R, et al. 2018. Cannabis Use Disorder in Young Adults with Acute Myocardial Infarction: Trend Inpatient Study from 2010 to 2014 in the United States. Cureus 10:e3241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Frost L, Mostofsky E, Rosenbloom JI, Mukamal KJ, Mittleman MA. 2013. Marijuana use and long-term mortality among survivors of acute myocardial infarction. Am Heart J 165:170–5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.McEvoy JW, Blaha MJ, DeFilippis AP, Lima JA, Bluemke DA, et al. 2015. Cigarette smoking and cardiovascular events: role of inflammation and subclinical atherosclerosis from the MultiEthnic Study of Atherosclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology 35:700–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Wei TT, Chandy M, Nishiga M, Zhang A, Kumar KK, et al. 2022. Cannabinoid receptor 1 antagonist genistein attenuates marijuana-induced vascular inflammation. Cell 185:1676–93 e23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Bycroft C, Freeman C, Petkova D, Band G, Elliott LT, et al. 2018. The UK Biobank resource with deep phenotyping and genomic data. Nature 562:203–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Paik DT, Chandy M, Wu JC. 2020. Patient and Disease-Specific Induced Pluripotent Stem Cells for Discovery of Personalized Cardiovascular Drugs and Therapeutics. Pharmacol Rev 72:320–42 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Steffens S, Veillard NR, Arnaud C, Pelli G, Burger F, et al. 2005. Low dose oral cannabinoid therapy reduces progression of atherosclerosis in mice. Nature 434:782–6 [DOI] [PubMed] [Google Scholar]
  • 50.Kariyanna PT, Jayarangaiah A, Singh N, Song T, Soroka S, et al. 2018. Marijuana Induced Myocarditis: A New Entity of Toxic Myocarditis. Am J Med Case Rep 6:169–72 [Google Scholar]
  • 51.Wei TT, Chandy M, Nishiga M, Zhang A, Kumar KK, et al. 2022. Cannabinoid receptor 1 antagonist genistein attenuates marijuana-induced vascular inflammation. Cell 185:1676–93.e23 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Sultan SR, Millar SA, O'Sullivan SE, England TJ. 2018. A Systematic Review and Meta-Analysis of the In Vivo Haemodynamic Effects of Delta(8)-Tetrahydrocannabinol. Pharmaceuticals (Basel) 11 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Richards JR, Blohm E, Toles KA, Jarman AF, Ely DF, Elder JW. 2020. The association of cannabis use and cardiac dysrhythmias: a systematic review. Clin Toxicol (Phila) 58:861–9 [DOI] [PubMed] [Google Scholar]
  • 54.Patel RS, Gonzalez MD, Ajibawo T, Baweja R. 2021. Cannabis use disorder and increased risk of arrhythmia-related hospitalization in young adults. Am J Addict 30:578–84 [DOI] [PubMed] [Google Scholar]
  • 55.Chouairi F, Miller PE, Guha A, Clarke JR, Reinhardt SW, et al. 2021. Cannabis use disorder among atrial fibrillation admissions, 2008-2018. Pacing Clin Electrophysiol 44:1934–8 [DOI] [PubMed] [Google Scholar]
  • 56.Ezzati M, Lopez AD. 2003. Estimates of global mortality attributable to smoking in 2000. Lancet 362:847–52 [DOI] [PubMed] [Google Scholar]
  • 57.National Center for Chronic Disease Prevention and Health Promotion (US) Office on Smoking and Health. In The Health Consequences of Smoking-50 Years of Progress: A Report of the Surgeon General. Atlanta (GA). Centers for Disease Control and Prevention (US); 2014. 8, Cardiovascular Diseases. [PubMed] [Google Scholar]
  • 58.Wu TC, Tashkin DP, Djahed B, Rose JE. 1988. Pulmonary hazards of smoking marijuana as compared with tobacco. N Engl J Med 318:347–51 [DOI] [PubMed] [Google Scholar]
  • 59.Jayakumar N, Chaiton M, Goodwin R, Schwartz R, O'Connor S, Kaufman P. 2021. Co-use and Mixing Tobacco With Cannabis Among Ontario Adults. Nicotine Tob Res 23:171–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Hancox RJ, Poulton R, Ely M, Welch D, Taylor DR, et al. 2010. Effects of cannabis on lung function: a population-based cohort study. Eur Respir J 35:42–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Tan WC, Bourbeau J, Aaron SD, Hogg JC, Maltais F, et al. 2019. The effects of marijuana smoking on lung function in older people. Eur Respir J 54 [DOI] [PubMed] [Google Scholar]
  • 62.Benowitz NL, Fraiman JB. 2017. Cardiovascular effects of electronic cigarettes. Nat. Rev. Cardiol 14:447–56 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Gotts JE, Jordt S-E, McConnell R, Tarran R. 2019. What are the respiratory effects of e-cigarettes? BMJ 366:l5275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Butt YM, Smith ML, Tazelaar HD, Vaszar LT, Swanson KL, et al. 2019. Pathology of Vaping-Associated Lung Injury. N Engl J Med [DOI] [PubMed] [Google Scholar]
  • 65.Hayes D Jr., Board A, Calfee CS, Ellington S, Pollack LA, et al. 2022. Pulmonary and Critical Care Considerations for e-Cigarette, or Vaping, Product Use-Associated Lung Injury. Chest 162:256–64 [DOI] [PubMed] [Google Scholar]
  • 66.Blount BC, Karwowski MP, Shields PG, Morel-Espinosa M, Valentin-Blasini L, et al. 2019. Vitamin E Acetate in Bronchoalveolar-Lavage Fluid Associated with EVALI. N Engl J Med [Google Scholar]
  • 67.Martin BR, Dewey WL, Harris LS, Beckner J. 1975. Marihuana-like activity of new synthetic tetrahydrocannabinols. Pharmacol Biochem Behav 3:849–53 [DOI] [PubMed] [Google Scholar]
  • 68.Adams IB, Martin BR. 1996. Cannabis: pharmacology and toxicology in animals and humans. Addiction 91:1585–614 [PubMed] [Google Scholar]
  • 69.Bonnet U, Preuss UW. 2017. The cannabis withdrawal syndrome: current insights. Subst Abuse Rehabil 8:9–37 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Gajendran M, Sifuentes J, Bashashati M, McCallum R. 2020. Cannabinoid hyperemesis syndrome: definition, pathophysiology, clinical spectrum, insights into acute and long-term management. J Investig Med 68:1309–16 [DOI] [PubMed] [Google Scholar]
  • 71.Chu F, Cascella M. 2023. Cannabinoid Hyperemesis Syndrome. In StatPearls. Treasure Island (FL). Number of. [PubMed] [Google Scholar]
  • 72.Patel J, Marwaha R. 2023. Cannabis Use Disorder. In StatPearls. Treasure Island (FL). Number of. [Google Scholar]
  • 73.Cerda M, Mauro C, Hamilton A, Levy NS, Santaella-Tenorio J, et al. 2020. Association Between Recreational Marijuana Legalization in the United States and Changes in Marijuana Use and Cannabis Use Disorder From 2008 to 2016. JAMA Psychiatry 77:165–71 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Lucatch AM, Coles AS, Hill KP, George TP. 2018. Cannabis and Mood Disorders. Curr Addict Rep 5:336–45 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Pinto JV, Medeiros LS, Santana da Rosa G, Santana de Oliveira CE, Crippa JAS, et al. 2019. The prevalence and clinical correlates of cannabis use and cannabis use disorder among patients with bipolar disorder: A systematic review with meta-analysis and meta-regression. Neurosci Biobehav Rev 101:78–84 [DOI] [PubMed] [Google Scholar]
  • 76.Di Forti M, Quattrone D, Freeman TP, Tripoli G, Gayer-Anderson C, et al. 2019. The contribution of cannabis use to variation in the incidence of psychotic disorder across Europe (EU-GEI): a multicentre case-control study. The Lancet. Psychiatry 6:427–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Campolongo P, Trezza V, Cassano T, Gaetani S, Morgese MG, et al. 2007. Perinatal exposure to delta-9-tetrahydrocannabinol causes enduring cognitive deficits associated with alteration of cortical gene expression and neurotransmission in rats. Addict Biol 12:485–95 [DOI] [PubMed] [Google Scholar]
  • 78.Di Forti M, Iyegbe C, Sallis H, Kolliakou A, Falcone MA, et al. 2012. Confirmation that the AKT1 (rs2494732) genotype influences the risk of psychosis in cannabis users. Biological Psychiatry 72:811–6 [DOI] [PubMed] [Google Scholar]
  • 79.Kochling J, Geis B, Chao CM, Dieks JK, Wirth S, Hensel KO. 2021. The hazardous (mis)perception of Self-estimated Alcohol intoxication and Fitness to drivE-an avoidable health risk: the SAFE randomised trial. Harm Reduction Journal 18:122. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Rogeberg O, Elvik R. 2016. The effects of cannabis intoxication on motor vehicle collision revisited and revised. Addiction 111:1348–59 [DOI] [PubMed] [Google Scholar]
  • 81.Doucette ML, Frattaroli S, Vernick JS. 2018. Oral fluid testing for marijuana intoxication: enhancing objectivity for roadside DUI testing. Inj Prev 24:78–80 [DOI] [PubMed] [Google Scholar]
  • 82.Lynch KL, Luo YR, Hooshfar S, Yun C. 2019. Correlation of Breath and Blood Delta(9)-Tetrahydrocannabinol Concentrations and Release Kinetics Following Controlled Administration of Smoked Cannabis. Clin Chem 65:1171–9 [DOI] [PubMed] [Google Scholar]
  • 83.Yeung MEM, Weaver CG, Hartmann R, Haines-Saah R, Lang E. 2021. Emergency Department Pediatric Visits in Alberta for Cannabis After Legalization. Pediatrics 148 [DOI] [PubMed] [Google Scholar]
  • 84.Payne KS, Mazur DJ, Hotaling JM, Pastuszak AW. 2019. Cannabis and Male Fertility: A Systematic Review. J Urol 202:674–81 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Harlow AF, Wesselink AK, Hatch EE, Rothman KJ, Wise LA. 2021. Male Preconception Marijuana Use and Spontaneous Abortion: A Prospective Cohort Study. Epidemiology 32:239–47 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Koren G, Cohen R. 2020. The use of cannabis for Hyperemesis Gravidarum (HG). J Cannabis Res 2:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.2017. In The Health Effects of Cannabis and Cannabinoids: The Current State of Evidence and Recommendations for Research. Washington (DC). Number of. [PubMed] [Google Scholar]
  • 88.Gunn JK, Rosales CB, Center KE, Nunez A, Gibson SJ, et al. 2016. Prenatal exposure to cannabis and maternal and child health outcomes: a systematic review and meta-analysis. BMJ Open 6:e009986. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Roncero C, Valriberas-Herrero I, Mezzatesta-Gava M, Villegas JL, Aguilar L, Grau-Lopez L. 2020. Cannabis use during pregnancy and its relationship with fetal developmental outcomes and psychiatric disorders. A systematic review. Reprod Health 17:25. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Bowles DW, O’Bryant CL, Camidge DR, Jimeno A. 2012. The intersection between cannabis and cancer in the United States. Critical Reviews in Oncology/Hematology 83:1–10 [DOI] [PubMed] [Google Scholar]
  • 91.Guzman M, Duarte MJ, Blazquez C, Ravina J, Rosa MC, et al. 2006. A pilot clinical study of Delta9-tetrahydrocannabinol in patients with recurrent glioblastoma multiforme. Br J Cancer 95:197–203 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Ghasemiesfe M, Barrow B, Leonard S, Keyhani S, Korenstein D. 2019. Association Between Marijuana Use and Risk of Cancer: A Systematic Review and Meta-analysis. JAMA Network Open 2:e1916318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Aldington S, Harwood M, Cox B, Weatherall M, Beckert L, et al. 2008. Cannabis use and risk of lung cancer: a case-control study. Eur Respir J 31:280–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Pacher P, Mechoulam R. 2011. Is lipid signaling through cannabinoid 2 receptors part of a protective system? Prog Lipid Res 50:193–211 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Turcotte C, Blanchet MR, Laviolette M, Flamand N. 2016. The CB(2) receptor and its role as a regulator of inflammation. Cell Mol Life Sci 73:4449–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Bie B, Wu J, Foss JF, Naguib M. 2018. An overview of the cannabinoid type 2 receptor system and its therapeutic potential. Curr Opin Anaesthesiol 31:407–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Despres JP, Golay A, Sjostrom L. 2005. Effects of rimonabant on metabolic risk factors in overweight patients with dyslipidemia. N Engl J Med 353:2121–34 [DOI] [PubMed] [Google Scholar]
  • 98.Onakpoya IJ, Heneghan CJ, Aronson JK. 2016. Worldwide withdrawal of medicinal products because of adverse drug reactions: a systematic review and analysis. Critical Reviews in Toxicology 46:477–89 [DOI] [PubMed] [Google Scholar]
  • 99.Cinar R, Iyer MR, Kunos G. 2020. The therapeutic potential of second and third generation CB(1)R antagonists. Pharmacol Ther 208:107477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Fulp A, Bortoff K, Seltzman H, Zhang Y, Mathews J, et al. 2012. Design and synthesis of cannabinoid receptor 1 antagonists for peripheral selectivity. Journal of Medicinal Chemistry 55:2820–34 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES