1
|
Strong SJ, Thomas HA, Adams ZW, Hulvershorn LA. Comorbid Cannabis Use and Mood Disorders Among Adolescents. FOCUS (AMERICAN PSYCHIATRIC PUBLISHING) 2025; 23:133-140. [PMID: 40235605 PMCID: PMC11995898 DOI: 10.1176/appi.focus.20240049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Adolescence is a unique developmental period marked by increased exploration and risk-taking, as well as important brain development milestones. Many people who use substances will begin using during adolescence, and cannabis is the most commonly used illicit substance among adolescents. For adolescents with mood disorders, cannabis use (and, by extension, cannabis use disorder) is even more likely, and the associated consequences are even more significant. In this review, we explore the assessment of cannabis use disorder among adolescents, the impact of cannabis use on mood symptoms, level-of-care recommendations for adolescents with comorbid cannabis use and mood disorders, and effective treatment options.
Collapse
Affiliation(s)
- Stephane J Strong
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis (all authors); Department of Psychology, Wayne State University, Detroit, Michigan (Thomas)
| | - Halle A Thomas
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis (all authors); Department of Psychology, Wayne State University, Detroit, Michigan (Thomas)
| | - Zachary W Adams
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis (all authors); Department of Psychology, Wayne State University, Detroit, Michigan (Thomas)
| | - Leslie A Hulvershorn
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis (all authors); Department of Psychology, Wayne State University, Detroit, Michigan (Thomas)
| |
Collapse
|
2
|
Gray KM, Tomko RL, Baker NL, McClure EA, McRae-Clark AL, Squeglia LM. N-acetylcysteine for youth cannabis use disorder: randomized controlled trial main findings. Neuropsychopharmacology 2025; 50:731-738. [PMID: 39910268 PMCID: PMC11914066 DOI: 10.1038/s41386-025-02061-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/14/2025] [Accepted: 01/27/2025] [Indexed: 02/07/2025]
Abstract
Cannabis use disorder is particularly prevalent and impairing among young people, and evidence-based treatments are limited. Prior trials of N-acetylcysteine, added to contingency management as a platform behavioral intervention, yielded positive findings in youth but not in adults. This trial sought to rigorously evaluate whether N-acetylcysteine is efficacious in youth when not paired with a robust behavioral treatment platform. Treatment-seeking youth with cannabis use disorder (N = 192, ages 14-21) were randomized to receive a double-blind 12-week course of oral N-acetylcysteine 1200 mg or placebo twice daily; all received weekly medical management and brief behavioral counseling. The primary efficacy outcome was the proportion of negative urine cannabinoid tests during treatment, compared between groups. An array of self-report and urine testing measures were examined secondarily to assess cannabis use reduction and cessation outcomes. The N-acetylcysteine and placebo groups did not differ in proportion of negative urine cannabinoid tests (RR = 0.93, 95% CI = 0.53, 1.64; p = 0.80) or self-reported cannabis abstinence (RR = 1.02, 95% CI = 0.63, 1.65; p = 0.93) during treatment. The mean percentage of cannabis use days and grams of cannabis used per using day decreased over time during treatment but did not differ between groups. More N-acetylcysteine than placebo treated participants reported gastrointestinal adverse events (63/98 versus 37/94, χ21 = 11.9 p < 0.001); adverse events were otherwise similar between groups. Findings indicate that N-acetylcysteine is not efficacious for youth cannabis use disorder when not paired with contingency management, highlighting the potentially crucial role of a robust behavioral treatment platform in facilitating prior positive efficacy findings with N-acetylcysteine.Trial Registration: Clinicaltrials.gov identifier NCT03055377.
Collapse
Affiliation(s)
- Kevin M Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA.
| | - Rachel L Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Nathaniel L Baker
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Erin A McClure
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Aimee L McRae-Clark
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Lindsay M Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
3
|
Castro EM, Lotfipour S, Leslie FM. Neuroglia in substance use disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:347-369. [PMID: 40148055 DOI: 10.1016/b978-0-443-19102-2.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2025]
Abstract
Substance use disorders (SUD) remain a major public health concern in which individuals are unable to control their use of substances despite significant harm and negative consequences. Drugs of abuse dysregulate major brain and behavioral functions. Glial cells, primarily microglia and astrocytes, play a crucial role in these drug-induced molecular and behavioral changes. This review explores preclinical and clinical studies of how neuroglia and their associated neuroinflammatory responses contribute to SUD and reward-related properties. We evaluate preclinical and clinical evidence for targeting neuroglia as therapeutic interventions. In addition, we evaluate the literature on the gut microbiome and its role in SUD. Clinical treatments are most effective for reducing drug cravings, and some have yielded promising results in other measures of drug use. N-Acetylcysteine, through modulation of cysteine-glutamate antiporter of glial cells, shows encouraging results across a variety of drug classes. Neuroglia and gut microbiome interactions are important factors to consider with regard to SUD and could lead to novel therapeutic avenues. Age- and sex-dependent properties of neuroglia, gut microbiome, and drug use behaviors are important areas in need of further investigation.
Collapse
Affiliation(s)
- Emily M Castro
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States; Department of Emergency Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States; Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, Irvine, CA, United States
| | - Frances M Leslie
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California, Irvine, Irvine, CA, United States.
| |
Collapse
|
4
|
De Felice M, Szkudlarek HJ, Uzuneser TC, Rodríguez-Ruiz M, Sarikahya MH, Pusparajah M, Galindo Lazo JP, Whitehead SN, Yeung KKC, Rushlow WJ, Laviolette SR. The Impacts of Adolescent Cannabinoid Exposure on Striatal Anxiety- and Depressive-Like Pathophysiology Are Prevented by the Antioxidant N-Acetylcysteine. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2024; 4:100361. [PMID: 39257692 PMCID: PMC11381987 DOI: 10.1016/j.bpsgos.2024.100361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 09/12/2024] Open
Abstract
Background Exposure to Δ9-tetrahydrocannabinol (THC) is an established risk factor for later-life neuropsychiatric vulnerability, including mood- and anxiety-related symptoms. The psychotropic effects of THC on affect and anxiogenic behavioral phenomena are known to target the striatal network, particularly the nucleus accumbens, a neural region linked to mood and anxiety disorder pathophysiology. THC may increase neuroinflammatory responses via the redox system and dysregulate inhibitory and excitatory neural balance in various brain circuits, including the striatum. Thus, interventions that can induce antioxidant effects may counteract the neurodevelopmental impacts of THC exposure. Methods In the current study, we used an established preclinical adolescent rat model to examine the impacts of adolescent THC exposure on various behavioral, molecular, and neuronal biomarkers associated with increased mood and anxiety disorder vulnerability. Moreover, we investigated the protective properties of the antioxidant N-acetylcysteine against THC-related pathology. Results We demonstrated that adolescent THC exposure induced long-lasting anxiety- and depressive-like phenotypes concomitant with differential neuronal and molecular abnormalities in the two subregions of the nucleus accumbens, the shell and the core. In addition, we report for the first time that N-acetylcysteine can prevent THC-induced accumbal pathophysiology and associated behavioral abnormalities. Conclusions The preventive effects of this antioxidant intervention highlight the critical role of redox mechanisms underlying cannabinoid-induced neurodevelopmental pathology and identify a potential intervention strategy for the prevention and/or reversal of these pathophysiological sequelae.
Collapse
Affiliation(s)
- Marta De Felice
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Hanna J Szkudlarek
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Taygun C Uzuneser
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mar Rodríguez-Ruiz
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Mohammed H Sarikahya
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | | | | | - Shawn N Whitehead
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
| | - Ken K-C Yeung
- Department of Chemistry, Western University, London, Ontario, Canada
- Department of Biochemistry, Western University, London, Ontario, Canada
| | - Walter J Rushlow
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Steven R Laviolette
- Addiction Research Group, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, Ontario, Canada
- Department of Psychiatry, Schulich School of Medicine & Dentistry, University of Western Ontario, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
- Division of Maternal, Fetal and Newborn Health, Children's Health Research Institute (CHRI), London, Ontario, Canada
| |
Collapse
|
5
|
Alayoubi M, Henry BA, Cahill CM, Cooper ZD. Exploring Novel Pharmacotherapy Candidates for Cannabis Use Disorder: Uncovering Promising Agents on the Horizon by Mechanism of Action. Drugs 2024; 84:1395-1417. [PMID: 39388076 PMCID: PMC11602823 DOI: 10.1007/s40265-024-02098-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 10/15/2024]
Abstract
With rapid expansion of cannabis legalization worldwide, rates of cannabis use and cannabis use disorder (CUD) are increasing; the need for safe and effective medications to treat CUD is urgent. This narrative review evaluates evidence for promising pharmacotherapies to treat CUD from randomized, placebo-controlled trials. Pharmacotherapies for CUD are categorized based on compound targets (e.g., cannabinoid receptor 1 [CB1] agonists such as nabilone, serotonergic compounds such as bupropion, GABAergic compounds such as zolpidem) and outcomes are organized by predetermined withdrawal symptoms, cannabis craving, and cannabis relapse/use. Most promising pharmacotherapies for CUD are drugs that act on the endocannabinoid system and specifically at the CB1 receptor. Priority populations such as females, certain racial/ethnic groups, and age groups experience a different course of CUD progression, symptoms, and drug effects that are important to consider when evaluating outcomes related to CUD. Possible explanations for these disparities are explored, along with the clinical trials that explore these demographics in treating CUD with pharmacotherapies.
Collapse
Affiliation(s)
- Myra Alayoubi
- UCLA, Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, USA
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, USA
- Shirley and Stefan Hatos Center for Neuropharmacology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, USA
| | - Brittany A Henry
- UCLA, Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, USA
- Department of Psychiatry and Biobehavioral Science, UCLA Center for Cannabis and Cannabinoids, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, 760 Westwood Plaza, 37-418, Los Angeles, CA, 90095, USA
| | - Catherine M Cahill
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, USA
- Shirley and Stefan Hatos Center for Neuropharmacology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, USA
| | - Ziva D Cooper
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California, Los Angeles, USA.
- Department of Psychiatry and Biobehavioral Science, UCLA Center for Cannabis and Cannabinoids, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, 760 Westwood Plaza, 37-418, Los Angeles, CA, 90095, USA.
- Department of Anesthesiology and Perioperative Medicine, David Geffen School of Medicine, University of California, Los Angeles, USA.
| |
Collapse
|
6
|
Imperio CG, Levin FR, Martinez D. The Neurocircuitry of Substance Use Disorder, Treatment, and Change: A Resource for Clinical Psychiatrists. Am J Psychiatry 2024; 181:958-972. [PMID: 39380375 PMCID: PMC11926739 DOI: 10.1176/appi.ajp.20231023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
Substance use disorder (SUD) is common in psychiatric patients and has a negative impact on health and well-being. However, SUD often goes untreated, and there is a need for psychiatrists, of all specialties, to address this pervasive clinical problem. In this review, the authors' goal is to provide a resource that describes treatments for SUD, using neuroscience as a framework. They discuss the effect of pharmacotherapy on craving, intoxication, and withdrawal and its ability to interrupt the cycle of substance use in SUD. The neuroscience of stress is reviewed, including medications targeting neurotransmitter systems activated by alarm and fear. Neuroplasticity and promising treatments that use this mechanism, including ketamine, psilocybin, and transcranial magnetic stimulation (TMS), are discussed. The authors conclude by listing resources and practice guidelines for physicians interested in learning more about treatments for SUD.
Collapse
Affiliation(s)
- Caesar G Imperio
- Division on Substance Use Disorders, New York State Psychiatric Institute, New York; Department of Psychiatry, Columbia University Irving Medical Center, New York
| | - Frances R Levin
- Division on Substance Use Disorders, New York State Psychiatric Institute, New York; Department of Psychiatry, Columbia University Irving Medical Center, New York
| | - Diana Martinez
- Division on Substance Use Disorders, New York State Psychiatric Institute, New York; Department of Psychiatry, Columbia University Irving Medical Center, New York
| |
Collapse
|
7
|
McClure EA, Neelon B, Tomko RL, Gray KM, McRae-Clark AL, Baker NL. Association of Cannabis Use Reduction With Improved Functional Outcomes: An Exploratory Aggregated Analysis From Seven Cannabis Use Disorder Treatment Trials to Extract Data-Driven Cannabis Reduction Metrics. Am J Psychiatry 2024; 181:988-996. [PMID: 39380374 DOI: 10.1176/appi.ajp.20230508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
OBJECTIVE This exploratory analysis sought to determine whether decreases in cannabis use are associated with improvements in cannabis-related problems and functional outcomes, and if so, what percentage decrease is associated with improvement. METHODS Data were aggregated from seven cannabis use disorder treatment trials conducted in the United States (N=920; ages 13 years and older; mean age, 25 years; 30% female, 7% Black, 11% Hispanic/Latinx). Outcome measures included the patient-reported Marijuana Problems Scale (MPS), Health-Related Quality of Life scale (HRQOL), and Pittsburgh Sleep Quality Index and the clinician-rated Clinical Global Impressions (CGI) severity and improvement scales (CGI-S and CGI-I). Generalized estimating equations tested the association between changes in 4-week cannabis use and improvements in functional outcomes. Classification and regression tree (CART) models were developed to determine what reductions in cannabis use could be used as classifiers of improvement. RESULTS Decreases in the amount and frequency of cannabis use were significantly associated with improvements in MPS severity and total scores as well as improvements on the CGI-I and in sleep quality, but not improvements on the HRQOL. CART models performed best for CGI-I scores (72%-75% correct classification), while other outcome measures did not perform as well (40%-62% correct classification). CART models showed improvements on the CGI at 74% reduction in use amounts and 47% reduction in use days. CONCLUSIONS Reductions in cannabis use (∼50% reduction in use days and ∼75% reduction in use amounts) were associated with clinician-assessed improvement, which suggests that cannabis use reduction may yield benefit among individuals with cannabis use disorder. These exploratory results extract a data-driven metric to inform future studies, clinicians, patients, and policy recommendations.
Collapse
Affiliation(s)
- Erin A McClure
- Department of Psychiatry and Behavioral Sciences (McClure, Tomko, Gray, McRae-Clark), Hollings Cancer Center (McClure), and Department of Public Health Sciences (Neelon, Baker), Medical University of South Carolina, Charleston; Ralph H. Johnson VA Medical Center, Charleston (McRae-Clark)
| | - Brian Neelon
- Department of Psychiatry and Behavioral Sciences (McClure, Tomko, Gray, McRae-Clark), Hollings Cancer Center (McClure), and Department of Public Health Sciences (Neelon, Baker), Medical University of South Carolina, Charleston; Ralph H. Johnson VA Medical Center, Charleston (McRae-Clark)
| | - Rachel L Tomko
- Department of Psychiatry and Behavioral Sciences (McClure, Tomko, Gray, McRae-Clark), Hollings Cancer Center (McClure), and Department of Public Health Sciences (Neelon, Baker), Medical University of South Carolina, Charleston; Ralph H. Johnson VA Medical Center, Charleston (McRae-Clark)
| | - Kevin M Gray
- Department of Psychiatry and Behavioral Sciences (McClure, Tomko, Gray, McRae-Clark), Hollings Cancer Center (McClure), and Department of Public Health Sciences (Neelon, Baker), Medical University of South Carolina, Charleston; Ralph H. Johnson VA Medical Center, Charleston (McRae-Clark)
| | - Aimee L McRae-Clark
- Department of Psychiatry and Behavioral Sciences (McClure, Tomko, Gray, McRae-Clark), Hollings Cancer Center (McClure), and Department of Public Health Sciences (Neelon, Baker), Medical University of South Carolina, Charleston; Ralph H. Johnson VA Medical Center, Charleston (McRae-Clark)
| | - Nathaniel L Baker
- Department of Psychiatry and Behavioral Sciences (McClure, Tomko, Gray, McRae-Clark), Hollings Cancer Center (McClure), and Department of Public Health Sciences (Neelon, Baker), Medical University of South Carolina, Charleston; Ralph H. Johnson VA Medical Center, Charleston (McRae-Clark)
| |
Collapse
|
8
|
Arjmand S, Ilaghi M, Shafie'ei M, Gobira PH, Grassi-Oliveira R, Wegener G. Exploring the potential link between ΔFosB and N-acetylcysteine in craving/relapse dynamics: can N-acetylcysteine stand out as a possible treatment candidate? Acta Neuropsychiatr 2024; 37:e31. [PMID: 39415655 DOI: 10.1017/neu.2024.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
From a neuroscientific point of view, one of the unique archetypes of substance use disorders is its road to relapse, in which the reward system plays a crucial role. Studies on the neurobiology of substance use disorders have highlighted the central role of a protein belonging to the Fos family of transcription factors, ΔFosB. Relying on the roles ΔFosB plays in the pathophysiology of substance use disorders, we endeavour to present some evidence demonstrating that N-acetylcysteine, a low-cost and well-tolerated over-the-counter medicine, may influence the downstream pathway of ΔFosB, thereby serving as a treatment strategy to mitigate the risk of relapse in cases of substance use.
Collapse
Affiliation(s)
- Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Mehran Ilaghi
- Institute of Neuropharmacology, Kerman Neuroscience Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Shafie'ei
- Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Pedro H Gobira
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Rodrigo Grassi-Oliveira
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Affective Disorders, Aarhus University Hospital-Psychiatry, Aarhus, Denmark
| |
Collapse
|
9
|
Cuocina M, Aiello G, Cutrufelli P, Rampello M, Rapisarda L, Rodolico A, Cantarella G, Signorelli MS, Bernardini R. Effect of N-acetylcysteine on craving in substance use disorders (SUD): a meta-analysis of randomized controlled trials. Front Pharmacol 2024; 15:1462612. [PMID: 39309000 PMCID: PMC11412889 DOI: 10.3389/fphar.2024.1462612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 08/28/2024] [Indexed: 09/25/2024] Open
Abstract
Background N-acetyl cysteine (NAC) appears promising as a treatment in patients with substance use disorder (SUD) as it helps rebalance glutamate levels in the central nervous system (CNS). Basal concentrations of glutamate are indeed reduced in SUD patients but increased during craving. Materials and Methods We conducted a systematic review and meta-analysis of randomized controlled trials (RCTs). We assessed whether NAC reduce craving rating as compared to a placebo in SUD patients. Secondary outcomes were withdrawal symptoms (WS), side effects (SE) and drop-outs. Estimates are presented as standardized mean differences (SMD) or risk ratio (RR) with 95% confidence interval (CI). Results Eleven RCTs were included. NAC reduced craving rating (SMD -0.61 (-1.17, -0.06), p = 0.03, I2 = 85%), with no differences in the subgroup analysis according to the drug addiction (alcohol, cocaine, poly-drugs, amphetamine, nicotine) (p = 0.98). Among the secondary outcomes, for WS data showed no significant difference between groups (SMD -0.18 (-0.43, 0.08), p = 0.17); for SE no substantial difference was observed between the two treatment groups (RR = 1.06 (0.89-1.27), p = 0.52, I2 = 0%); for dropouts the results are in favor of the placebo but no statistically significant (RR 1.17 (0.85, 1.61), p = 0.34; I2 = 0%). Conclusion NAC seem to reduce craving rating in SUD patients, but evidence is weak. More studies are needed to confirm this finding.
Collapse
Affiliation(s)
- Micol Cuocina
- Department Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, Catania, Italy
| | - Giuseppe Aiello
- Department Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, Catania, Italy
| | - Pierfelice Cutrufelli
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Martina Rampello
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Laura Rapisarda
- Department Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, Catania, Italy
| | - Alessandro Rodolico
- Technical University of Munich, TUM School of Medicine and Health, Department of Psychiatry and Psychotherapy, Munich, Germany
| | - Giuseppina Cantarella
- Department Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, Catania, Italy
| | - Maria Salvina Signorelli
- Psychiatry Unit, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Renato Bernardini
- Department Biomedical and Biotechnological Sciences (BIOMETEC), Section of Pharmacology, University of Catania, Catania, Italy
- Clinical Toxicology Unit, University Hospital of Catania, Catania, Italy
| |
Collapse
|
10
|
Guida CR, Maia JM, Ferreira LFR, Rahdar A, Branco LGS, Soriano RN. Advancements in addressing drug dependence: A review of promising therapeutic strategies and interventions. Prog Neuropsychopharmacol Biol Psychiatry 2024; 134:111070. [PMID: 38908501 DOI: 10.1016/j.pnpbp.2024.111070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Substance dependence represents a pervasive global concern within the realm of public health. Presently, it is delineated as a persistent and recurrent neurological disorder stemming from drug-triggered neuroadaptations in the brain's reward circuitry. Despite the availability of various therapeutic modalities, there has been a steady escalation in the mortality rate attributed to drug overdoses. Substantial endeavors have been directed towards the exploration of innovative interventions aimed at mitigating cravings and drug-induced repetitive behaviors. Within this review, we encapsulate the most auspicious contemporary treatment methodologies, accentuating meta-analyses of efficacious pharmacological and non-pharmacological approaches: including gabapentin, topiramate, prazosin, physical exercise regimens, and cerebral stimulation techniques.
Collapse
Affiliation(s)
- Clara Rodrigues Guida
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | - Juliana Marino Maia
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, MG 35032-620, Brazil
| | | | - Abbas Rahdar
- Department of Physics, Faculty of Sciences, University of Zabol, Zabol 538-98615, Iran
| | - Luiz G S Branco
- Department of Basic and Oral Biology, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-904, Brazil; Department of Physiology, Medical School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP 14040-900, Brazil.
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, MG 35020-360, Brazil.
| |
Collapse
|
11
|
Kelley AT, Incze MA, Baumgartner M, Campbell ANC, Nunes EV, Scharfstein DO. Predictors of urine toxicology and other biologic specimen missingness in randomized trials of substance use disorders. Drug Alcohol Depend 2024; 261:111368. [PMID: 38896944 PMCID: PMC11405181 DOI: 10.1016/j.drugalcdep.2024.111368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/08/2024] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND High levels of missing outcome data for biologically confirmed substance use (BCSU) threaten the validity of substance use disorder (SUD) clinical trials. Underlying attributes of clinical trials could explain BCSU missingness and identify targets for improved trial design. METHODS We reviewed 21 clinical trials funded by the NIDA National Drug Abuse Treatment Clinical Trials Network (CTN) and published from 2005 to 2018 that examined pharmacologic and psychosocial interventions for SUD. We used configurational analysis-a Boolean algebra approach that identifies an attribute or combination of attributes predictive of an outcome-to identify trial design features and participant characteristics associated with high levels of BCSU missingness. Associations were identified by configuration complexity, consistency, coverage, and robustness. We limited results using a consistency threshold of 0.75 and summarized model fit using the product of consistency and coverage. RESULTS For trial design features, the final solution consisted of two pathways: psychosocial treatment as a trial intervention OR larger trial arm size (complexity=2, consistency=0.79, coverage=0.93, robustness score=0.71). For participant characteristics, the final solution consisted of two pathways: interventions targeting individuals with poly- or nonspecific substance use OR younger age (complexity=2, consistency=0.75, coverage=0.86, robustness score=1.00). CONCLUSIONS Psychosocial treatments, larger trial arm size, interventions targeting individuals with poly- or nonspecific substance use, and younger age among trial participants were predictive of missing BCSU data in SUD clinical trials. Interventions to mitigate missing data that focus on these attributes may reduce threats to validity and improve utility of SUD clinical trials.
Collapse
Affiliation(s)
- A Taylor Kelley
- Division of General Internal Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Greater Intermountain Node, National Institute on Drug Abuse Clinical Trial Network, Program of Addiction Research, Clinical Care, Knowledge, and Advocacy (PARCKA), Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Informatics, Decision Enhancement, and Analytic Sciences (IDEAS) Center, VA Salt Lake City Health Care System, Salt Lake City, UT, USA; Vulnerable Veteran Patient-Aligned Care Team, VA Salt Lake City Health Care System, Salt Lake City, UT, USA.
| | - Michael A Incze
- Division of General Internal Medicine, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA; Greater Intermountain Node, National Institute on Drug Abuse Clinical Trial Network, Program of Addiction Research, Clinical Care, Knowledge, and Advocacy (PARCKA), Division of Epidemiology, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Aimee N C Campbell
- New York State Psychiatric Institute, Division on Substance Use Disorders, New York, NY, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Edward V Nunes
- New York State Psychiatric Institute, Division on Substance Use Disorders, New York, NY, USA; Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, USA
| | - Daniel O Scharfstein
- Division of Biostatistics, Department of Population Health Sciences, University of Utah School of Medicine, Salt Lake City, UT, USA
| |
Collapse
|
12
|
Soares-Cardoso C, Leal S, Sá SI, Dantas-Barros R, Dinis-Oliveira RJ, Faria J, Barbosa J. Unraveling the Hippocampal Molecular and Cellular Alterations behind Tramadol and Tapentadol Neurobehavioral Toxicity. Pharmaceuticals (Basel) 2024; 17:796. [PMID: 38931463 PMCID: PMC11206790 DOI: 10.3390/ph17060796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/11/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Tramadol and tapentadol are chemically related opioids prescribed for the analgesia of moderate to severe pain. Although safer than classical opioids, they are associated with neurotoxicity and behavioral dysfunction, which arise as a concern, considering their central action and growing misuse and abuse. The hippocampal formation is known to participate in memory and learning processes and has been documented to contribute to opioid dependence. Accordingly, the present study assessed molecular and cellular alterations in the hippocampal formation of Wistar rats intraperitoneally administered with 50 mg/kg tramadol or tapentadol for eight alternate days. Alterations were found in serum hydrogen peroxide, cysteine, homocysteine, and dopamine concentrations upon exposure to one or both opioids, as well as in hippocampal 8-hydroxydeoxyguanosine and gene expression levels of a panel of neurotoxicity, neuroinflammation, and neuromodulation biomarkers, assessed through quantitative real-time polymerase chain reaction (qRT-PCR). Immunohistochemical analysis of hippocampal formation sections showed increased glial fibrillary acidic protein (GFAP) and decreased cluster of differentiation 11b (CD11b) protein expression, suggesting opioid-induced astrogliosis and microgliosis. Collectively, the results emphasize the hippocampal neuromodulator effects of tramadol and tapentadol, with potential behavioral implications, underlining the need to prescribe and use both opioids cautiously.
Collapse
Affiliation(s)
- Cristiana Soares-Cardoso
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Sandra Leal
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Toxicologic Pathology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Susana I. Sá
- RISE-HEALTH, Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Rua Dr. Plácido da Costa, 4200-450 Porto, Portugal;
| | - Rita Dantas-Barros
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- FOREN-Forensic Science Experts, Av. Dr. Mário Moutinho 33-A, 1400-136 Lisboa, Portugal
| | - Juliana Faria
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Joana Barbosa
- Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, 4585-116 Gandra, Portugal; (C.S.-C.); (S.L.); (R.D.-B.); or (R.J.D.-O.)
- UCIBIO—Applied Molecular Biosciences Unit, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), 4585-116 Gandra, Portugal
| |
Collapse
|
13
|
Poliakova N, Shrier LA, Harris SK, Bélanger RE. Predicting Time to Return to Cannabis Use After a Cessation Attempt: Impact of Cumulated Exposure to Nicotine-Containing Products. Tob Use Insights 2024; 17:1179173X241259603. [PMID: 38846268 PMCID: PMC11155328 DOI: 10.1177/1179173x241259603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 05/09/2024] [Indexed: 06/09/2024] Open
Abstract
Objective: Cannabis is frequently co-used with tobacco/nicotine products, especially among young adults. Little is known about the effects of this co-use on cannabis cessation outcomes. Within a sample of young adults using cannabis frequently (current use of ≥5 days/week in the past 3 months), this study aimed to (a) document sources of exposure to tobacco/nicotine products, whether used simultaneously with cannabis or on different occasions, (b) examine if the level of cumulated exposure to tobacco/nicotine (self-reported or from biochemical testing) could predict time to cannabis lapse during a cannabis abstinence period, and (c) explore the relationship between nicotine/tobacco exposure and time to cannabis lapse according to tobacco cigarette smoking status. Method: Urine cotinine measures and self-reported data on use of different tobacco/nicotine products, collected from 32 participants (aged 19 to 23), were analyzed to predict time to lapse during a 2-week period of attempted abstinence from cannabis, controlling for cannabis dependence and sex. Results: Half of participants (56.3%) used at least one tobacco/nicotine product. Higher urine cotinine, representing higher cumulated tobacco/nicotine exposure, was related to a higher risk of lapsing (Hazard Ratio [HR] = 1.64; 95%CI [1.04, 2.58]). The risk of lapsing was even higher ([HR] = 3.46; 95%CI [1.17, 10.25]) among heavily tobacco/nicotine exposed (>600 ng/mL, urine cotinine) participants than among unexposed (<50 ng/mL) or lightly/moderately exposed (50-600 ng/mL) participants. Among those smoking cigarettes (solely or in combination with other products), there was no relation between cotinine level and time to lapse, likely due to a reduced variability in abstinence probability and a high likelihood of lapse observed for higher cotinine levels, mainly achieved by cigarette use. Conclusions: With a rapidly changing landscape of tobacco/nicotine use, our results underscore the need to consider all sources of tobacco/nicotine exposure to fully understand the specific and cumulative contributions of tobacco/nicotine to cannabis cessation outcomes.
Collapse
Affiliation(s)
- Natalia Poliakova
- Research Centre of CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Lydia A. Shrier
- Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Sion Kim Harris
- Boston Children’s Hospital and Harvard Medical School, Boston, MA, USA
| | - Richard E. Bélanger
- Research Centre of CHU de Québec-Université Laval, Quebec City, QC, Canada
- Faculty of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
14
|
Donlon J, Kumari P, Varghese SP, Bai M, Florentin OD, Frost ED, Banks J, Vadlapatla N, Kam O, Shad MU, Rahman S, Abulseoud OA, Stone TW, Koola MM. Integrative Pharmacology in the Treatment of Substance Use Disorders. J Dual Diagn 2024; 20:132-177. [PMID: 38117676 DOI: 10.1080/15504263.2023.2293854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
The detrimental physical, mental, and socioeconomic effects of substance use disorders (SUDs) have been apparent to the medical community for decades. However, it has become increasingly urgent in recent years to develop novel pharmacotherapies to treat SUDs. Currently, practitioners typically rely on monotherapy. Monotherapy has been shown to be superior to no treatment at all for most substance classes. However, many randomized controlled trials (RCTs) have revealed that monotherapy leads to poorer outcomes when compared with combination treatment in all specialties of medicine. The results of RCTs suggest that monotherapy frequently fails since multiple dysregulated pathways, enzymes, neurotransmitters, and receptors are involved in the pathophysiology of SUDs. As such, research is urgently needed to determine how various neurobiological mechanisms can be targeted by novel combination treatments to create increasingly specific yet exceedingly comprehensive approaches to SUD treatment. This article aims to review the neurobiology that integrates many pathophysiologic mechanisms and discuss integrative pharmacology developments that may ultimately improve clinical outcomes for patients with SUDs. Many neurobiological mechanisms are known to be involved in SUDs including dopaminergic, nicotinic, N-methyl-D-aspartate (NMDA), and kynurenic acid (KYNA) mechanisms. Emerging evidence indicates that KYNA, a tryptophan metabolite, modulates all these major pathophysiologic mechanisms. Therefore, achieving KYNA homeostasis by harmonizing integrative pathophysiology and pharmacology could prove to be a better therapeutic approach for SUDs. We propose KYNA-NMDA-α7nAChRcentric pathophysiology, the "conductor of the orchestra," as a novel approach to treat many SUDs concurrently. KYNA-NMDA-α7nAChR pathophysiology may be the "command center" of neuropsychiatry. To date, extant RCTs have shown equivocal findings across comparison conditions, possibly because investigators targeted single pathophysiologic mechanisms, hit wrong targets in underlying pathophysiologic mechanisms, and tested inadequate monotherapy treatment. We provide examples of potential combination treatments that simultaneously target multiple pathophysiologic mechanisms in addition to KYNA. Kynurenine pathway metabolism demonstrates the greatest potential as a target for neuropsychiatric diseases. The investigational medications with the most evidence include memantine, galantamine, and N-acetylcysteine. Future RCTs are warranted with novel combination treatments for SUDs. Multicenter RCTs with integrative pharmacology offer a promising, potentially fruitful avenue to develop novel therapeutics for the treatment of SUDs.
Collapse
Affiliation(s)
- Jack Donlon
- Cooper Medical School of Rowan University, Camden, New Jersey, USA
| | - Pooja Kumari
- Community Living Trent Highlands, Peterborough, Canada
| | - Sajoy P Varghese
- Addiction Recovery Treatment Services, Veterans Affairs Northern California Health Care System, University of California, Davis, Sacramento, California, USA
| | - Michael Bai
- Columbia University, New York, New York, USA
| | - Ori David Florentin
- Department of Psychiatry, Westchester Medical Center, Valhalla, New York, USA
| | - Emma D Frost
- Department of Neurology, Cooper University Health Care, Camden, New Jersey, USA
| | - John Banks
- Talkiatry Mental Health Clinic, New York, New York, USA
| | - Niyathi Vadlapatla
- Thomas Jefferson High School for Science and Technology, Alexandria, Virginia, USA
| | - Olivia Kam
- Stony Brook University Renaissance School of Medicine, Stony Brook, New York, USA
| | - Mujeeb U Shad
- Department of Psychiatry, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Shafiqur Rahman
- Department of Pharmaceutical Sciences, College of Pharmacy, South Dakota State University, Brookings, South Dakota, USA
| | - Osama A Abulseoud
- Department of Psychiatry and Psychology, Alix School of Medicine at Mayo Clinic, Phoenix, Arizona, USA
| | - Trevor W Stone
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences (NDORMS), University of Oxford, Oxford, UK
| | - Maju Mathew Koola
- Department of Psychiatry and Behavioral Health, Cooper University Health Care, Cooper Medical School of Rowan University, Camden, New Jersey, USA
| |
Collapse
|
15
|
Petrilli K, Lawn W, Lees R, Mokrysz C, Borissova A, Ofori S, Trinci K, Dos Santos R, Leitch H, Soni S, Hines LA, Lorenzetti V, Curran HV, Freeman TP. Enhanced cannabis timeline followback (EC-TLFB): Comprehensive assessment of cannabis use including standard THC units and validation through biological measures. Addiction 2024; 119:772-783. [PMID: 38105033 DOI: 10.1111/add.16405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023]
Abstract
AIMS The aims of this study were to present an enhanced cannabis timeline followback (EC-TLFB) enabling comprehensive assessment of cannabis use measures, including standard tetrahydrocannabinol (THC) units, and to validate these against objectively indexed urinary 11-nor-9-carboxy-tetrahydrocannabinol (THC-COOH) concentrations. DESIGN We used cross-sectional baseline data from the 'CannTeen' observational longitudinal study. SETTING The study was conducted in London, UK. PARTICIPANTS A total of 147 participants who used cannabis regularly took part in the study (n = 71 female, n = 76 male; mean age = 21.90, standard deviation = 5.32). MEASUREMENTS The EC-TLFB was used to calculate frequency of cannabis use, method of administration, including co-administration with tobacco, amount of cannabis used (measured with unaided self-report and also using pictorial aided self-report) and type of cannabis product (flower, hash) which was used to estimate THC concentration (both from published data on THC concentration of products and analysis of cannabis samples donated by participants in this study). We calculated total weekly standard THC units (i.e. 5 mg THC for all cannabis products and methods of administration) using the EC-TLFB. The outcome variable for validation of past week EC-TLFB assessments was creatinine-normalized carboxy-tetrahydrocannabinol (THC-COOH) in urine. FINDINGS All measures of cannabis exposure included in this analysis were positively correlated with levels of THC-COOH in urine (r = 0.41-0.52). Standard THC units, calculated with average concentrations of THC in cannabis in the UK and unaided self-report measures of amount of cannabis used in grams showed the strongest correlation with THC-COOH in urine (r = 0.52, 95% bias-corrected and accelerated = 0.26-0.70). CONCLUSIONS The enhanced cannabis timeline followback (EC-TLFB) can provide a valid assessment of a comprehensive set of cannabis use measures including standard tetrahydrocannabinol units as well as and traditional TLFB assessments (e.g. frequency of use and grams of cannabis use).
Collapse
Affiliation(s)
- Kat Petrilli
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, Bath, UK
| | - Will Lawn
- Department of Psychology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Department of Addictions, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Clinical Psychopharmacology Unit, University College London, London, UK
| | - Rachel Lees
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, Bath, UK
| | - Claire Mokrysz
- Clinical Psychopharmacology Unit, University College London, London, UK
| | - Anya Borissova
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- NIHR University College London Hospitals Biomedical Research Centre, University College Hospital, London, UK
| | - Shelan Ofori
- Clinical Psychopharmacology Unit, University College London, London, UK
| | - Katie Trinci
- Clinical Psychopharmacology Unit, University College London, London, UK
| | | | - Harry Leitch
- Clinical Psychopharmacology Unit, University College London, London, UK
| | - Shilpa Soni
- Clinical Psychopharmacology Unit, University College London, London, UK
| | - Lindsey A Hines
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, Bath, UK
- Population Health Science, Bristol Medical School, University of Bristol, Bristol, UK
| | - Valentina Lorenzetti
- Neuroscience of Addiction and Mental Health Programme, the Healthy Brain and Mind Research Centre, School of Behavioural and Health Sciences, Faculty of Health Sciences, Australian Catholic University, Melbourne, Australia
| | - H Valerie Curran
- Clinical Psychopharmacology Unit, University College London, London, UK
| | - Tom P Freeman
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, Bath, UK
| |
Collapse
|
16
|
Hudson D, Howarth N, Idalsoaga F, Song YN, Islam A, Theiventhiran S, Díaz LA, Arab JP. Addiction and Liver Disease: Exploring the Complex Relationship and Implications for Clinical Management. CURRENT HEPATOLOGY REPORTS 2024; 23:110-122. [DOI: 10.1007/s11901-024-00630-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 01/01/2024] [Indexed: 01/12/2025]
|
17
|
Abstract
This review summarizes treatments for cannabis use disorder (CUD) in adolescents. The best supported CUD treatments are cognitive behavioral psychotherapies, including family-based models that facilitate environmental changes and youth-focused models that incorporate skills training, motivational interviewing, and contingency management to promote reductions in cannabis use. Some medications show promise in reducing cannabis craving and withdrawal symptoms. Further research is needed on the efficacy and implementation of existing treatments given the changes in cannabis use trends over time and on emerging technologies that may expand access to evidence-based CUD treatments.
Collapse
Affiliation(s)
- Zachary W Adams
- Department of Psychiatry, Indiana University School of Medicine, 410 West 10th Street, Suite 2000, Indianapolis, IN 46202, USA.
| | - Brigid R Marriott
- Department of Psychiatry, Indiana University School of Medicine, 410 West 10th Street, Suite 2000, Indianapolis, IN 46202, USA
| | - Leslie A Hulvershorn
- Department of Psychiatry, Indiana University School of Medicine, 1002 Wishard Boulevard, Suite 4110, Indianapolis, IN 46202, USA
| | - Jesse D Hinckley
- Department of Psychiatry, University of Colorado School of Medicine, 13001 East 17th Place, MS-F546, Aurora, CO 80045, USA. https://twitter.com/JHinckleyMDPhD
| |
Collapse
|
18
|
Tomko RL, Wolf BJ, McClure EA, Carpenter MJ, Magruder KM, Squeglia LM, Gray KM. Who responds to a multi-component treatment for cannabis use disorder? Using multivariable and machine learning models to classify treatment responders and non-responders. Addiction 2023; 118:1965-1974. [PMID: 37132085 PMCID: PMC10524796 DOI: 10.1111/add.16226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 04/13/2023] [Indexed: 05/04/2023]
Abstract
BACKGROUND AND AIMS Treatments for cannabis use disorder (CUD) have limited efficacy and little is known about who responds to existing treatments. Accurately predicting who will respond to treatment can improve clinical decision-making by allowing clinicians to offer the most appropriate level and type of care. This study aimed to determine whether multivariable/machine learning models can be used to classify CUD treatment responders versus non-responders. METHODS This secondary analysis used data from a National Drug Abuse Treatment Clinical Trials Network multi-site outpatient clinical trial in the United States. Adults with CUD (n = 302) received 12 weeks of contingency management, brief cessation counseling and were randomized to receive additionally either (1) N-Acetylcysteine or (2) placebo. Multivariable/machine learning models were used to classify treatment responders (i.e. two consecutive negative urine cannabinoid tests or a 50% reduction in days of use) versus non-responders using baseline demographic, medical, psychiatric and substance use information. RESULTS Prediction performance for various machine learning and regression prediction models yielded area under the curves (AUCs) >0.70 for four models (0.72-0.77), with support vector machine models having the highest overall accuracy (73%; 95% CI = 68-78%) and AUC (0.77; 95% CI = 0.72, 0.83). Fourteen variables were retained in at least three of four top models, including demographic (ethnicity, education), medical (diastolic/systolic blood pressure, overall health, neurological diagnosis), psychiatric (depressive symptoms, generalized anxiety disorder, antisocial personality disorder) and substance use (tobacco smoker, baseline cannabinoid level, amphetamine use, age of experimentation with other substances, cannabis withdrawal intensity) characteristics. CONCLUSIONS Multivariable/machine learning models can improve on chance prediction of treatment response to outpatient cannabis use disorder treatment, although further improvements in prediction performance are likely necessary for decisions about clinical care.
Collapse
Affiliation(s)
- Rachel L. Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Bethany J. Wolf
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Erin A. McClure
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Matthew J. Carpenter
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Kathryn M. Magruder
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Lindsay M. Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Kevin M. Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
19
|
Herbst ED, Pennington DL, Borsari B, Manuel J, Yalch M, Alcid E, Martinez Rivas M, Delacruz J, Rossi N, Garcia B, Wong N, Batki SL. N-acetylcysteine for smoking cessation among dual users of tobacco and cannabis: Protocol and rationale for a randomized controlled trial. Contemp Clin Trials 2023; 131:107250. [PMID: 37271412 PMCID: PMC10847050 DOI: 10.1016/j.cct.2023.107250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/25/2023] [Accepted: 05/30/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND Tobacco and cannabis co-use is a growing public health problem. The synergistic effects of cannabis and nicotine on neurobiological systems that mediate reward and shared environmental cues reinforcing use may make tobacco smoking cessation more difficult. N-acetylcysteine (NAC), an FDA-approved medication and over-the-counter supplement, has shown promise in animal studies and randomized controlled trials (RCTs) in reducing tobacco and cannabis craving and use. NAC's potential efficacy in treating addiction may be attributable to its central nervous system effects in reducing excessive glutamatergic activity, oxidative stress, and inflammation. To date, no RCT has examined NAC for smoking cessation among dual users of tobacco and cannabis. METHOD In a double-blind, placebo-controlled RCT, we will examine NAC for smoking cessation among dual users of tobacco and cannabis. Sixty adult cigarette-cannabis co-users are randomized to receive NAC 3600 mg per day or placebo over 8 weeks. Participants in both groups receive 8 weekly cognitive behavioral therapy sessions addressing smoking cessation and cannabis reduction. Outcomes are assessed at Weeks 0, 4, 8, and 12. Primary aims are to determine NAC's efficacy in decreasing cigarette craving, nicotine dependence, and use; and cannabis craving and use. Exploratory aims include examination of changes in neurocognition with NAC and their potential mediational effects on cigarette and cannabis use outcomes. CONCLUSION Results will inform smoking cessation treatment among dual users of tobacco and cannabis. CLINICALTRIALS gov Identifier: NCT04627922.
Collapse
Affiliation(s)
- Ellen D Herbst
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA.
| | - David L Pennington
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Brian Borsari
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Jennifer Manuel
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Matthew Yalch
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA; Palo Alto University, 1791 Arastradero Rd., Palo Alto, CA 94304, USA
| | - Eric Alcid
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Madeline Martinez Rivas
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Joannalyn Delacruz
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Nathan Rossi
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Brianna Garcia
- California Institute of Technology, 1200 E California Blvd, Pasadena, CA 91125, USA
| | - Natalie Wong
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| | - Steven L Batki
- Mental Health Service (116B), San Francisco VA Health Care System, 4150 Clement St., San Francisco, CA 94121, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, 401 Parnassus Ave., San Francisco, CA 94143, USA
| |
Collapse
|
20
|
Preto MC, Kortas GT, Blaas IK, Lassi DLS, Waisman Campos M, Torales J, Ventriglio A, de Azevedo-Marques Périco C, de Andrade AG, Castaldelli-Maia JM. Unravelling the landscape of Cannabis craving pharmacological treatments: a PRISMA-guided review of evidence. Int Rev Psychiatry 2023; 35:434-449. [PMID: 38299652 DOI: 10.1080/09540261.2023.2231540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/27/2023] [Indexed: 02/02/2024]
Abstract
Currently, few treatments are available for craving in general, and none of them have received approval for cannabis craving. The objective of this review is to evaluate existing studies analysing treatments for cannabis craving and explore novel treatment possibilities for these patients. The study followed PRISMA guidelines and conducted an extensive database search. Inclusion criteria included human randomised controlled trials examining drug effects on craving symptoms. Exclusion criteria involved studies unrelated to craving, non-pharmacological treatments, duplicates, and non-English/Spanish/Portuguese articles. Our included 22 studies that investigated a wide range of compounds used for cravings related to other drugs, as well as interventions based on healthcare professionals' empirical knowledge. The current pharmacological treatments largely involve off-label drug use and the utilisation of cannabinoid-based medications, such as combinations of THC and lofexidine, oxytocin, progesterone, and N-acetylcysteine. These emerging treatments show promise and have the potential to revolutionise current clinical practices, but further investigation is needed to establish their efficacy. In this context, it is essential to consider non-pharmacological interventions, such as psychotherapy and behavioural treatments. These approaches play a crucial role in complementing pharmacological interventions and addressing the complex nature of the disorder.
Collapse
Affiliation(s)
- Mayra Cruz Preto
- Medical School, Universidade Cidade de São Paulo (UNICID), São Paulo, Brazil
| | - Guilherme Trevizan Kortas
- Perdizes Intitute (IPer), Clinics Hospital of Medical School (HCFMUSP), University of São Paulo, São Paulo, Brazil
- Department of Psychiatry, Medical School, University of São Paulo, São Paulo, Brazil
- Sirio-Libanês Hospital, São Paulo, Brazil
| | - Israel Kanaan Blaas
- Perdizes Intitute (IPer), Clinics Hospital of Medical School (HCFMUSP), University of São Paulo, São Paulo, Brazil
- Department of Neuroscience, Medical School, FMABC University Center, Santo André, Brazil
| | - Dangela Layne Silva Lassi
- Perdizes Intitute (IPer), Clinics Hospital of Medical School (HCFMUSP), University of São Paulo, São Paulo, Brazil
| | - Marcela Waisman Campos
- Department of Cognitive Neurology, Neuropsychiatry, and Neuropsychology, FLENI, Buenos Aires, Argentina
| | - Julio Torales
- Department of Medical Psychology, School of Medical Sciences, University of Asuncion, San Lorenzo, Paraguay
| | - Antonio Ventriglio
- Department of Experimental Medicine, Medical School, Medical School, University of Foggia, Foggia, Italy
| | | | - Arthur Guerra de Andrade
- Perdizes Intitute (IPer), Clinics Hospital of Medical School (HCFMUSP), University of São Paulo, São Paulo, Brazil
- Department of Psychiatry, Medical School, University of São Paulo, São Paulo, Brazil
- Sirio-Libanês Hospital, São Paulo, Brazil
- Department of Neuroscience, Medical School, FMABC University Center, Santo André, Brazil
| | - João Mauricio Castaldelli-Maia
- Department of Psychiatry, Medical School, University of São Paulo, São Paulo, Brazil
- Department of Neuroscience, Medical School, FMABC University Center, Santo André, Brazil
| |
Collapse
|
21
|
Yeap ZJS, Marsault J, George TP, Mizrahi R, Rabin RA. Does tobacco dependence worsen cannabis withdrawal in people with and without schizophrenia-spectrum disorders? Am J Addict 2023; 32:367-375. [PMID: 36815595 DOI: 10.1111/ajad.13394] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Rates of cannabis use disorder (CUD) are higher in people with schizophrenia than in the general population. Irrespective of psychiatric diagnosis, tobacco co-use is prevalent in those with CUD and leads to poor cannabis cessation outcomes. The cannabis withdrawal syndrome is well-established and increases cannabis relapse risk. We investigated whether cannabis withdrawal severity differed as a function of high versus no/low tobacco dependence and psychiatric diagnosis in individuals with CUD. METHOD Men with CUD (N = 55) were parsed into four groups according to schizophrenia diagnosis and tobacco dependence severity using the Fagerstrom Test for Nicotine Dependence (FTND): men with schizophrenia with high tobacco dependence (SCT+, n = 13; FTND ≥ 5) and no/low tobacco dependence (SCT-, n = 22; FTND ≤ 4), and nonpsychiatric controls with high (CCT+, n = 7; FTND ≥ 5) and no/low (CCT-, n = 13; FTND ≤ 4) tobacco dependence. Participants completed the Marijuana Withdrawal Checklist following 12-h of cannabis abstinence. RESULTS There was a significant main effect of tobacco dependence on cannabis withdrawal severity (p < .001). Individuals with high tobacco dependence had significantly greater cannabis withdrawal severity (M = 13.85 [6.8]) compared to individuals with no/low tobacco dependence (M = 6.49, [4.9]). Psychiatric diagnosis and the interaction effects were not significant. Lastly, cannabis withdrawal severity positively correlated with FTND (r = .41, p = .002). CONCLUSION AND SCIENTIFIC SIGNIFICANCE Among individuals with CUD and high tobacco dependence, cannabis withdrawal severity was elevated twofold, irrespective of diagnosis, relative to individuals with CUD and no/low tobacco dependence. Findings from this study emphasize the importance of addressing tobacco co-use when treating CUD.
Collapse
Affiliation(s)
- Zac J S Yeap
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Justine Marsault
- Integrated Program in Neuroscience, McGill University, Montreal, Quebec, Canada
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
| | - Tony P George
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Centre for Complex Interventions and Addictions Division, Centre for Addiction and Mental Health (CAMH), Toronto, Ontario, Canada
| | - Romina Mizrahi
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| | - Rachel A Rabin
- Douglas Mental Health University Institute, Verdun, Quebec, Canada
- Department of Psychiatry, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
22
|
Schmidt RD, Horigian VE, Shmueli-Blumberg D, Hefner K, Feinberg J, Kondapaka R, Feaster DJ, Duan R, Gonzalez S, Davis C, Vena A, Marín-Navarrete R, Tross S. High suicidality predicts overdose events among people with substance use disorder: A latent class analysis. Front Public Health 2023; 11:1150062. [PMID: 37261240 PMCID: PMC10228506 DOI: 10.3389/fpubh.2023.1150062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 03/14/2023] [Indexed: 06/02/2023] Open
Abstract
Introduction Suicide is the tenth leading cause of death in the United States and continues to be a major public health concern. Suicide risk is highly prevalent among individuals with co-occurring substance use disorders (SUD) and mental health disorders, making them more prone to adverse substance use related outcomes including overdose. Identifying individuals with SUD who are suicidal, and therefore potentially most at risk of overdose, is an important step to address the synergistic epidemics of suicides and overdose fatalities in the United States. The current study assesses whether patterns of suicidality endorsement can indicate risk for substance use and overdose. Methods Latent class analysis (LCA) was used to assess patterns of item level responses to the Concise Health Risk Tracking Self-Report (CHRT-SR), which measures thoughts and feelings associated with suicidal propensity. We used data from 2,541 participants with SUD who were enrolled across 8 randomized clinical trials in the National Drug Abuse Treatment Clinical Trials Network from 2012 to 2021. Characteristics of individuals in each class were assessed, and multivariable logistic regression was performed to examine class membership as a predictor of overdose. LCA was also used to analyze predictors of substance use days. Results Three classes were identified and discussed: Class (1) Minimal Suicidality, with low probabilities of endorsing each CHRT-SR construct; Class (2) Moderate Suicidality, with high probabilities of endorsing pessimism, helplessness, and lack of social support, but minimal endorsement of despair or suicidal thoughts; and Class (3) High Suicidality with high probabilities of endorsing all constructs. Individuals in the High Suicidality class comprise the highest proportions of males, Black/African American individuals, and those with a psychiatric history and baseline depression, as compared with the other two classes. Regression analysis revealed that those in the High Suicidality class are more likely to overdose as compared to those in the Minimal Suicidality class (p = 0.04). Conclusion Suicidality is an essential factor to consider when building strategies to screen, identify, and address individuals at risk for overdose. The integration of detailed suicide assessment and suicide risk reduction is a potential solution to help prevent suicide and overdose among people with SUD.
Collapse
Affiliation(s)
- Renae D. Schmidt
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Viviana E. Horigian
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | | | - Judith Feinberg
- Departments of Behavioral Medicine and Psychiatry and Medicine/Infectious Diseases, West Virginia University School of Medicine, Morgantown, WV, United States
| | | | - Daniel J. Feaster
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rui Duan
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Sophia Gonzalez
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Carly Davis
- Department of Public Health Sciences, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Ashley Vena
- The Emmes Company, LLC, Rockville, MD, United States
| | - Rodrigo Marín-Navarrete
- Division of Research and Translational Education, Centros de Integración Juvenil A.C, Mexico City, Mexico
| | - Susan Tross
- Department of Psychiatry, Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
23
|
Walters KJ, Baker NL, Tomko RL, Gray KM, Carpenter MJ, McClure EA. Determining the impact of cannabis use and severity on tobacco cessation outcomes: study protocol for a prospective tobacco treatment trial. BMC Psychol 2023; 11:25. [PMID: 36698194 PMCID: PMC9875760 DOI: 10.1186/s40359-023-01060-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 01/20/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Several evidence-based tobacco cessation treatment strategies exist, though significant barriers to cessation remain which must be addressed to improve abstinence rates for sub-populations of those smoking cigarettes. Cannabis co-use among those who use tobacco is common and appears to be increasing among adults in the United States (US). The literature evaluating the impact of cannabis use on tobacco cessation has been mixed and has several important limitations, which precludes development of treatment recommendations specific to individuals who use tobacco and co-use cannabis. To date, no prospective studies have evaluated the impact of cannabis use and severity on tobacco cessation or quantified cannabis use changes during tobacco treatment to assess for concurrent reductions, abstinence, or compensatory (i.e., increased) cannabis use. This study's aims are to: (1) evaluate tobacco cessation outcomes among participants who co-use cannabis compared to participants only using tobacco, (2) using daily diaries and biochemical verification, assess changes in cannabis use during tobacco treatment, and (3) assess for a dose-dependent impact of cannabis use on tobacco cessation. METHOD A multi-site, prospective, quasi-experimental 12-week tobacco treatment trial enrolling treatment-seeking adults (ages 18-40; N = 208) from three sites across South Carolina (US) who use tobacco daily and oversampling (2:1) those who co-use cannabis. Participants receive tobacco cessation pharmacotherapy (varenicline) paired with behavioral support, while cannabis use is not addressed as part of treatment. The primary outcome is 7-day point prevalence tobacco abstinence at the week 12 end of treatment visit, measured via biochemical verification and self-report. Secondary outcome measures include changes in cannabis use (via biochemical verification and self-report) during tobacco cessation treatment. DISCUSSION Results from this trial have the potential to inform tobacco treatment among those co-using cannabis, which may require a tailored approach to address the role of cannabis in quitting tobacco. TRIAL REGISTRATION The trial is registered with ClinicalTrials.gov: NCT04228965. January 14th, 2020.
Collapse
Affiliation(s)
- Kyle J Walters
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., MSC 861, Charleston, SC, 29425, USA
| | - Nathaniel L Baker
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Rachel L Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., MSC 861, Charleston, SC, 29425, USA
| | - Kevin M Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., MSC 861, Charleston, SC, 29425, USA
| | - Matthew J Carpenter
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., MSC 861, Charleston, SC, 29425, USA
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St., Charleston, SC, 29425, USA
| | - Erin A McClure
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, 67 President St., MSC 861, Charleston, SC, 29425, USA.
- Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St., Charleston, SC, 29425, USA.
| |
Collapse
|
24
|
Adams ZW, Marriott BR, Hulvershorn LA, Hinckley J. Treatment of Adolescent Cannabis Use Disorders. Child Adolesc Psychiatr Clin N Am 2023; 32:141-155. [PMID: 36410901 PMCID: PMC10097012 DOI: 10.1016/j.chc.2022.07.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This review summarizes treatments for cannabis use disorder (CUD) in adolescents. The best supported CUD treatments are cognitive behavioral psychotherapies, including family-based models that facilitate environmental changes and youth-focused models that incorporate skills training, motivational interviewing, and contingency management to promote reductions in cannabis use. Some medications show promise in reducing cannabis craving and withdrawal symptoms. Further research is needed on the efficacy and implementation of existing treatments given the changes in cannabis use trends over time and on emerging technologies that may expand access to evidence-based CUD treatments.
Collapse
Affiliation(s)
- Zachary W Adams
- Department of Psychiatry, Indiana University School of Medicine, 410 West 10th Street, Suite 2000, Indianapolis, IN 46202, USA.
| | - Brigid R Marriott
- Department of Psychiatry, Indiana University School of Medicine, 410 West 10th Street, Suite 2000, Indianapolis, IN 46202, USA
| | - Leslie A Hulvershorn
- Department of Psychiatry, Indiana University School of Medicine, 1002 Wishard Boulevard, Suite 4110, Indianapolis, IN 46202, USA
| | - Jesse Hinckley
- Department of Psychiatry, University of Colorado School of Medicine, 13001 East 17th Place, MS-F546, Aurora, CO 80045, USA. https://twitter.com/JHinckleyMDPhD
| |
Collapse
|
25
|
Padoei F, Mamsharifi P, Hazegh P, Boroumand H, Ostadmohammady F, Abbaszadeh-Mashkani S, Banafshe HR, Matini AH, Ghaderi A, Dehkohneh SG. The therapeutic effect of N-acetylcysteine as an add-on to methadone maintenance therapy medication in outpatients with substance use disorders: A randomized, double-blind, placebo-controlled clinical trial. Brain Behav 2023; 13:e2823. [PMID: 36448959 PMCID: PMC9847617 DOI: 10.1002/brb3.2823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 10/20/2022] [Accepted: 11/01/2022] [Indexed: 12/02/2022] Open
Abstract
OBJECTIVE Patients with substance use disorders (SUD) under methadone maintenance therapy (MMT) are susceptible to a number of complications (psychological and metabolic disorders). Evidence studies have shown the roles of the glutamatergic system in addiction. N-Acetylcysteine (NAC) enhances extracellular glutamate, and is effective in the treatment of neuropsychiatric disorders. We assessed oral NAC as an add-on to MMT medication for the treatment of SUD. METHODS In the current randomized, double-blind, placebo-controlled clinical trial, outpatients with SUD under MMT who were 18-60 years old received 2400 mg/day NAC (n = 30) or placebo (n = 30) for 12 weeks. Psychological status and metabolic biomarkers were assessed at baseline and the end of the trial. RESULTS Compared with the placebo group, NAC treatment resulted in a significant improvement in depression score (β -2.36; 95% CI, -3.97, -0.76; p = .005), and anxiety score (β -1.82; 95% CI, -3.19, -0.44; p = .01). Furthermore, NAC treatment resulted in a significant elevation in total antioxidant capacity levels (β 72.28 mmol/L; 95% CI, 11.36, 133.19; p = .02), total glutathione (GSH) levels (β 81.84 μmol/L; 95% CI, 15.40, 148.28; p = .01), and a significant reduction in high-sensitivity C-reactive protein levels (β -0.89 mg/L; 95% CI, -1.50, -0.28; p = .005), and homeostasis model of assessment-insulin resistance (β -0.33; 95% CI, -0.65, -0.009; p = .04), compared with the placebo group. CONCLUSION In the current study, improvement in depression and anxiety symptoms as well as some metabolic profiles with NAC treatment for 12 weeks in outpatients with SUD under MMT was detected.
Collapse
Affiliation(s)
- Fateme Padoei
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Peyman Mamsharifi
- Department of Psychology, Allameh Tabataba'i University, Tehran, Iran
| | - Pooya Hazegh
- Department of Psychiatry, Kashan University of Medical Sciences, Kashan, Iran
| | - Homa Boroumand
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | | | | | - Hamid Reza Banafshe
- Physiology Research Center, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Hassan Matini
- Department of Clinical Pathology, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Ghaderi
- Department of Addiction studies, School of Medical, Kashan University of Medical Sciences, Kashan, Iran.,Clinical Research Development Unit-Matini/Kargarnejad Hospital, Kashan University of Medical Sciences, Kashan, Iran
| | - Somayeh Ghadami Dehkohneh
- Department of Pharmacy, Acharya BM ready college of Pharmacy, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| |
Collapse
|
26
|
Roydhouse J, Tomko RL, Gray KM, Gutman R. Assessment of patient perception of treatment assignment and patient-reported outcomes in a cannabis use disorder trial. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2022; 48:651-661. [PMID: 35904459 DOI: 10.1080/00952990.2022.2097918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Background: Blinding is a cornerstone of trial methodology. Prior work indicates participant-perceived assignment may be associated with trial outcomes. Less is known about how perception changes over time and if this is associated with outcomes.Objectives: To evaluate if participants change their perception of assignment over time in a blinded trial, and if perception is associated with different types of patient-reported outcomes (PROs).Methods: This was a secondary analysis of data from the Achieving Cannabis Cessation-Evaluating N-Acetylcysteine Treatment (ACCENT) trial, which evaluated the efficacy of N-acetylcysteine (NAC) relative to placebo for treating cannabis use disorder. Participants (N = 234; 164 men, 70 women) were asked at weeks 5 and 9 what treatment (placebo or NAC) they believed they were receiving. We included PROs proximal (cannabis-associated problems, craving) and distal (anxiety) to the intervention. Analysis was by multiple linear regression and mixed models.Results: Approximately 20% of participants in both arms changed their perception over time. Relative to participants who consistently perceived assignment to placebo, participants who consistently perceived assignment to NAC did not always have comparatively better average scores (coefficient -3.3 [95% CI: -7.0, 0.5]). In some analyses, participants who switched to guessing NAC from placebo had comparatively better average scores (coefficient -3.0 [95% CI: -9.3, 3.4]), but this was inconsistent across outcomes or strata defined by actual assignment or guess accuracy.Conclusion: The study suggests that the proportion of individuals who switch their perception over time is modest. However, this group may influence the estimates of intervention effects on some PROs.
Collapse
Affiliation(s)
- Jessica Roydhouse
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.,Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI, USA
| | - Rachel L Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Kevin M Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Roee Gutman
- Department of Biostatistics, Brown University School of Public Health, Providence, RI, USA
| |
Collapse
|
27
|
Smiley CE, Wood SK. Stress- and drug-induced neuroimmune signaling as a therapeutic target for comorbid anxiety and substance use disorders. Pharmacol Ther 2022; 239:108212. [PMID: 35580690 DOI: 10.1016/j.pharmthera.2022.108212] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 10/18/2022]
Abstract
Stress and substance use disorders remain two of the most highly prevalent psychiatric conditions and are often comorbid. While individually these conditions have a debilitating impact on the patient and a high cost to society, the symptomology and treatment outcomes are further exacerbated when they occur together. As such, there are few effective treatment options for these patients, and recent investigation has sought to determine the neural processes underlying the co-occurrence of these disorders to identify novel treatment targets. One such mechanism that has been linked to stress- and addiction-related conditions is neuroimmune signaling. Increases in inflammatory factors across the brain have been heavily implicated in the etiology of these disorders, and this review seeks to determine the nature of this relationship. According to the "dual-hit" hypothesis, also referred to as neuroimmune priming, prior exposure to either stress or drugs of abuse can sensitize the neuroimmune system to be hyperresponsive when exposed to these insults in the future. This review completes an examination of the literature surrounding stress-induced increases in inflammation across clinical and preclinical studies along with a summarization of the evidence regarding drug-induced alterations in inflammatory factors. These changes in neuroimmune profiles are also discussed within the context of their impact on the neural circuitry responsible for stress responsiveness and addictive behaviors. Further, this review explores the connection between neuroimmune signaling and susceptibility to these conditions and highlights the anti-inflammatory pharmacotherapies that may be used for the treatment of stress and substance use disorders.
Collapse
Affiliation(s)
- Cora E Smiley
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| | - Susan K Wood
- Department of Pharmacology, Physiology, and Neuroscience; University of South Carolina School of Medicine, Columbia, SC 29209, United States of America; WJB Dorn Veterans Administration Medical Center, Columbia, SC 29209, United States of America.
| |
Collapse
|
28
|
Horigian VE, Schmidt RD, Shmueli-Blumberg D, Hefner K, Feinberg J, Kondapaka R, Feaster DJ, Duan R, Gonzalez S, Davis C, Marín-Navarrete R, Tross S. Suicidality as a Predictor of Overdose among Patients with Substance Use Disorders. J Clin Med 2022; 11:6400. [PMID: 36362628 PMCID: PMC9657076 DOI: 10.3390/jcm11216400] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 10/22/2022] [Accepted: 10/23/2022] [Indexed: 06/04/2024] Open
Abstract
Increasing rates of overdose and overdose deaths are a significant public health problem. Research has examined co-occurring mental health conditions, including suicidality, as a risk factor for intentional and unintentional overdose among individuals with substance use disorder (SUD). However, this research has been limited to single site studies of self-reported outcomes. The current research evaluated suicidality as a predictor of overdose events in 2541 participants who use substances enrolled across eight multi-site clinical trials completed within the National Drug Abuse Treatment Clinical Trials Network between 2012 to 2021. The trials assessed baseline suicidality with the Concise Health Risk Tracking Self-Report (CHRT-SR). Overdose events were determined by reports of adverse events, cause of death, or hospitalization due to substance overdose, and verified through a rigorous adjudication process. Multivariate logistic regression was performed to assess continuous CHRT-SR score as a predictor of overdose, controlling for covariates. CHRT-SR score was associated with overdose events (p = 0.03) during the trial; the likelihood of overdose increased as continuous CHRT score increased (OR 1.02). Participants with lifetime heroin use were more likely to overdose (OR 3.08). Response to the marked rise in overdose deaths should integrate suicide risk reduction as part of prevention strategies.
Collapse
Affiliation(s)
- Viviana E. Horigian
- Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Miami, FL 33136, USA
| | - Renae D. Schmidt
- Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Miami, FL 33136, USA
| | | | - Kathryn Hefner
- The Emmes Company, LLC, 401 N. Washington St., Suite 700, Rockville, MD 20850, USA
| | - Judith Feinberg
- Departments of Behavioral Medicine and Psychiatry & Medicine/Infectious Diseases, West Virginia University School of Medicine, 930 Chestnut Ridge Road, Morgantown, WV 26505, USA
| | - Radhika Kondapaka
- The Emmes Company, LLC, 401 N. Washington St., Suite 700, Rockville, MD 20850, USA
| | - Daniel J. Feaster
- Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Miami, FL 33136, USA
| | - Rui Duan
- Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Miami, FL 33136, USA
| | - Sophia Gonzalez
- Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Miami, FL 33136, USA
| | - Carly Davis
- Department of Public Health Sciences, University of Miami Miller School of Medicine, 1120 Northwest 14th Street, Miami, FL 33136, USA
| | - Rodrigo Marín-Navarrete
- Division of Research and Translational Education, Centros de Integración Juvenil A.C., San Jerónimo Avenue 372, Jardines del Pedregal, Mexico City 01900, Mexico
| | - Susan Tross
- Department of Psychiatry, Columbia University, 1051 Riverside Drive, New York, NY 10032, USA
| |
Collapse
|
29
|
Mechanistic Effects and Use of N-acetylcysteine in Substance Use Disorders. Curr Behav Neurosci Rep 2022. [DOI: 10.1007/s40473-022-00250-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
30
|
Fredriksson I, Jayaram-Lindström N, Kalivas PW, Melas PA, Steensland P. N-acetylcysteine improves impulse control and attenuates relapse-like alcohol intake in long-term drinking rats. Behav Brain Res 2022; 436:114089. [PMID: 36063970 DOI: 10.1016/j.bbr.2022.114089] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/25/2022] [Accepted: 08/29/2022] [Indexed: 11/17/2022]
Abstract
Increasing evidence suggests that individuals with alcohol use disorder (AUD) present with a disrupted glutamatergic system that underlies core components of addictive disorders, including drug relapse and low impulse control. N-acetylcysteine (NAC) is a cystine prodrug that has been found to promote glutamate homeostasis and drug abstinence. However, no studies to date have evaluated NAC's effect on impulsivity in substance use disorders. Here we determined whether NAC would decrease alcohol-intake behaviors, in addition to improving impulse control, in long-term alcohol drinking male Wistar-Han rats. Before the start of the experiments, all rats were exposed to long-term intermittent access to 20% ethanol for at least seven weeks. Next, in different groups of rats, the effect of NAC (60 and/or 90mg/kg) was evaluated on (i) voluntary alcohol drinking using a two-bottle free choice paradigm, (ii) the motivation to self-administer alcohol under a progressive ratio schedule of reinforcement, and (iii) relapse-like drinking using the alcohol deprivation effect model. Finally, (iv) NAC's effect on impulse control was evaluated using the five-choice serial reaction time task. Results showed that NAC administration at 90mg/kg significantly reduced relapse-like drinking and improved impulse control. In contrast, NAC had no effect on levels of alcohol drinking or motivation to drink alcohol. In conclusion, our findings continue to support the use of NAC as an adjuvant treatment for the maintenance of abstinence in AUD. Moreover, we provide evidence for NAC's efficacy in improving impulse control following drinking, which warrants further investigation in substance use settings.
Collapse
Affiliation(s)
- Ida Fredriksson
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, 11364 Stockholm, Sweden
| | - Nitya Jayaram-Lindström
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, 11364 Stockholm, Sweden
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Ralph Johnson Veterans Administration, Charleston, SC, USA
| | - Philippe A Melas
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, 11364 Stockholm, Sweden; Center for Molecular Medicine, L8:00, Karolinska University Hospital, 17176 Stockholm, Sweden.
| | - Pia Steensland
- Center for Psychiatry Research, Department of Clinical Neuroscience, Karolinska Institutet & Stockholm Health Care Services, 11364 Stockholm, Sweden
| |
Collapse
|
31
|
Dennen CA, Blum K, Bowirrat A, Khalsa J, Thanos PK, Baron D, Badgaiyan RD, Gupta A, Braverman ER, Gold MS. Neurogenetic and Epigenetic Aspects of Cannabinoids. EPIGENOMES 2022; 6:27. [PMID: 36135314 PMCID: PMC9498086 DOI: 10.3390/epigenomes6030027] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/17/2022] [Accepted: 08/23/2022] [Indexed: 12/04/2022] Open
Abstract
Cannabis is one of the most commonly used and abused illicit drugs in the world today. The United States (US) currently has the highest annual prevalence rate of cannabis consumption in the world, 17.9% in individuals aged 12 or older, and it is on the rise. With increasing cannabis use comes the potential for an increase in abuse, and according to the Substance Abuse and Mental Health Services Administration (SAMHSA), approximately 5.1% of Americans had Cannabis Use Disorder (CUD) in 2020. Research has shown that genetics and epigenetics play a significant role in cannabis use and CUD. In fact, approximately 50-70% of liability to CUD and 40-48% of cannabis use initiation have been found to be the result of genetic factors. Cannabis usage and CUD have also been linked to an increased risk of psychiatric disorders and Reward Deficiency Syndrome (RDS) subsets like schizophrenia, depression, anxiety, and substance use disorder. Comprehension of the genetic and epigenetic aspects of cannabinoids is necessary for future research, treatment plans, and the production of pure cannabinoid compounds, which will be essential for FDA approval. In conclusion, having a better understanding of the epigenetic and genetic underpinnings of cannabis use, CUD, and the endocannabinoid system as a whole will aid in the development of effective FDA-approved treatment therapies and the advancement of personalized medicine.
Collapse
Affiliation(s)
- Catherine A. Dennen
- Department of Family Medicine, Jefferson Health Northeast, Philadelphia, PA 19114, USA
| | - Kenneth Blum
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, Austin, TX 78701, USA
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of the Provost), Graduate College, Western University of Health Sciences, Pomona, CA 91766, USA
- Institute of Psychology, ELTE Eötvös Loránd University, 1075 Budapest, Hungary
- Department of Psychiatry, University of Vermont, Burlington, VT 05405, USA
- Department of Psychiatry, Wright University Boonshoft School of Medicine, Dayton, OH 45324, USA
| | - Abdalla Bowirrat
- Department of Molecular Biology, Adelson School of Medicine, Ariel University, Ariel 40700, Israel
| | - Jag Khalsa
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20037, USA
- Medical Consequences of Drug Abuse and Infections Branch, National Institute on Drug Abuse, National Institutes of Health, Bethesda, MD 20852, USA
| | - Panayotis K. Thanos
- Behavioral Neuropharmacology and Neuroimaging Laboratory on Addictions, Clinical Research Institute on Addictions, Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biosciences, State University of New York at Buffalo, Buffalo, NY 14260, USA
| | - David Baron
- Division of Addiction Research & Education, Center for Psychiatry, Medicine & Primary Care (Office of the Provost), Graduate College, Western University of Health Sciences, Pomona, CA 91766, USA
| | - Rajendra D. Badgaiyan
- Department of Psychiatry, Icahn School of Medicine Mt Sinai, New York, NY 10029, USA
- Department of Psychiatry, South Texas Veteran Health Care System, Audie L. Murphy Memorial VA Hospital, San Antonio, TX 78229, USA
- Long School of Medicine, University of Texas Medical Center, San Antonio, TX 78229, USA
| | - Ashim Gupta
- Future Biologics, Lawrenceville, GA 30043, USA
| | - Eric R. Braverman
- Division of Nutrigenomics, The Kenneth Blum Behavioral Neurogenetic Institute, Austin, TX 78701, USA
| | - Mark S. Gold
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
32
|
Tijani AO, Garg J, Frempong D, Verana G, Kaur J, Joga R, Sabanis CD, Kumar S, Kumar N, Puri A. Sustained drug delivery strategies for treatment of common substance use disorders: Promises and challenges. J Control Release 2022; 348:970-1003. [PMID: 35752256 DOI: 10.1016/j.jconrel.2022.06.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 06/16/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
Substance use disorders (SUDs) are a leading cause of death and other ill health effects in the United States and other countries in the world. Several approaches ranging from detoxification, behavioral therapy, and the use of antagonists or drugs with counter effects are currently being applied for its management. Amongst these, drug therapy is the mainstay for some drug abuse incidences, as is in place specifically for opioid abuse or alcohol dependence. The severity of the havocs observed with the SUDs has triggered constant interest in the discovery and development of novel medications as well as suitable or most appropriate methods for the delivery of these agents. The chronic need of such drugs in users warrants the need for their prolonged or sustained systemic availability. Further, the need to improve patient tolerance to medication, limit invasive drug use and overall treatment outcome are pertinent considerations for embracing sustained release designs for medications used in managing SUDs. This review aims to provide an overview on up-to-date advances made with regards to sustained delivery systems for the drugs for treatment of different types of SUDs such as opioid, alcohol, tobacco, cocaine, and cannabis use disorders. The clinical relevance, promises and the limitations of deployed sustained release approaches along with future opportunities are discussed.
Collapse
Affiliation(s)
- Akeemat O Tijani
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Jivesh Garg
- University Institute of Pharmaceutical Sciences (UIPS), Panjab University, Chandigarh 160014, India
| | - Dorcas Frempong
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Gabrielle Verana
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Jagroop Kaur
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Chetan D Sabanis
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Sandeep Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Neeraj Kumar
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad 500 037, Telangana, India.
| | - Ashana Puri
- Department of Pharmaceutical Sciences, Bill Gatton College of Pharmacy, East Tennessee State University, Johnson City, TN 37614, USA.
| |
Collapse
|
33
|
McKee SA, McRae-Clark AL. Consideration of sex and gender differences in addiction medication response. Biol Sex Differ 2022; 13:34. [PMID: 35761351 PMCID: PMC9235243 DOI: 10.1186/s13293-022-00441-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 06/08/2022] [Indexed: 12/22/2022] Open
Abstract
Substance use continues to contribute to significant morbidity and mortality in the United States, for both women and men, more so than another other preventable health condition. To reduce the public health burden attributable to substances, the National Institute on Drug Abuse and the National Institute on Alcohol Abuse and Alcoholism have identified that medication development for substance use disorder is a high priority research area. Furthermore, both Institutes have stated that research on sex and gender differences in substance use medication development is a critical area. The purpose of the current narrative review is to highlight how sex and gender have been considered (or not) in medication trials for substance use disorders to clarify and summarize what is known regarding sex and gender differences in efficacy and to provide direction to the field to advance medication development that is consistent with current NIH 'sex as a biological variable' (SABV) policy. To that end, we reviewed major classes of abused substances (nicotine, alcohol, cocaine, cannabis, opioids) demonstrating that, sex and gender have not been well-considered in addiction medication development research. However, when adequate data on sex and gender differences have been evaluated (i.e., in tobacco cessation), clinically significant differences in response have been identified between women and men. Across the other drugs of abuse reviewed, data also suggest sex and gender may be predictive of outcome for some agents, although the relatively low representation of women in clinical research samples limits making definitive conclusions. We recommend the incorporation of sex and gender into clinical care guidelines and improved access to publicly available sex-stratified data from medication development investigations.
Collapse
Affiliation(s)
- Sherry A. McKee
- Yale School of Medicine, 2 Church St South, Suite 109, New Haven, CT 06519 USA
| | | |
Collapse
|
34
|
Sharma R, Tikka SK, Bhute AR, Bastia BK. N-acetyl cysteine in the treatment of cannabis use disorder: A systematic review of clinical trials. Addict Behav 2022; 129:107283. [PMID: 35189496 DOI: 10.1016/j.addbeh.2022.107283] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 01/17/2022] [Accepted: 02/13/2022] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND AIM Cannabis is the most consumed illicit drug globally, with a high risk of developing cannabis use disorder (CUD). No approved pharmacological treatment exists for CUD, but N-Acetyl Cysteine (NAC) has shown promising results in different clinical studies. This study aims to conduct a systematic review of NAC clinical trials for the treatment of CUD. METHODS Systematic review of randomized controlled trials (RCTs) was conducted to determine the effect of NAC for the treatment of cannabis dependence/cannabis use disorder (CUD). Articles were electronically searched across different databases using PubMed, Google Scholar, EMBASE, Cochrane Library, Medline and PsycINFO from inception to June 2021. Several study characteristics, including study duration, sample size, study population and age group, intervention, adverse effects, and outcome measure were extracted. A PICO table was used for data extraction. RESULTS We included 08 RCTs in the qualitative analysis. The risk of bias (RoB) was assessed according to Cochrane RoB criteria, and a 5 point grading system according to the Oxford Centre for Evidence-Based Medicine was used to rate the methodological quality (level of evidence) of the included articles. Mild and well-tolerated adverse events were reported in the placebo and NAC group. CONCLUSIONS The studies collectively offer mixed results, although the strength of the evidence available on which to make a recommendation is strong. NAC has shown to be effective in promoting abstinence, medication adherence and reducing cannabis use and craving among cannabis dependent users. This review also suggests recommendations for future research.
Collapse
|
35
|
Burke CW, Wilens TE. Transitional-Age Youth With Substance Use Disorders. Psychiatr Ann 2022. [DOI: 10.3928/00485713-20220520-01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
36
|
Montebello M, Jefferies M, Mills L, Bruno R, Copeland J, McGregor I, Rivas C, Jackson MA, Silsbury C, Dunlop A, Lintzeris N. Mood, sleep and pain comorbidity outcomes in cannabis dependent patients: Findings from a nabiximols versus placebo randomised controlled trial. Drug Alcohol Depend 2022; 234:109388. [PMID: 35316689 DOI: 10.1016/j.drugalcdep.2022.109388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 02/03/2022] [Accepted: 02/28/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Mood, sleep and pain problems are common comorbidities among treatment-seeking cannabis-dependent patients. There is limited evidence suggesting treatment for cannabis dependence is associated with their improvement. This study explored the impact of cannabis dependence treatment on these comorbidities. METHODS This is a secondary analysis from a 12-week double-blind placebo-controlled trial testing the efficacy of a cannabis agonist (nabiximols) against placebo in reducing illicit cannabis use in 128 cannabis-dependent participants. Outcome measurements including DASS-21 (Depression, Anxiety, and Stress subscales); Insomnia Severity Index (ISI); and Brief Pain Inventory (BPI), were performed at weeks 0, 4, 8, 12 and 24. Each was analysed as continuous outcomes and as binary cases based on validated clinical cut-offs. RESULTS Among those whose DASS and ISI scores were in the moderate to severe range at baseline, after controlling for cannabis use, there was a gradual decrease in severity of symptoms over the course of the trial. BPI decreased significantly until week 12 and then rose again in the post-treatment period during weeks 12-24. Neither pharmacotherapy type (nabiximols vs placebo) nor number of counselling sessions contributed significant explanatory power to any of the models and were excluded from the final analyses for both continuous and categorical outcomes. CONCLUSIONS Participants in this trial who qualified as cases at baseline had elevated comorbidity symptoms. There was no evidence that nabiximols treatment is a barrier to achieving reductions in the comorbid symptoms examined. Cannabis dependence treatment reduced illicit cannabis use and improved comorbidity symptoms, even when complete abstinence was not achieved.
Collapse
Affiliation(s)
- Mark Montebello
- Drug and Alcohol Services, Northern Sydney Local Health District, Level 1, 2c Herbert Street, St Leonards, NSW 2065, Australia; Specialty of Addiction Medicine, Faculty of Medicine and Health, University of Sydney, City Road, Camperdown, NSW 2006, Australia; National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St, Randwick, NSW 2031, Australia; NSW Drug and Alcohol Clinical Research and Improvement Network (DACRIN), NSW, Australia.
| | - Meryem Jefferies
- Drug Health, Western Sydney Local Health District, 5 Fleet St, North Parramatta, NSW 2151, Australia.
| | - Llewellyn Mills
- Specialty of Addiction Medicine, Faculty of Medicine and Health, University of Sydney, City Road, Camperdown, NSW 2006, Australia; NSW Drug and Alcohol Clinical Research and Improvement Network (DACRIN), NSW, Australia; Drug and Alcohol Services, South Eastern Sydney Local Health District, The Langton Centre, 591 South Dowling St, Surry Hills, NSW 2010, Australia.
| | - Raimondo Bruno
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St, Randwick, NSW 2031, Australia; School of Psychological Sciences, University of Tasmania, Private Bag 30, Hobart, Tasmania 7001, Australia.
| | - Jan Copeland
- National Drug and Alcohol Research Centre, University of New South Wales, 22-32 King St, Randwick, NSW 2031, Australia; Sunshine Coast Mind and Neuroscience Thompson Institute, University of the Sunshine Coast, Locked Bag 4, Maroochydore BC, QLD 4558, Australia.
| | - Iain McGregor
- Lambert Initiative for Cannabinoid Therapeutics, University of Sydney, City Road, Camperdown, NSW 2006, Australia.
| | - Consuelo Rivas
- Drug and Alcohol Services, South Eastern Sydney Local Health District, The Langton Centre, 591 South Dowling St, Surry Hills, NSW 2010, Australia.
| | - Melissa A Jackson
- NSW Drug and Alcohol Clinical Research and Improvement Network (DACRIN), NSW, Australia; Drug and Alcohol Clinical Services, Hunter New England Local Health District, Level 3, 670 Hunter Street, Newcastle, NSW 2300, Australia; School of Medicine and Public Health, University of Newcastle, University Dr, Callaghan, NSW 2308, Australia.
| | - Catherine Silsbury
- Drug Health, Western Sydney Local Health District, 5 Fleet St, North Parramatta, NSW 2151, Australia.
| | - Adrian Dunlop
- NSW Drug and Alcohol Clinical Research and Improvement Network (DACRIN), NSW, Australia; Drug and Alcohol Clinical Services, Hunter New England Local Health District, Level 3, 670 Hunter Street, Newcastle, NSW 2300, Australia.
| | - Nicholas Lintzeris
- Specialty of Addiction Medicine, Faculty of Medicine and Health, University of Sydney, City Road, Camperdown, NSW 2006, Australia; NSW Drug and Alcohol Clinical Research and Improvement Network (DACRIN), NSW, Australia; Drug and Alcohol Services, South Eastern Sydney Local Health District, The Langton Centre, 591 South Dowling St, Surry Hills, NSW 2010, Australia.
| | | |
Collapse
|
37
|
Courtney D, Hammond CJ, Rizwan B, Giasson S, Gupta M. The Cannabis Ask: What's a Psychiatrist to Do? JOURNAL OF THE CANADIAN ACADEMY OF CHILD AND ADOLESCENT PSYCHIATRY = JOURNAL DE L'ACADEMIE CANADIENNE DE PSYCHIATRIE DE L'ENFANT ET DE L'ADOLESCENT 2022; 31:28-37. [PMID: 35251194 PMCID: PMC8862599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
|
38
|
Hodebourg R, Kalivas PW, Kruyer A. Extrasynaptic therapeutic targets in substance use and stress disorders. Trends Pharmacol Sci 2022; 43:56-68. [PMID: 34753604 PMCID: PMC8688303 DOI: 10.1016/j.tips.2021.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/05/2021] [Accepted: 10/08/2021] [Indexed: 01/03/2023]
Abstract
Treatments for substance use and stress disorders are based on ameliorating behavioral symptoms, not on reversing the synaptic pathology that has the potential to cure disorders. This failing arises in part from a research focus on how pre- and postsynaptic physiology is changed even though key neuropathology exists in the perisynaptic neuropil that homeostatically regulates synaptic transmission. We explore recent findings from the substance use and stress disorder literature pointing to a key role for perisynaptic astroglia and signaling in the extracellular matrix (ECM) in regulating synaptic pathology. We conclude that drugs and stress initiate long-lasting changes in brain synapses via enduring neuroadaptations in astroglia and the ECM, and that modulating extrasynaptic regulators may be therapeutically useful.
Collapse
Affiliation(s)
- Ritchy Hodebourg
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA
| | - Peter W Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA.
| | - Anna Kruyer
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC 29464, USA
| |
Collapse
|
39
|
Bradlow RCJ, Berk M, Kalivas PW, Back SE, Kanaan RA. The Potential of N-Acetyl-L-Cysteine (NAC) in the Treatment of Psychiatric Disorders. CNS Drugs 2022; 36:451-482. [PMID: 35316513 PMCID: PMC9095537 DOI: 10.1007/s40263-022-00907-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/17/2022] [Indexed: 12/22/2022]
Abstract
N-acetyl-L-cysteine (NAC) is a compound of increasing interest in the treatment of psychiatric disorders. Primarily through its antioxidant, anti-inflammatory, and glutamate modulation activity, NAC has been investigated in the treatment of neurodevelopmental disorders, schizophrenia spectrum disorders, bipolar-related disorders, depressive disorders, anxiety disorders, obsessive compulsive-related disorders, substance-use disorders, neurocognitive disorders, and chronic pain. Whilst there is ample preclinical evidence and theoretical justification for the use of NAC in the treatment of multiple psychiatric disorders, clinical trials in most disorders have yielded mixed results. However, most studies have been underpowered and perhaps too brief, with some evidence of benefit only after months of treatment with NAC. Currently NAC has the most evidence of having a beneficial effect as an adjuvant agent in the negative symptoms of schizophrenia, severe autism, depression, and obsessive compulsive and related disorders. Future research with well-powered studies that are of sufficient length will be critical to better understand the utility of NAC in the treatment of psychiatric disorders.
Collapse
Affiliation(s)
| | - Michael Berk
- IMPACT-The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Deakin University, Geelong, VIC Australia ,Orygen, The National Centre of Excellence in Youth Mental Health, Centre for Youth Mental Health, Melbourne, VIC Australia ,Florey Institute of Neuroscience and Mental Health, Melbourne, VIC Australia ,Department of Psychiatry, University of Melbourne, Parkville, VIC Australia
| | - Peter W. Kalivas
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC USA ,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC USA
| | - Sudie E. Back
- Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC USA ,Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC USA
| | - Richard A. Kanaan
- Florey Institute of Neuroscience and Mental Health, Melbourne, VIC Australia ,Department of Psychiatry, University of Melbourne, Parkville, VIC Australia
| |
Collapse
|
40
|
Rømer Thomsen K, Thylstrup B, Kenyon EA, Lees R, Baandrup L, Feldstein Ewing SW, Freeman TP. Cannabinoids for the treatment of cannabis use disorder: New avenues for reaching and helping youth? Neurosci Biobehav Rev 2022; 132:169-180. [PMID: 34822876 PMCID: PMC11577263 DOI: 10.1016/j.neubiorev.2021.11.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/17/2021] [Accepted: 11/21/2021] [Indexed: 11/16/2022]
Abstract
Cannabis use peaks during adolescence and emerging adulthood, and cannabis use disorder (CUD) is associated with a wide range of adverse outcomes. This is particularly pertinent in youth, because the developing brain may be more vulnerable to adverse effects of frequent cannabis use. Combining evidence-based psychosocial interventions with safe and effective pharmacotherapy is a potential avenue to improve youth outcomes, but we lack approved CUD pharmacotherapies. Here, we review new potential avenues for helping youth with CUD, with a particular focus on cannabinoid-based treatments. Evidence from placebo-controlled RCTs suggests synthetic delta-9-tetrahydrocannabinol (THC) decreases withdrawal symptoms, but not cannabis use, in adults with daily cannabis use/CUD, while findings regarding formulations containing THC combined with cannabidiol (CBD) are mixed. Preliminary evidence from two placebo-controlled RCTs in adults with CUD suggests that both Fatty Acid Amide Hydrolase inhibitors and CBD can reduce cannabis use. However, larger trials are needed to strengthen the evidence. Findings from adults point to cannabinoid-based treatments as a potential strategy that should be examined in youth with CUD.
Collapse
Affiliation(s)
- Kristine Rømer Thomsen
- Centre for Alcohol and Drug Research, Department of Psychology and Behavioral Sciences, Aarhus University, Denmark.
| | - Birgitte Thylstrup
- Centre for Alcohol and Drug Research, Department of Psychology and Behavioral Sciences, Aarhus University, Denmark
| | - Emily A Kenyon
- Department of Psychology, University of Rhode Island, USA
| | - Rachel Lees
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, UK
| | - Lone Baandrup
- Mental Health Centre Copenhagen and Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Sarah W Feldstein Ewing
- Centre for Alcohol and Drug Research, Department of Psychology and Behavioral Sciences, Aarhus University, Denmark; Department of Psychology, University of Rhode Island, USA
| | - Tom P Freeman
- Addiction and Mental Health Group (AIM), Department of Psychology, University of Bath, UK
| |
Collapse
|
41
|
Arancini L, Mohebbi M, Berk M, Dean OM, Bortolasci CC, Spolding B, Zazula R, Dodd S. A placebo-controlled, randomised pilot trial of N-acetylcysteine or placebo for cessation of tobacco smoking. Eur Neuropsychopharmacol 2021; 53:120-126. [PMID: 34757312 DOI: 10.1016/j.euroneuro.2021.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 11/18/2022]
Abstract
Smoking represents a significant health threat to the population, however there remains a core group of consistent smokers that are largely unable to break the addiction. Novel therapies are required to assist this group with cessation. N-acetylcysteine (NAC) is a nutraceutical supplement that has shown efficacy compared to placebo in previous pilot studies for assisting smokers to quit or reduce their consumption of cigarettes. A double-blind, randomised trial with a treatment period of 16 weeks and a final follow-up at 42 weeks was conducted comparing 1.8g of effervescent NAC per day (n=47) with placebo (n=47) as an aide to smoking cessation. Both study arms received adjunctive online support through the QuitCoach program. Participants reported smoking at each timepoint (baseline and weeks 8, 16 & 42), which was confirmed through salivary cotinine and exhaled carbon monoxide testing. Primary and secondary analyses were undertaken using a modified intent-to-treat basis, including all participants with at least one valid post baseline outcome, regardless of treatment received or their withdrawal from the study. There was no significant difference in smoking outcomes between intervention groups among the 24 participants that competed follow-up. There were no significant differences in age, gender, or body mass index (BMI) between the groups lost to follow-up or recorded at follow-up. This study found no evidence to support NAC as a therapy for smoking cessation. The negative outcome could be the result of lack of treatment efficacy, or alternatively, small sample size, participant retention difficulties, dose, or duration of follow-up. Trial Registration: Australian New Zealand Clinical Trials registry (ANZCTR), ACTRN12617001478303. Registered on 19 October 2017.
Collapse
Affiliation(s)
- Lauren Arancini
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia.
| | - Mohammadreza Mohebbi
- Deakin University, Faculty of Health, Biostatistics Unit, Geelong, VIC, Australia.
| | - Michael Berk
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia; University Hospital Geelong, Barwon Health, Geelong, VIC, Australia; Department of Psychiatry, the University of Melbourne, Parkville, VIC, Australia; Orygen, the National Centre for Excellence in Youth Mental Health, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
| | - Olivia M Dean
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia; University Hospital Geelong, Barwon Health, Geelong, VIC, Australia; Department of Psychiatry, the University of Melbourne, Parkville, VIC, Australia; Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia.
| | - Chiara C Bortolasci
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia.
| | - Briana Spolding
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia
| | - Robson Zazula
- Federal University for Latin American Integration, Foz do Iguacu, Brazil
| | - Seetal Dodd
- Deakin University, IMPACT, the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, VIC, Australia; University Hospital Geelong, Barwon Health, Geelong, VIC, Australia; Department of Psychiatry, the University of Melbourne, Parkville, VIC, Australia; Orygen, the National Centre for Excellence in Youth Mental Health, Parkville, VIC, Australia.
| |
Collapse
|
42
|
McRae-Clark AL, Gray KM, Baker NL, Sherman BJ, Squeglia L, Sahlem GL, Wagner A, Tomko R. Varenicline as a treatment for cannabis use disorder: A placebo-controlled pilot trial. Drug Alcohol Depend 2021; 229:109111. [PMID: 34655945 PMCID: PMC8665036 DOI: 10.1016/j.drugalcdep.2021.109111] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 09/16/2021] [Accepted: 09/23/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND An efficacious pharmacotherapy for cannabis use disorder (CUD) has yet to be established. This study preliminarily evaluated the safety and efficacy of varenicline for CUD in a proof-of-concept clinical trial. METHODS Participants in this 6-week randomized, placebo-controlled pilot trial received either varenicline (n = 35) or placebo (n = 37), added to a brief motivational enhancement therapy intervention. Outcomes included cannabis withdrawal, cannabis abstinence, urine cannabinoid levels, percent cannabis use days, and cannabis sessions per day. RESULTS Both treatment groups noted significant decreases in self-reported cannabis withdrawal, percentage of days used, and use sessions per day during treatment compared to baseline. While this pilot trial was not powered to detect statistically significant between-group differences, participants randomized to varenicline evidenced numerically greater rates of self-reported abstinence at the final study visit [Week 6 intent-to-treat (ITT): Varenicline: 17.1% vs. Placebo: 5.4%; RR = 3.2 (95% CI: 0.7,14.7)]. End-of-treatment urine creatinine corrected cannabinoid levels were numerically lower in the varenicline group and higher in the placebo group compared to baseline [Change from baseline: Varenicline -1.7 ng/mg (95% CI: -4.1,0.8) vs. Placebo: 1.9 ng/mg (95% CI: -0.4,4.3); Δ = 3.5 (95% CI: 0.1,6.9)]. Adverse events related to study treatment did not reveal new safety signals. CONCLUSIONS Findings support the feasibility of conducting clinical trials of varenicline as a candidate pharmacotherapy for CUD, and indicate that a full-scale efficacy trial, powered based on effect sizes and variability yielded in this study, is warranted.
Collapse
Affiliation(s)
- Aimee L. McRae-Clark
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC,Ralph H. Johnson VA Medical Center, Charleston, SC
| | - Kevin M. Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | - Nathaniel L. Baker
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC
| | - Brian J. Sherman
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | - Lindsay Squeglia
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | | | - Amanda Wagner
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| | - Rachel Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC
| |
Collapse
|
43
|
Bahji A, Meyyappan AC, Hawken ER, Tibbo PG. Pharmacotherapies for cannabis use disorder: A systematic review and network meta-analysis. THE INTERNATIONAL JOURNAL OF DRUG POLICY 2021; 97:103295. [PMID: 34062288 PMCID: PMC8881089 DOI: 10.1016/j.drugpo.2021.103295] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 04/28/2021] [Accepted: 04/30/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE This study aimed to determine the efficacy and acceptability of pharmacotherapies for cannabis use disorder (CUD). METHODS We conducted a systematic review and frequentist network meta-analysis, searching five electronic databases for randomized placebo-controlled trials of individuals diagnosed with CUD receiving pharmacotherapy with or without concomitant psychotherapy. Primary outcomes were the reduction in cannabis use and retention in treatment. Secondary outcomes were adverse events, discontinuation due to adverse events, total abstinence, withdrawal symptoms, cravings, and CUD severity. We applied a frequentist, random-effects Network Meta-Analysis model to pool effect sizes across trials using standardized mean differences (SMD, g) and rate ratios (RR) with their 95% confidence intervals. RESULTS We identified a total of 24 trials (n=1912, 74.9% male, mean age 30.2 years). Nabilone (d=-4.47 [-8.15; -0.79]), topiramate (d=-3.80 [-7.06; -0.54]), and fatty-acid amyl hydroxylase inhibitors (d=-2.30 [-4.75; 0.15]) reduced cannabis use relative to placebo. Dronabinol improved retention in treatment (RR=1.27 [1.02; 1.57]), while topiramate worsened treatment retention (RR=0.62 [0.42; 0.91]). Gabapentin reduced cannabis cravings (d=-2.42 [-3.53; -1.32], while vilazodone worsened craving severity (d=1.69 [0.71; 2.66]. Buspirone (RR=1.14 [1.00; 1.29]), venlafaxine (RR=1.78 [1.40; 2.26]), and topiramate (RR=9.10 [1.27; 65.11]) caused more adverse events, while topiramate caused more dropouts due to adverse events. CONCLUSIONS Based on this review, some medications appeared to show promise for treating individual aspects of CUD. However, there is a lack of robust evidence to support any particular pharmacological treatment. There is a need for additional studies to expand the evidence base for CUD pharmacotherapy. While medication strategies may become an integral component for CUD treatment one day, psychosocial interventions should remain the first line given the limitations in the available evidence.
Collapse
Affiliation(s)
- Anees Bahji
- Department of Psychiatry, University of Calgary, Calgary, AB, Canada.
| | - Arthi Chinna Meyyappan
- Department of Psychiatry, Queen's University, Kingston, ON, Canada; Centre for Neurosciences, Queen's University, Kingston, ON, Canada; Providence Care Hospital, Kingston, ON, Canada
| | - Emily R Hawken
- Department of Psychiatry, Queen's University, Kingston, ON, Canada; Centre for Neurosciences, Queen's University, Kingston, ON, Canada; Providence Care Hospital, Kingston, ON, Canada
| | - Philip G Tibbo
- Department of Psychiatry, Dalhousie University, Halifax, NS, Canada; Nova Scotia Early Psychosis Program, Halifax, NS, Canada
| |
Collapse
|
44
|
Roydhouse JK, Floden L, Tomko RL, Gray KM, Bell ML. The estimand framework and its application in substance use disorder clinical trials: a case study. THE AMERICAN JOURNAL OF DRUG AND ALCOHOL ABUSE 2021; 47:658-663. [PMID: 34702088 PMCID: PMC10124131 DOI: 10.1080/00952990.2021.1976199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Relapse rates among individuals with substance use disorder (SUD) remain high and new treatment approaches are needed, which require evaluation in randomized controlled trials (RCTs). Measurement and interpretation challenges for SUD RCT data are often ignored or presented only in statistical analysis plans. Since different analytic approaches may result in different estimates and thus interpretations of the treatment effect, it is important to present this clearly throughout the trial. Inconsistencies between study analyses and objectives present further challenges for interpretation and cross-study comparisons. The recent International Council for Harmonization (ICH) addendum provides standardized language and a common framework for aligning trial objectives, design, conduct, and analysis. The framework focuses on estimands, which describe the treatment effect and link the trial objective with the scientific question and the analytic approach. The use of estimands offers SUD researchers and clinicians the opportunity to explicitly address events that affect measurement and interpretation at the outset of the trial. Furthermore, the use of standard terminology can lead to clearer interpretations of SUD trials and the treatments evaluated in SUD trials. Resources for understanding and applying estimands are needed to optimize the use of this new, helpful framework. This Perspective provides this resource for SUD researchers. Specifically, it highlights the relevance of estimands for SUD trials. Furthermore, it demonstrates how estimands can be used to develop clinically relevant analyses to address challenges in SUD trials. It also shows how a standardized framework can be employed to improve the interpretation and presentation of SUD study findings.
Collapse
Affiliation(s)
- Jessica K Roydhouse
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia.,Department of Health Services, Policy and Practice, Brown University School of Public Health, Providence, RI, USA
| | | | - Rachel L Tomko
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Kevin M Gray
- Department of Psychiatry and Behavioral Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Melanie L Bell
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, AZ, USA.,Psycho-Oncology Co-operative Research Group, School of Psychology, University of Sydney, Camperdown, Australia
| |
Collapse
|
45
|
McKetin R, Dean OM, Turner A, Kelly PJ, Quinn B, Lubman DI, Dietze P, Carter G, Higgs P, Sinclair B, Reid D, Baker AL, Manning V, Pas NT, Thomas T, Bathish R, Raftery DK, Wrobel A, Saunders L, Arunogiri S, Cordaro F, Hill H, Hall S, Clare PJ, Mohebbi M, Berk M. N-acetylcysteine (NAC) for methamphetamine dependence: A randomised controlled trial. EClinicalMedicine 2021; 38:101005. [PMID: 34308314 PMCID: PMC8283342 DOI: 10.1016/j.eclinm.2021.101005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Methamphetamine dependence is a significant global health concern for which there are no approved medications. The cysteine prodrug, N-acetylcysteine (NAC), has been found to ameliorate glutamate dysregulation in addiction, and to reduce craving for methamphetamine and other drugs. We evaluated the efficacy and safety of NAC as a pharmacotherapy for methamphetamine dependence. METHODS A parallel double-blind randomised placebo-controlled trial of people dependent on methamphetamine recruited from Geelong, Melbourne and Wollongong, Australia, between July 2018 and December 2019. Participants were randomised to receive either 12 weeks of oral NAC (2400 mg/day) or matched placebo, delivered as a take-home medication. The primary outcome was methamphetamine use, measured in two ways: (a) change in days of use in the past 4 weeks from baseline to weeks 4, 8 and 12, assessed using the Timeline Followback; and (b) methamphetamine-positive oral fluid samples taken weekly. Analyses were intention-to-treat and based on imputed data. Secondary outcomes were craving, severity of dependence, withdrawal severity and psychiatric symptoms (depression, suicidality, hostility and psychotic symptoms). Significance levels were p < 0.025 for primary outcomes and p < 0.01 for secondary outcomes. Adverse events were compared between groups by system organ class. The study was prospectively registered, ACTRN12618000366257. RESULTS Participants (N = 153; 59% male, mean [SD] age 38 [8]) were randomised to placebo (n = 77) or NAC (n = 76). Both groups had a median (IQR) of 24 (15-28) days of methamphetamine use in the 4 weeks prior to baseline. Both groups significantly reduced methamphetamine use (mean [SE] reduction of 7.3 [1.2]) days for placebo, 6.8 [1.2] for NAC) but NAC did not reduce days of methamphetamine use more than placebo (group difference of 0.5 days, 97.5% CI -3.4-4.3). There was no significant effect of NAC on methamphetamine-positive oral fluid samples (placebo 79%, NAC 76%; mean difference -2.6, 97.5% CI -12.6-7.4). NAC did not significantly reduce craving, severity of dependence, withdrawal, suicidality, depression, hostility or psychotic symptoms relative to placebo. Adverse events did not differ significantly between placebo and NAC groups. INTERPRETATION These findings suggest that take-home oral NAC has no significant effect on methamphetamine use or most clinically related outcomes amongst people who are dependent on the drug.
Collapse
Affiliation(s)
- Rebecca McKetin
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Olivia M. Dean
- Deakin University, School of Medicine, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
| | - Alyna Turner
- Deakin University, School of Medicine, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Peter J. Kelly
- Illawarra Health and Medical Research Institute and School of Psychology, University of Wollongong, Wollongong, Australia
| | - Brendan Quinn
- Behaviours and Health Risks Program, Burnet Institute, Melbourne, Australia
| | - Dan I. Lubman
- Monash Addiction Research Centre, Eastern Health Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
- Turning Point, Eastern Health, Richmond, Australia
| | - Paul Dietze
- Behaviours and Health Risks Program, Burnet Institute, Melbourne, Australia
- National Drug Research Institute, Curtin University, Australia
| | - Gregory Carter
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Peter Higgs
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Barbara Sinclair
- Drug and Alcohol Services, Illawarra Shoalhaven Local Health District, Wollongong, Australia
| | - David Reid
- Drug and Alcohol Services, Illawarra Shoalhaven Local Health District, Wollongong, Australia
| | - Amanda L. Baker
- School of Medicine and Public Health, University of Newcastle, Callaghan, Australia
| | - Victoria Manning
- Monash Addiction Research Centre, Eastern Health Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
- Turning Point, Eastern Health, Richmond, Australia
| | - Nina te Pas
- National Drug Research Institute, Curtin University, Australia
| | - Tamsin Thomas
- Illawarra Health and Medical Research Institute and School of Psychology, University of Wollongong, Wollongong, Australia
| | - Ramez Bathish
- Monash Addiction Research Centre, Eastern Health Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
- Turning Point, Eastern Health, Richmond, Australia
| | - Dayle K. Raftery
- Illawarra Health and Medical Research Institute and School of Psychology, University of Wollongong, Wollongong, Australia
| | - Anna Wrobel
- Deakin University, School of Medicine, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Lucy Saunders
- Deakin University, School of Medicine, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
| | - Shalini Arunogiri
- Monash Addiction Research Centre, Eastern Health Clinical School, Faculty of Medicine, Nursing & Health Sciences, Monash University, Melbourne, Australia
- Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Australia
| | - Frank Cordaro
- Drug and Alcohol Services, Illawarra Shoalhaven Local Health District, Wollongong, Australia
| | - Harry Hill
- Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Australia
| | - Scott Hall
- Monash Alfred Psychiatry Research Centre, Monash University, Melbourne, Australia
| | - Philip J. Clare
- National Drug and Alcohol Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
- Prevention Research Collaboration, School of Public Health, University of Sydney, Australia
| | - Mohammadreza Mohebbi
- Deakin University, School of Medicine, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
- Faculty of Health, Biostatistics Unit, Deakin University, Geelong, Australia
| | - Michael Berk
- Deakin University, School of Medicine, IMPACT – the Institute for Mental and Physical Health and Clinical Translation, Geelong, Australia
- Florey Institute for Neuroscience and Mental Health, University of Melbourne, Melbourne, Australia
- Department of Psychiatry, University of Melbourne, Parkville, Australia
- Health Drug and Alcohol Services, Geelong, Australia
- Orygen, The National Centre of Excellence in Youth Mental Health, University of Melbourne, Parkville, Australia
| |
Collapse
|
46
|
McClure EA, Wahlquist AE, Tomko RL, Baker NL, Carpenter MJ, Bradley ED, Cato PA, Gipson CD, Gray KM. Evaluating N-acetylcysteine for early and end-of-treatment abstinence in adult cigarette smokers. Drug Alcohol Depend 2021; 225:108815. [PMID: 34171822 PMCID: PMC8282766 DOI: 10.1016/j.drugalcdep.2021.108815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 04/02/2021] [Accepted: 04/10/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND There is robust preclinical literature and preliminary clinical findings supporting the use of N-Acetylcysteine (NAC) to treat substance use disorders, including tobacco use disorder (TUD). However, randomized controlled trials have yielded mixed results and NAC's efficacy for TUD has not been established. The goals of this study were to assess the efficacy of NAC in promoting early and end-of-treatment abstinence and preventing relapse among adult smokers. METHODS This randomized, double-blinded clinical trial enrolled adult, daily smokers (N = 114; ages 23-64; 51 % female; 65 % White; 29 % Black/African American; 7% Hispanic/Latinx), who were randomized 1:1 to receive NAC (n = 59) or placebo (n = 55) (1200 mg b.i.d.) for eight weeks. Participants received brief cessation counseling and incentives for abstinence during the first three days of the quit attempt. Primary outcomes: (i) carbon monoxide (CO)-confirmed abstinence during the first three days of the quit attempt. SECONDARY OUTCOMES (ii) time to relapse; (iii) biologically confirmed abstinence at Week 8. RESULTS No differences were found between NAC and placebo groups on measures of early abstinence (3-day quit attempt; 11 % for NAC vs. 15 % for placebo; all p > 0.11), time to relapse (p = 0.19), and end-of-treatment abstinence (7% for NAC vs. 11 % for placebo; all p > 0.40]. CONCLUSIONS Results indicate that NAC is a well-tolerated pharmacotherapy but is unlikely to be efficacious as a monotherapy for TUD in adults. Considered in the collective context of other research, NAC may potentially be more useful in a younger population, as a combination pharmacotherapy, or in the presence of more intensive psychosocial treatment.
Collapse
Affiliation(s)
- Erin A. McClure
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, 67 President St., MSC 861, Charleston SC, 29425, USA,Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St., Charleston SC, 29425, USA,Corresponding Author: Erin A. McClure, Ph.D., Medical University of South Carolina, 67 President St, MSC 861, Charleston, SC 29425, Phone: 843-792-7192,
| | - Amy E. Wahlquist
- East Tennessee State University, Center for Rural Health Research, Department of Biostatistics and Epidemiology, 104 Lamb Hall, Johnson City, TN, 37612, USA
| | - Rachel L. Tomko
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, 67 President St., MSC 861, Charleston SC, 29425, USA
| | - Nathaniel L. Baker
- Medical University of South Carolina, Department of Public Health Sciences, 135 Cannon St., Charleston SC, 29425, USA
| | - Matthew J. Carpenter
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, 67 President St., MSC 861, Charleston SC, 29425, USA,Hollings Cancer Center, Medical University of South Carolina, 86 Jonathan Lucas St., Charleston SC, 29425, USA
| | - Elizabeth D. Bradley
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, 67 President St., MSC 861, Charleston SC, 29425, USA
| | - Patrick A. Cato
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, 67 President St., MSC 861, Charleston SC, 29425, USA
| | - Cassandra D. Gipson
- Department of Family and Community Medicine, University of Kentucky, 2195 Harrodsburg Rd., Lexington KY, 40504, USA
| | - Kevin M. Gray
- Medical University of South Carolina, Department of Psychiatry and Behavioral Sciences, 67 President St., MSC 861, Charleston SC, 29425, USA
| |
Collapse
|
47
|
Levin FR, Mariani JJ, Choi CJ, Basaraba C, Brooks DJ, Brezing CA, Pavlicova M. Non-abstinent treatment outcomes for cannabis use disorder. Drug Alcohol Depend 2021; 225:108765. [PMID: 34087745 PMCID: PMC8282674 DOI: 10.1016/j.drugalcdep.2021.108765] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 04/09/2021] [Accepted: 04/13/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Abstinence remains a standard outcome for potential treatment interventions for Cannabis Use Disorder (CUD). However, there needs to be validation of non-abstinent outcomes. This study explores reductions in self-reported days of use as another viable outcome measure using data from three completed randomized placebo-controlled clinical trials of pharmacological interventions for CUD. METHODS The three trials tested the effect of quetiapine (QTP, n = 113); dronabinol (DRO, n = 156); and lofexidine + dronabinol (LFD, n = 122). Self-reported cannabis use was categorized into three use-groups/week: heavy (5-7 days/week), moderate (2-4 days/week) and light use (0-1 days/week). Multinomial logistic regressions analyzed the treatment by time effect on the likelihood of light and moderate use compared to heavy use in each study. RESULTS Across the three trials, there was no significant overall time-by-treatment interaction (QTP: p = .06; DRO: p = .15; LFD: p = .21). However, the odds of moderate compared to heavy use were significantly higher in treatment than in placebo groups starting around the midpoint of each trial. No treatment differences were found between the odds of light compared to heavy use. CONCLUSIONS While study-end abstinence rates have been a standard treatment outcome for CUD trials, reduction from heavy to moderate use has not been standardly assessed. During the last several weeks of each trial, those on active medication were more likely to move from heavy to moderate use, which suggests that certain medications may be more impactful than previously assessed. Future studies should determine if this pattern is associated with less CUD severity and/or improved quality of life.
Collapse
Affiliation(s)
- Frances R. Levin
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA,Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032 USA,Address correspondence to: Frances R. Levin, M.D., 1051 Riverside Drive, Unit 66, New York, New York 10032 USA, Telephone: (646) 774-6137 FAX: (212) 543-6018,
| | - John J. Mariani
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA,Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032 USA
| | - C. Jean Choi
- New York State Psychiatric Institute, Division of Mental Health Data Science, 1051 Riverside Drive, New York, NY 10032 USA
| | - Cale Basaraba
- New York State Psychiatric Institute, Division of Mental Health Data Science, 1051 Riverside Drive, New York, NY 10032 USA
| | - Daniel J. Brooks
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA
| | - Christina A. Brezing
- New York State Psychiatric Institute, Division on Substance Use Disorders, 1051 Riverside Drive, New York, NY 10032 USA,Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032 USA
| | - Martina Pavlicova
- Department of Biostatistics, Columbia University, 722 West 168th Street, New York, NY 10032 USA
| |
Collapse
|
48
|
Ullrich HS, Torbati A, Fan W, Arbona C, Cano MA, Essa S, Harvey L, Vaughan EL, de Dios MA. Race, psychosocial characteristics, and treatment outcomes among individuals undergoing treatment for cannabis use disorder: A latent profile analysis based on preferred method of using cannabis. J Subst Abuse Treat 2021; 131:108561. [PMID: 34275690 DOI: 10.1016/j.jsat.2021.108561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/29/2021] [Accepted: 07/04/2021] [Indexed: 11/30/2022]
Abstract
INTRODUCTION There are a wide variety of methods for using combustible cannabis which may impact an individual's pattern of use as well as their response to cannabis use disorder (CUD) treatment. Previous research has noted racial/ethnic differences in cannabis users' preferred method of use. METHOD The current study examined data from a randomized placebo-controlled trial of a pharmacological intervention for adults with CUD. Latent profile analysis classified participants (N = 302) based on their primary method of combustible cannabis use. RESULTS A four profile solution emerged which identified participants who demonstrated 1) Primarily Joint (n = 50), 2) Primarily Blunt (n = 106), 3) Mixed MoU (n = 30), and 4) Primarily Pipe (i.e., pipe or bong; n = 116) use. Profiles were compared on socio-demographic characteristics and racial differences were found among the four latent profiles as well as differences in their level of use. Cannabis users with a preference for joints were more likely to be White as compared to other racial groups. In contrast, a greater proportion of participants with a preference for blunts were African American. The Primarily Joint profile was found to have the highest cannabis relapse rate at 1-month follow-up (94%) which was significantly greater than the Mixed MoU (74%, x2 = 5.06, p < .05) and Primarily Pipe (78%, x2 = 9.24, p < .01) profiles. Interestingly, there was no difference in 1-Month Follow-up cannabis relapse rates between the Primarily Joint and Blunt profiles (87%, x2 = 9.24, p > .05). CONCLUSIONS Findings suggest that treatment-seeking individuals who primarily use joints or blunts may face unique challenges that may impact cannabis abstinence. Along with other cannabis-related characteristics, an individual's preferred method of use may represent an important factor to consider in the treatment of CUD.
Collapse
Affiliation(s)
- Helen S Ullrich
- Department of Psychological, Health, & Learning Sciences, University of Houston, 3657 Cullen Blvd, Houston, TX 77204, United States.
| | - Autena Torbati
- Department of Psychological, Health, & Learning Sciences, University of Houston, 3657 Cullen Blvd, Houston, TX 77204, United States.
| | - Weihua Fan
- Department of Psychological, Health, & Learning Sciences, University of Houston, 3657 Cullen Blvd, Houston, TX 77204, United States.
| | - Consuelo Arbona
- Department of Psychological, Health, & Learning Sciences, University of Houston, 3657 Cullen Blvd, Houston, TX 77204, United States.
| | - Miguel A Cano
- Department of Epidemiology, Florida International University, 11200 SW 8(th) St AHC5, Miami, FL 33199, United States.
| | - Saman Essa
- Department of Psychological, Health, & Learning Sciences, University of Houston, 3657 Cullen Blvd, Houston, TX 77204, United States.
| | - Laura Harvey
- Department of Psychological, Health, & Learning Sciences, University of Houston, 3657 Cullen Blvd, Houston, TX 77204, United States.
| | - Ellen L Vaughan
- Department of Counseling and Educational Psychology, Indiana University, 201 N Rose Ave, Bloomington, IN 47405, United States.
| | - Marcel A de Dios
- Department of Psychological, Health, & Learning Sciences, University of Houston, 3657 Cullen Blvd, Houston, TX 77204, United States.
| |
Collapse
|
49
|
N-acetylcysteine in substance use disorder: a lesson from preclinical and clinical research. Pharmacol Rep 2021; 73:1205-1219. [PMID: 34091880 PMCID: PMC8460563 DOI: 10.1007/s43440-021-00283-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 05/19/2021] [Accepted: 05/19/2021] [Indexed: 12/29/2022]
Abstract
Substance use disorder (SUD) is a chronic brain condition, with compulsive and uncontrollable drug-seeking that leads to long-lasting and harmful consequences. The factors contributing to the development of SUD, as well as its treatment settings, are not fully understood. Alterations in brain glutamate homeostasis in humans and animals implicate a key role of this neurotransmitter in SUD, while the modulation of glutamate transporters has been pointed as a new strategy to diminish the excitatory glutamatergic transmission observed after drugs of abuse. N-acetylcysteine (NAC), known as a safe mucolytic agent, is involved in the regulation of this system and may be taken into account as a novel pharmacotherapy for SUD. In this paper, we summarize the current knowledge on the ability of NAC to reduce drug-seeking behavior induced by psychostimulants, opioids, cannabinoids, nicotine, and alcohol in animals and humans. Preclinical studies showed a beneficial effect in animal models of SUD, while the clinical efficacy of NAC has not been fully established. In summary, NAC will be a small add-on to usual treatment and/or psychotherapy for SUD, however, further studies are required.
Collapse
|
50
|
Chang CT, Hsieh PJ, Lee HC, Lo CH, Tam KW, Loh EW. Effectiveness of N-acetylcysteine in Treating Clinical Symptoms of Substance Abuse and Dependence: A Meta-analysis of Randomized Controlled Trials. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE 2021; 19:282-293. [PMID: 33888657 PMCID: PMC8077050 DOI: 10.9758/cpn.2021.19.2.282] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/15/2022]
Abstract
Objective Treatment with N-acetylcysteine (NAC) is believed to reduce the clinical symptoms among individuals with substance abuse or dependence. We conducted a meta-analysis of randomized controlled trials to evaluate the effectiveness of NAC in treating substance abuse and dependence. Methods PubMed, EMBASE, ClinicalTrials.gov registry, and the Cochrane Library were searched for trials published before June 2020. Results A total of 16 trials were analyzed. The treatment effectiveness domains assessed in this study were craving and depressive symptoms, withdrawal syndrome, adverse events, and smoking frequency. Standardized mean difference (SMD), weighted mean difference (WMD), and odds ratio (OR) were used for evaluation where appropriate. A significant decrease in craving symptoms was observed in the NAC treatment group compared with the control group (SMD, −0.67; 95% confidence interval [CI], −1.21 to 0.21). When withdrawal and depressive symptoms were considered as a single domain, the NAC treatment group demonstrated a significantly higher overall improvement than the control group (SMD, −0.35; 95% CI, −0.64 to −0.06). No between-group differences in term of the OR of adverse events (OR, 1.18; 95% CI, 0.68 to 2.06) and a non-significant trend toward reduction in smoking frequency was observed in the NAC treatment group compared with the control group (WMD, −3.09; 95% CI, −6.50 to 0.32). Conclusion NAC provides certain noticeable benefits in attenuating substance craving and might help alleviate depressive symptoms and withdrawal syndrome. Precautious measures should be considered when using NAC although no difference in adverse effects was found between NAC treatment and control group.
Collapse
Affiliation(s)
- Chung-Ting Chang
- Department of Primary Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Pei-Ju Hsieh
- Department of Medical Education, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan City, Taiwan
| | - Hsin-Chien Lee
- Department of Psychiatry, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Department of Psychiatry, Taipei Medical University Hospital, Taipei, Taiwan
| | - Chun-Hong Lo
- Department of Primary Medicine, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| | - Ka-Wai Tam
- Center for Evidence-Based Health Care and Shared Decision Making Resource Center, Department of Medical Research, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cochrane Taiwan, Taipei Medical University, Taipei, Taiwan.,Division of General Surgery, Department of Surgery, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - El-Wui Loh
- Center for Evidence-Based Health Care and Shared Decision Making Resource Center, Department of Medical Research, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Cochrane Taiwan, Taipei Medical University, Taipei, Taiwan.,Department of Dentistry, Taipei Medical University Shuang Ho Hospital, New Taipei City, Taiwan
| |
Collapse
|