Exenatide once weekly for smoking cessation: study protocol for a randomized clinical trial

Luba Yammine, Thomas R Kosten, Paul M Cinciripini, Charles E Green, Janet C Meininger, Jennifer A Minnix, Thomas F Newton, Luba Yammine, Thomas R Kosten, Paul M Cinciripini, Charles E Green, Janet C Meininger, Jennifer A Minnix, Thomas F Newton

Abstract

Background: Cigarette smoking is the greatest preventable cause of morbidity and premature mortality in the United States. Approved pharmacological treatments for smoking cessation are marginally effective, underscoring the need for improved pharmacotherapies. A novel approach might use glucagon-like peptide-1 (GLP-1) agonists, which reduce alcohol and drug use in preclinical studies. GLP-1 is produced in the intestinal L-cells and in the hindbrain. The peptide maintains glucose homeostasis and reduces food intake. Several GLP-1 agonists are used clinically to treat type 2 diabetes and obesity, but none have been tested in humans to reduce smoking.

Aims: We will examine whether extended-release exenatide reduces smoking, craving, and withdrawal symptoms, as well as cue-induced craving for cigarettes.

Methods: We will enroll prediabetic and/or overweight treatment seeking smokers (n = 90) into a double-blind, placebo-controlled, randomized clinical trial. Participants will be randomized in a 1:1 ratio to receive exenatide or placebo. All participants will receive transdermal nicotine replacement therapy (NRT) and behavioral counseling. Abstinence from smoking (verified via expired CO level of ≤5 ppm), craving (Questionnaire of Smoking Urges score), and withdrawal symptoms (Wisconsin Scale of Withdrawal Symptoms score) will be assessed weekly during 6 weeks of treatment and at 1 and 4 weeks posttreatment. Cue-induced craving for cigarettes will be assessed at baseline and at 3 weeks of treatment following virtual reality exposure.

Expected outcomes: We hypothesize that exenatide will increase the number of participants able to achieve complete smoking abstinence above that achieved via standard NRT and that exenatide will reduce craving and withdrawal symptoms, as well as cue-induced craving for cigarettes.

Conflict of interest statement

The authors have no conflicts of interest to disclose.

Copyright © 2017 The Authors. Published by Wolters Kluwer Health, Inc. All rights reserved.

Figures

https://www.ncbi.nlm.nih.gov/pmc/articles/instance/5943874/bin/medi-97-e9567-g002.jpg

References

    1. The Health Consequences of Smoking-50 Years of Progress: A Report of the Surgeon General. Atlanta Georgia 2014.
    1. Polosa R, Benowitz NL. Treatment of nicotine addiction: present therapeutic options and pipeline developments. Trends Pharmacol Sci 2011;32:281–9.
    1. Dani JA, Jenson D, Broussard JI, et al. Neurophysiology of nicotine addiction. J Addict Res Ther 2011;S1:
    1. Shirazi RH, Dickson SL, Skibicka KP. Gut peptide GLP-1 and its analogue, exendin-4, decrease alcohol intake and reward. PLoS One 2013;8:e61965.
    1. Egecioglu E, Steensland P, Fredriksson I, et al. The glucagon-like peptide 1 analogue Exendin-4 attenuates alcohol mediated behaviors in rodents. Psychoneuroendocrinology 2013;38:1259–70.
    1. Egecioglu E, Engel JA, Jerlhag E. The glucagon-like Peptide 1 analogue, exendin-4, attenuates the rewarding properties of psychostimulant drugs in mice. PLoS One 2013;8:e69010.
    1. Graham DL, Erreger K, Galli A, et al. GLP-1 analog attenuates cocaine reward. Mol Psychiatry 2013;18:961–2.
    1. Erreger K, Davis AR, Poe AM, et al. Exendin-4 decreases amphetamine-induced locomotor activity. Physiol Behav 2012;106:574–8.
    1. Egecioglu E, Engel JA, Jerlhag E. The glucagon-like peptide 1 analogue exendin-4 attenuates the nicotine-induced locomotor stimulation, accumbal dopamine release, conditioned place preference as well as the expression of locomotor sensitization in mice. PLoS One 2013;8:e77284.
    1. Reimann F. Molecular mechanisms underlying nutrient detection by incretin-secreting cells. Int Dairy J 2010;20:236–42.
    1. Han VK, Hynes MA, Jin C, et al. Cellular localization of proglucagon/glucagon-like peptide I messenger RNAs in rat brain. J Neurosci Res 1986;16:97–107.
    1. Larsen PJ, Tang-Christensen M, Holst JJ, et al. Distribution of glucagon-like peptide-1 and other preproglucagon-derived peptides in the rat hypothalamus and brainstem. Neuroscience 1997;77:257–70.
    1. Jin SL, Han VK, Simmons JG, et al. Distribution of glucagonlike peptide I (GLP-I), glucagon, and glicentin in the rat brain: an immunocytochemical study. J Comp Neurol 1988;271:519–32.
    1. Holst JJ, Seino Y. GLP-1 receptor agonists: targeting both hyperglycaemia and disease processes in diabetes. Diabetes Res Clin Pract 2009;85:1–3.
    1. Matsuyama T, Komatsu R, Namba M, et al. Glucagon-like peptide-1 (7-36 amide): a potent glucagonostatic and insulinotropic hormone. Diabetes Res Clin Pract 1988;5:281–4.
    1. Gutniak M, Orskov C, Holst JJ, et al. Antidiabetogenic effect of glucagon-like peptide-1 (7-36)amide in normal subjects and patients with diabetes mellitus. N Engl J Med 1992;326:1316–22.
    1. Engel JA, Jerlhag E. Role of appetite-regulating peptides in the pathophysiology of addiction: implications for pharmacotherapy. CNS Drugs 2014;28:875–86.
    1. Skibicka KP. The central GLP-1: implications for food and drug reward. Front Neurosci 2013;7:181.
    1. Naslund E, Schmidt PT, Hellstrom PM. Gut peptide hormones: importance for food intake. Scand J Gastroenterol 2005;40:250–8.
    1. Tang-Christensen M, Larsen PJ, Goke R, et al. Central administration of GLP-1-(7-36) amide inhibits food and water intake in rats. Am J Physiol 1996;271:R848–56.
    1. Turton MD, O'Shea D, Gunn I, et al. A role for glucagon-like peptide-1 in the central regulation of feeding. Nature 1996;379:69–72.
    1. Merchenthaler I, Lane M, Shughrue P. Distribution of pre-pro-glucagon and glucagon-like peptide-1 receptor messenger RNAs in the rat central nervous system. J Comp Neurol 1999;403:261–80.
    1. Alhadeff AL, Rupprecht LE, Hayes MR. GLP-1 neurons in the nucleus of the solitary tract project directly to the ventral tegmental area and nucleus accumbens to control for food intake. Endocrinology 2012;153:647–58.
    1. West R, Schneider N. Craving for cigarettes. Br J Addict 1987;82:407–15.
    1. West RJ, Hajek P, Belcher M. Severity of withdrawal symptoms as a predictor of outcome of an attempt to quit smoking. Psychol Med 1989;19:981–5.
    1. West R. The multiple facets of cigarette addiction and what they mean for encouraging and helping smokers to stop. COPD 2009;6:277–83.
    1. Wray JM, Gass JC, Tiffany ST. A systematic review of the relationships between craving and smoking cessation. Nicotine Tob Res 2013;15:1167–82.
    1. Bush MA, Matthews JE, De Boever EH, et al. Safety, tolerability, pharmacodynamics and pharmacokinetics of albiglutide, a long-acting glucagon-like peptide-1 mimetic, in healthy subjects. Diabetes Obes Metab 2009;11:498–505.
    1. Rosenstock J, Klaff LJ, Schwartz S, et al. Effects of exenatide and lifestyle modification on body weight and glucose tolerance in obese subjects with and without pre-diabetes. Diabetes Care 2010;33:1173–5.
    1. Daniele G, Abdul-Ghani M, DeFronzo RA. What are the pharmacotherapy options for treating prediabetes? Expert Opin Pharmacother 2014;15:2003–18.
    1. Armato J, DeFronzo RA, Abdul-Ghani M, et al. Successful treatment of prediabetes in clinical practice: targeting insulin resistance and beta-cell dysfunction. Endocr Pract 2012;18:342–50.
    1. Fineman M, Flanagan S, Taylor K, et al. Pharmacokinetics and pharmacodynamics of exenatide extended-release after single and multiple dosing. Clin Pharmacokinet 2011;50:65–74.
    1. Parkes DG, Mace KF, Trautmann ME. Discovery and development of exenatide: the first antidiabetic agent to leverage the multiple benefits of the incretin hormone, GLP-1. Expert Opin Drug Discov 2013;8:219–44.
    1. Li L, Shen J, Bala MM, et al. Incretin treatment and risk of pancreatitis in patients with type 2 diabetes mellitus: systematic review and meta-analysis of randomised and non-randomised studies. BMJ 2014;348:g2366.
    1. Chiu WY, Shih SR, Tseng CH. A review on the association between glucagon-like peptide-1 receptor agonists and thyroid cancer. Exp Diabetes Res 2012;2012:924168.
    1. Gray KM, McClure EA, Baker NL, et al. An exploratory short-term double-blind randomized trial of varenicline versus nicotine patch for smoking cessation in women. Addiction 2015;110:1027–34.
    1. Fiore MC, Jaen CR, Baker TB, et al. Treating tobacco use and dependence: 2008 update U.S. Public Health Service Clinical Practice Guideline executive summary. Respir Care 2008;53:1217–22.
    1. Cinciripini PM, Tsoh JY, Wetter DW, et al. Combined effects of venlafaxine, nicotine replacement, and brief counseling on smoking cessation. Exp Clin Psychopharmacol 2005;13:282–92.
    1. Cinciripini PM, Blalock JA, Minnix JA, et al. Effects of an intensive depression-focused intervention for smoking cessation in pregnancy. J Consult Clin Psychol 2010;78:44–54.
    1. Cinciripini PM, Robinson JD, Karam-Hage M, et al. Effects of varenicline and bupropion sustained-release use plus intensive smoking cessation counseling on prolonged abstinence from smoking and on depression, negative affect, and other symptoms of nicotine withdrawal. JAMA Psychiatry 2013;70:522–33.
    1. Thompson-Lake DG, Cooper KN, Mahoney JJ, 3rd, et al. Withdrawal symptoms and nicotine dependence severity predict virtual reality craving in cigarette-deprived smokers. Nicotine Tob Res 2015;17:796–802.
    1. Sheehan DV, Lecrubier Y, Sheehan KH, et al. The Mini-international neuropsychiatric interview (M.I. N. I.): the development and validation of a structured diagnostic psychiatric interview for DSM-IV and ICD-10. J Clin Psychiatry 1998;59(Suppl 20):22–33.
    1. Heatherton TF, Kozlowski LT, Frecker RC, et al. The Fagerstrom test for nicotine dependence: a revision of the Fagerstrom tolerance questionnaire. Br J Addict 1991;86:1119–27.
    1. Sobell LC, Sobell MB, Leo GI, et al. Reliability of a timeline method: assessing normal drinkers’ reports of recent drinking and a comparative evaluation across several populations. Br J Addict 1988;83:393–402.
    1. Welsch SK, Smith SS, Wetter DW, et al. Development and validation of the Wisconsin Smoking Withdrawal Scale. Exp Clin Psychopharmacol 1999;7:354–61.
    1. Cox LS, Tiffany ST, Christen AG. Evaluation of the brief questionnaire of smoking urges (QSU-brief) in laboratory and clinical settings. Nicotine Tob Res 2001;3:7–16.
    1. Watson D, Clark LA, Tellegen A. Development and validation of brief measures of positive and negative affect: the PANAS scales. J Pers Soc Psychol 1988;54:1063–70.
    1. Wijeysundera DN, Austin PC, Hux JE, et al. Bayesian statistical inference enhances the interpretation of contemporary randomized controlled trials. J Clin Epidemiol 2009;62:13.e5–21. e5.
    1. O’Neill RT. FDA's critical path initiative: a perspective on contributions of biostatistics. Biom J 2006;48:559–64.
    1. Woodcock J. FDA introductory comments: clinical studies design and evaluation issues. Clin Trials 2005;2:273–5.
    1. Temple R. How FDA currently makes decisions on clinical studies. Clin Trials 2005;2:276–81.
    1. Spiegelhalter DJ, Myles JP, Jones DR, et al. Methods in health service research. An introduction to Bayesian methods in health technology assessment. BMJ 1999;319:508–12.
    1. Lilford RJ, Thornton JG, Braunholtz D. Clinical trials and rare diseases: a way out of a conundrum. BMJ 1995;311:1621–5.
    1. Mills EJ, Wu P, Lockhart I, et al. Comparisons of high-dose and combination nicotine replacement therapy, varenicline, and bupropion for smoking cessation: a systematic review and multiple treatment meta-analysis. Ann Med 2012;44:588–97.

Source: PubMed

3
Sottoscrivi