Anton RF et al. Combined pharmacotherapies and behavioral interventions for alcohol dependence. The

COMBINE study: a randomized controlled trial. JAMA. 2006;295:2003.

Berglund M. Pharmacotherapy for alcohol dependence. In: Berglund M et al. eds. Treating Alcohol and Drug

Abuse: An Evidence Based Review. Weinheim, Germany: Wiley-VCH; 2003:313.

Kranzler H et al. A double-blind, randomized trial of sertraline for alcohol dependence: moderation by age of

onset and 5-HTTLPR genotype. J Clin Psychopharmacol. 2011;31:22.

Johnson BA et al. Topiramate for treating alcohol dependence: a randomized controlled trial. JAMA.

2007;298:1541.

Johnson BA et al. Improvement of physical health and quality of life of alcohol-dependent individuals with

topiramate treatment: US multisite randomized controlled trial. Arch Intern Med. 2008;168:1188.

Johnson BA et al. Pharmacogenetic approach at the serotonin transporter gene as a method to reduce severe

alcohol consumption. Am J Psychiatry. In press.

Anton RF et al. A randomized, multicenter, double-blind, placebo-controlled study of the efficacy and safety of

aripiprazole for the treatment of alcohol dependence. J Clin Psychopharmacol. 2008;28:5.

Garbutt JC et al. Efficacy and tolerability of long-acting injectable naltrexone for alcohol dependence: a

randomized controlled trial [published corrections appear in JAMA. 2005:293;2864; JAMA. 2005;293:1978].

JAMA. 2005; 293:1617.

George DT et al. Rimonabant (SR141716) has no effect on alcoholself-administration or endocrine measures in

nontreatment-seeking heavy alcohol drinkers. Psychopharmacology (Berl). 2010;208:37.

Farren CK et al. A double-blind, placebo-controlled study of sertraline with naltrexone for alcohol dependence.

Drug Alcohol Depend. 2009;99:317.

Veterans Administration Cooperative Study #1027. Quetiapine fumarate extended release (XR) for the

treatment of alcohol dependency in very heavy drinkers. NIAAA/NIH Final Study Report, February 15, 2010.

Angelini M, Brahmbhatt Y. A review of the pharmacologic options for the treatment of alcohol dependence.

Formulary. 2007;42:14–31.

Leggio L et al. Typologies of alcohol dependence. From Jellinek to genetics and beyond. Neuropsychol Rev.

2009;19:115.

Chick JK et al. Disulfiram treatment of alcoholism. Br J Psychiatry. 1992;161:84.

De Sousa A, De Sousa A. A one-year pragmatic trial of naltrexone vs disulfiram in the treatment of alcohol

dependence. Alcohol. 2004;39:528.

Diehl A et al. Why is disulfiram superior to acamprosate in the routine clinicalsetting? A retrospective long-term

study in 353 alcohol-dependent patients. Alcohol. 2010;45:271.

Petrakis IL et al. VA New England VISN I MIRECC Study Group. Naltrexone and disulfiram in patients with

alcohol dependence and comorbid psychiatric disorders. Biol Psychiatry. 2005;57:1128.

Besson J et al. Combined efficacy of acamprosate and disulfiram in the treatment of alcoholism: a controlled

study. Alcohol Clin Exp Res. 1998;22:573.

Fuller RK, Gordis E. Does disulfiram have a role in alcoholism treatment today? Addiction. 2004;99:21.

Brewer C. Recent developments in disulfiram treatment. Alcohol. 1993;28:383.

Helmbrecht GD, Hoskins IA. First trimester disulfiram exposure: report of two cases. Am J Perinatol.

1993;10:5.

Reitnauer PJ et al. Prenatal exposure to disulfiram implicated in the cause of malformations in discordant

monozygotic twins. Teratology. 1997;56:358.

Saxon A et al. Disulfiram use in patients with abnormal liver function test results. J Clin Psychiatry. 1998;59:313.

Wright C et al. Screening for disulfiram-induced liver test dysfunction in an inpatient alcoholism program.

Alcohol Clin Exp Res. 1993;17:184.

Wright C 4th et al. Disulfiram-induced fulminating hepatitis: guidelines for liver-panel monitoring. J Clin

.

.

.

.

.

.

.

223.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

243.

.

Psychiatry. 1988;49:430.

Daniel DG et al. Capgras delusion and seizures in association with therapeutic dosages of disulfiram. South Med

J. 1987;80:1577.

Amini M, Runyon BA. Alcoholic hepatitis 2010: a clinician’s guide to diagnosis and therapy. World J

Gastroenterol. 2010;16:4905.

Larson EW et al. Disulfiram treatment of patients with both alcohol dependence and other psychiatric disorders:

a review. Alcohol Clin Exp Res. 1992;16:125.

Mueser KT et al. Disulfiram treatment for alcoholism in severe mental illness. Am J Addict. 2003;12:242.

Ciraulo DA et al. Drug Interactions in Psychiatry. 3rd ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2006.

Mason BJ, Heyser CJ. Acamprosate: a prototypic neuromodulator in the treatment of alcohol dependence. CNS

Neurol Disord Drug Targets. 2010;9:23.

Slattery J et al. Prevention of relapse in alcohol dependence. Health Technology Assessment Report 3.

Glasgow: Health Technology Board for Scotland; 2003.

Miller WR et al. Mesa Grande: a methodological analysis of clinical trials of treatments for alcohol use disorders.

Addiction. 2002;97:265.

Mann K et al. The efficacy of acamprosate in the maintenance of abstinence in alcohol depending individuals:

results of a meta-analysis. Alcohol Clin Exp Res. 2003;28:51.

Chick J et al. United Kingdom Multicentre Acamprosate Study (UKMAS): a 6-month prospective study of

acamprosate versus placebo in preventing relapse after withdrawal from alcohol. Alcohol. 2000;35:176.

Gual A, Lehert P. Acamprosate during and after acute alcohol withdrawal: a double-blind placebo-controlled

study in Spain. Alcohol. 2001;36:413.

Poldrugo F. Acamprosate treatment in a long-term community-based alcohol rehabilitation programme.

Addiction. 1997;92:1537.

Kiritze -Topor P et al. A pragmatic trial of acamprosate in the treatment of alcohol dependence in primary care.

Alcohol. 2004;39:520.

Mason B. Individual patient data meta-analysis of predictors of outcome including U.S. and European studies in

acamprosate: new preclinical and clinical findings. Presented at the Research Society on Alcoholism; June 26,

2005; Santa Barbara, CA.

Verheul R et al. Predictors of acamprosate efficacy: results from a pooled analysis of seven European trials

including 1485 alcohol-dependent patients. Psychopharmacology (Berl). 2005;178:167.

Carmen B et al. Efficacy and safety of naltrexone and acamprosate in the treatment of alcohol dependence: a

systematic review. Addiction. 2004;99:811.

Kenna GA. Pharmacogenomics and the future of alcohol dependence treatment. In: Sher L, ed. Research on the

Neurobiology of Alcohol Use Disorders. New York, NY: Nova Publishers; 2008:79.

Mason BJ et al. Effect of oral acamprosate on abstinence in patients with alcohol dependence in a double-blind,

placebo-controlled trial: the role of patient motivation. J Psychiatr Res. 2006;40:383.

Chick J et al. A multicentre, randomized, double-blind, placebo-controlled trial of naltrexone in the treatment of

alcohol dependence or abuse. Alcohol. 2000;35:587.

Namkoong K et al. Acamprosate in Korean alcohol dependent patients: a multi-centre, randomized, double blind,

placebo-controlled study. Alcohol. 2003;38:135.

Roussaux JP et al. Does acamprosate diminish the appetite for alcohol in weaned alcoholics? [in French]. J

Pharm Belg. 1996;51:65.

Rosner S et al. Acamprosate for alcohol dependence. Cochrane Database Syst Rev. 2010;(9):CD004332.

Mason BJ et al. A pharmacokinetic and pharmacodynamic drug interaction study of acamprosate and

naltrexone. Neuropsychopharmacology. 2002;27:596.

Johnson BA et al. Dose-ranging kinetics and behavioral pharmacology of naltrexone and acamprosate, both

alone and combined, in alcohol-dependent subjects. J Clin Psychopharmacol. 2003;23:281.

Mark TL et al. Physicians’ opinions about medications to treat alcoholism. Addiction. 2003;98:617.

Kenna GA et al. Pharmacotherapy, pharmacogenomics, and the future of alcohol dependence treatment, part 1.

Am J Health Syst Pharm. 2004;61:2272.

Srisurapanont M, Jarusuraisin N. Opioid antagonists for alcohol dependence. Cochrane Database Syst Rev.

2005;(1): CD001867.

Kranzler HR, Van Kirk J. Efficacy of naltrexone and acamprosate for alcoholism treatment: a meta-analysis.

Alcohol Clin Exp Res. 2001;25:1335.

Kiefer F et al. Comparing and combining naltrexone and acamprosate in relapse prevention of alcoholism: a

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

.

double-blind, placebo-controlled study. Arch Gen Psychiatry. 2003;60:92.

McCaul ME et al. Naltrexone alters subjective and psychomotor responses to alcohol in heavy drinking subjects.

Neuropsychopharmacology. 2000;22:480.

Jaffe AJ et al. Naltrexone, relapse prevention and supportive therapy with alcoholics: an analysis of patienttreatment matching. J Consult Clin Psychol. 1996;64:1044.

Monti PM et al. Naltrexone’s effect on cue-elicited craving among alcoholics in treatment. Alcohol Clin Exp

Res. 1999;23:1386.

Volpicelli JR et al. Naltrexone and alcohol dependence. Role of subject compliance. Arch Gen Psychiatry.

1997;54:737.

Rubio G et al. Clinical predictors of response to naltrexone in alcoholic patients: who benefits most from

treatment with naltrexone? Alcohol. 2005;40:227.

Ray LA et al. Naltrexone for the treatment of alcoholism: clinical findings, mechanisms of action, and

pharmacogenetics. CNS Neurol Disord Drug Targets. 2010;9:13.

Soyka M, Rosner S. Opioid antagonists for pharmacological treatment of alcohol dependence—a critical review.

Curr Drug Abuse Rev. 2008;1:280.

Davidson D et al. Effects of naltrexone on alcohol self-administration in heavy drinkers. Alcohol Clin Exp Res.

1999; 23:195.

Kranzler HR et al. Naltrexone vs. nefazodone for treatment 5 of alcohol dependence. A placebo-controlled trial.

Neuropsychiatry. 2000;22:493.

Krystal JH et al; Veterans Affairs Naltrexone Cooperative Study 425 Group. Naltrexone in the treatment of

alcohol dependence. N EnglJ Med. 2001;345:1734.

Gastpar M et al. Lack of efficacy of naltrexone in the prevention of alcohol relapse: results from a German

multicenter study. J Clin Psychopharmacol. 2002;22:592.

Galloway GP et al. Pharmacokinetics, safety, and tolerability of a depot formulation of naltrexone in alcoholics:

an open label trial. BMC Psychiatry. 2005;5:18.

Kranzler HR et al. Drug Abuse Sciences Naltrexone Depot Study Group. Naltrexone depot for treatment of

alcohol dependence: a multicenter, randomized, placebo controlled clinical trial. Alcohol Clin Exp Res.

2004;28:1051.

Lee JD et al. Extended-release naltrexone for treatment of alcohol dependence in primary care. J Subst Abuse

Treat. 2010;39:14.

Rohsenow DJ. What place does naltrexone have in the treatment of alcoholism? CNS Drugs. 2004;18:547.

Vivitrol (naltrexone). Silver Spring, MD.: US Food and Drug Administration, US Department of Health and

Human Services; Last updated August 12, 2013.

http://www.fda.gov/safety/medwatch/safetyinformation/ucm208449.htm. Accessed January 31, 2017.

Gerasimov MR et al. GABAergic blockade of cocaine associated cue-induced increases in nucleus accumbens

dopamine. Eur J Pharmacol. 2001;414:205.

Gryder DS, Rogawski MA. Selective antagonism of Glu R5 kainate-receptor-mediated synaptic currents by

topiramate in rat basolateral amygdala neurons. J Neurosci. 2003;23:7069.

Swift RM. Topiramate for the treatment of alcohol dependence: initiating abstinence. Lancet. 2003;361:1666.

Topamax [package insert]. Raritan, NJ: Ortho-McNeil-Janssen Pharmaceutical Inc.; 2009.

Di Sclafani V et al. Psychiatric comorbidity in long term abstinent alcoholic individuals. Alcohol Clin Exp Res.

2007;31:795

p. 1902

Cigarette smoking is the single most preventable cause of premature

death in the United States. Smoking harms nearly every organ of the

body, causing many diseases (including, but not limited to,

cardiovascular disease, pulmonary disease, and cancers) and reducing

the health of smokers in general. Quitting smoking has immediate as

well as long-term benefits, reducing risks for diseases caused by

smoking and improving health in general.

Case 91-2 (Questions 1, 4),

Case 91-3 (Question 1),

Case 91-5 (Question 1),

Case 91-6 (Question 1)

Tobacco products are effective delivery systems for the drug nicotine.

Nicotine is a highly addictive drug that activates the dopamine reward

pathway in the brain that reinforces continued tobacco use. Nicotine

withdrawalsymptoms (e.g., irritability, anxiety, difficulty concentrating,

restlessness, depressed mood, insomnia, impaired performance,

increased appetite or weight gain, cravings) generally occur when

nicotine is discontinued.

Case 91-1 (Questions 2, 4),

Case 91-3 (Question 1),

Case 91-6 (Questions 1, 2)

Constituents in tobacco smoke are associated with a number of clinically

significant drug interactions.

Case 91-4 (Questions 1, 3),

Case 91-6 (Question 3)

Tobacco dependence, a chronic disease that often requires repeated

intervention and multiple attempts to quit, is characterized by physiologic

dependence (addiction to nicotine) and behavioral habit of using

tobacco.

Case 91-1 (Question 5),

Case 91-2 (Question 3),

Case 91-3 (Question 1)

Numerous effective medications, as delineated in the Clinical Practice

Guideline, are available for treating tobacco use and dependence. Most

patients should be encouraged to use one or more first-line agents,

which include the nicotine patch, nicotine gum, nicotine lozenge, nicotine

nasalspray, nicotine oral inhaler, sustained-release bupropion, and

varenicline. All first-line agents approximately double quit rates, and

therefore, the choice of therapy is based largely on contraindications,

precautions, patient preference, and tolerability. In some cases,

medications can be combined or used for extended durations. Although

alternative therapies are available, these are not recommended because

of insufficient evidence of efficacy.

Case 91-1 (Questions 1–3),

Case 91-2 (Questions 3, 4),

Case 91-3 (Question 1),

Case 91-4 (Question 2),

Case 91-6 (Questions 1, 4)

Comprehensive counseling, as defined by the Clinical Practice Guideline,

includes asking about tobacco use, advising patients to quit, assessing

readiness to quit, assisting patients with quitting, and arranging followCase 91-1 (Questions 2, 5),

Case 91-3 (Question 1),

Case 91-5 (Question 1),

up. This approach is referred to as “The 5 A’s.” Counseling and support

can be provided a variety of ways, such as through individual

counseling, group programs, telephone, or the Internet. Two components

of counseling are especially effective and should be applied when

assisting patients with quitting: practical counseling (problem solving or

skills training) and socialsupport delivered as part of treatment. Relapse

is common, and clinicians should work with patients throughout the quit

attempt to increase the chances for long-term abstinence.

Case 91-6 (Questions 1, 2, 4)

Brief tobacco dependence treatment (<3 minutes) is effective. In the

absence of time or expertise, ask about tobacco use, advise patients to

quit, and refer patients to other resources (e.g., telephone quitlines, webbased support, local programs) for additional assistance. This approach

is referred to as “Ask-Advise-Refer.”

Case 91-5 (Question 1)

p. 1903

p. 1904

Patients with psychiatric disorders exhibit a higher prevalence of tobacco

use and a disproportionately high level of tobacco-related morbidity and

mortality.

Case 91-6 (Question 1)

Cigarettes are the most common form of tobacco used in the United

States; however, other forms of tobacco exist (spit tobacco, pipes,

cigars, bidis, hookah). All forms of tobacco are harmful.

Case 91-2 (Question 2),

Narrated PowerPoint slides

on Forms of Tobacco

Tobacco is a detrimental substance, and its use dramatically increases a person’s

odds of dependence, disease, disability, and death. Cigarettes are the only marketed

consumable product that when used as intended will contribute to the death of half or

more of its users.

1 Tobacco products are carefully engineered formulations that

optimize the delivery of nicotine, a chemical that meets the criteria for an addictive

substance: (a) nicotine induces psychoactive effects, (b) it is used in a highly

controlled or compulsive manner, and (c) behavioral patterns of tobacco use are

reinforced by the pharmacologic effects of nicotine.

2 As a major risk factor for a

wide range of diseases, including cardiovascular conditions, cancers, and pulmonary

disorders, tobacco is the primary known preventable cause of premature death and

disease in our society.

3 Since the Surgeon General’s first report on smoking in 1964,

over 20 million American deaths have been attributed to smoking or secondhand

smoke exposure.

3 Globally, almost 7 million people die annually from smoking (6

million) or being exposed to secondhand smoke (890,000).

4

In the United States, smoking is responsible for more than 480,000 premature

deaths each year.

3

In addition to the harm imposed on users of tobacco, exposure to

secondhand smoke results in an estimated 50,000 deaths annually.

5,6 According to the

Office of the US Surgeon General, there is no risk-free level of exposure to tobacco

smoke.

2 Because of the health and societal burdens that it imposes, tobacco use and

dependence should be addressed during each clinical encounter with all tobacco

users.

7

EPIDEMIOLOGY OF TOBACCO USE AND

DEPENDENCE

In the United States, cigarettes are the most common form of tobacco that is

consumed, but other forms are also prevalent: smokeless tobacco (chewing tobacco,

oral snuff), pipes, cigars, clove cigarettes, bidis, and hookah. Electronic cigarettes

(e-cigarettes) are growing in popularity. Electronic cigarettes, electronic nicotine

delivery systems (ENDS), contain a liquid consisting of nicotine and other

substances that is heated by an atomizer and inhaled as a vapor. Among adults,

smoking prevalence varies by sociodemographic factors, including sex, race or

ethnicity, education level, age, and poverty level. The Centers for Disease Control

and Prevention (CDC) reported that in 2014, 21.3% of adults in the United States

used a tobacco product every day or some days, with 17% reporting cigarette use.

8

Smoking prevalence is increased among persons with mental illness, with over onethird smoking cigarettes.

9

Factors Contributing to Tobacco Use

Tobacco addiction is maintained by nicotine dependence.

10,11 Nicotine induces a

variety of pharmacologic effects that lead to dependence.

12 However, tobacco

dependence is not simply a matter of nicotine pharmacology—it is a result of the

interplay of complex processes, including the desire for the direct pharmacologic

actions of nicotine, the relief of withdrawal, learned associations, and environmental

cues (e.g., advertising, the smell of a cigarette, or observing others who are

smoking).

11 Physiologic factors, such as preexisting medical conditions (e.g.,

psychiatric comorbidities

9,11 and one’s genetic profile), also can predispose

individuals to tobacco use.

12,13

Nicotine, the addictive component of tobacco, is rapidly absorbed and passes

through the blood–brain barrier, contributing to its addictive nature. After inhalation,

nicotine reaches the brain within seconds.

10 As such, smokers experience nearly

immediate onset of the positive effects of nicotine, including pleasure, relief of

anxiety, improved task performance, improved memory, mood modulation, and

skeletal muscle relaxation.

10 These effects, mediated by alterations in

neurotransmitter levels, reinforce continued use of nicotine-containing products.

10,11

Nicotine Pharmacology

Nicotine (Nicotiana tabacum) is one of the few natural alkaloids that exist in the

liquid state. Nicotine is a clear, weak base with a pKα of 8.0.

14

In acidic media,

nicotine is ionized and poorly absorbed; conversely, in alkaline media, nicotine is

nonionized and well absorbed. Under physiologic conditions (pH = 7.4), a large

proportion of nicotine is nonionized and readily crosses cell membranes.

14 Given the

relation between pH and absorption, the tobacco industry and pharmaceutical

companies are able to titrate the pH of their tobacco products and nicotine

replacement therapy (NRT) products to maximize the absorption potential of

nicotine.

14–16

Once absorbed, nicotine induces a variety of central nervous system,

cardiovascular, and metabolic effects. Nicotine stimulates the release of several

neurotransmitters, inducing a range of pharmacologic effects such as pleasure

(dopamine), arousal (acetylcholine, norepinephrine), cognitive enhancement

(acetylcholine), appetite suppression (dopamine, norepinephrine, serotonin), learning

(glutamate), memory enhancement (glutamate), mood modulation (serotonin), and

reduction of anxiety and tension (β-endorphin and γ-aminobutyric acid [GABA]).

17

The dopamine reward pathway, a network that elicits feelings of pleasure in

response to certain stimuli, is central to drug-induced reward. The neurons of the

ventral tegmental area contain the neurotransmitter dopamine, which is released in

the nucleus accumbens and in the prefrontal cortex. Immediately after inhalation, a

bolus of nicotine enters the brain, stimulating the release of dopamine, which induces

nearly immediate feelings of pleasure, along with relief of the symptoms of nicotine

withdrawal. This rapid dose response reinforces repeated administration of the drug

and perpetuates the smoking behavior.

11,17

Chronic administration of nicotine has been shown to increase the number of

nicotine receptors in specific regions of the brain.

18 This may represent upregulation

in response to nicotine-mediated desensitization of the receptors and could play a

role in nicotine tolerance and dependence.

17,18 Chronic nicotine administration also

leads to tolerance of its behavioral and cardiovascular effects during the course of

the day; however, tobacco users regain sensitivity to the effects of nicotine after

overnight abstinence from nicotine,

10,11 as shown in Figure 91-1.

19 After smoking the

first cigarette of the day, the smoker experiences marked pharmacologic effects,

particularly arousal. No other cigarette throughout the day produces the same degree

of pleasure or arousal. For this reason, many smokers describe the first cigarette as

the most important one of the day. Shortly after the initial cigarette, tolerance begins

to develop. Accordingly, the threshold levels for both pleasure or arousal and

abstinence rise progressively throughout the day because the smoker becomes

tolerant to the effects of nicotine. With continued smoking, nicotine accumulates,

leading to an even greater degree of tolerance. Later in the day, each individual

cigarette produces only limited pleasure or arousal; instead, smoking primarily

alleviates nicotine withdrawal symptoms. Lack of exposure to nicotine overnight

results in resensitization of drug responses (i.e., loss of tolerance). Most dependent

smokers tend to smoke a certain number of cigarettes/day and tend to consume

sufficient nicotine/day to achieve the desired effects of cigarette smoking and

minimize the symptoms of nicotine withdrawal.

11,19 Withdrawal symptoms, which

include anger, anxiety, depression, difficulty concentrating, impatience, insomnia,

and restlessness, typically manifest within a few days after quitting, peak within a

week, and subside within 2 to 4 weeks.

20 Tobacco users become adept at titrating

their nicotine levels throughout the day to avoid withdrawal symptoms, maintain

pleasure and arousal, and modulate mood.

p. 1904

p. 1905

Figure 91-1 Nicotine addiction cycle throughout the day. The sawtooth line represents venous plasma

concentrations of nicotine because a cigarette is smoked every 40 minutes between 8 AM and 9 PM. The upper

solid line indicates the threshold concentration for nicotine to produce pleasure or arousal. The lower solid line

indicates the concentrations at which symptoms of abstinence (i.e., withdrawalsymptoms) from nicotine occur.

The shaded area represents the zone of nicotine concentrations (neutral zone) in which the smoker is comfortable

without experiencing either pleasure and arousal or abstinence symptoms. (Reprinted with permission from

Benowitz NL. Cigarette smoking and nicotine addiction. Med Clin North Am. 1992;76(2):415.)

Nicotine is extensively metabolized in the liver and, to a lesser extent, in the

kidney and lung. Approximately 70% to 80% of nicotine is metabolized to cotinine,

an inactive metabolite.

14 The rapid metabolism of nicotine (half-life [t1/2

] = 2 hours)

to inactive compounds underlies tobacco users’ needs for frequent, repeated

administration. The half-life of cotinine, however, is much longer (t1/2 = 18–20

hours), and for this reason, cotinine is commonly used as a marker of tobacco use as

well as a marker for exposure to secondhand smoke.

14 Measurement of cotinine

cannot, however, differentiate between the nicotine from tobacco products and the

nicotine from NRT products. Nicotine and other metabolites are excreted in the urine.

Urinary excretion is pH dependent; the excretion rate is increased in acidic urine.

14

Nicotine crosses the placenta and accumulates in breast milk.

14

Drug Interactions with Smoking

It is widely recognized that polycyclic aromatic hydrocarbons (PAHs) in tobacco

smoke are responsible for most drug interactions with smoking.

21,22 PAHs, which

result from incomplete combustion of tobacco, are potent inducers of several hepatic

cytochrome-P450 microsomal enzymes (CYP1A1, CYP1A2, and possibly CYP2E1).

Although other substances in tobacco smoke, including acetone, pyridines, benzene,

nicotine, carbon monoxide, and heavy metals (e.g., cadmium), might also interact

with hepatic enzymes, their effects appear to be less significant. Most drug

interactions with tobacco smoke are pharmacokinetic, resulting from the induction of

drug-metabolizing enzymes (especially CYP1A2) by compounds in tobacco smoke.

Table 91-1 summarizes key interactions with smoking.

21,22,23 Patients who begin

smoking, quit smoking, or dramatically alter their level of smoking might require

dosage adjustments for some medications.

Health Consequences of Tobacco Use

All forms of tobacco are harmful, and there is no safe level of exposure to tobacco

products. Smoking has a causal or contributory role in the development of a variety

of medical conditions, affecting almost every organ in the body.

2,3

SECONDHAND SMOKE EXPOSURE

Exposure to secondhand smoke, which includes the smoke from burning tobacco and

that exhaled by the smoker, affects an estimated 88 million nonsmokers older than the

age of 3 in the United States.

24 Secondhand smoke exposure causes disease and

premature death in children and adults who do not smoke resulting in an estimated

50,000 deaths annually.

6 Millions of American children and adults are still exposed

to secondhand smoke in their homes and workplaces despite substantial progress in

tobacco control. Evidence indicates that there is no risk-free level of exposure to

secondhand smoke. Only completely eliminating smoking in indoor spaces fully

protects nonsmokers from exposure to secondhand smoke. Separating smokers from

nonsmokers, cleaning the air, and ventilating buildings cannot eliminate exposure of

nonsmokers to secondhand smoke.

Benefits of Quitting

Benefits of smoking cessation incurred soon after quitting (e.g., within 2 weeks–3

months) include improvements in pulmonary function, circulation, and ambulation.

Smoking cessation results in measurable improvements in lung function (see Chapter

19, Chronic Obstructive Pulmonary Disease). One year after cessation, the excess

risk of coronary heart disease is reduced to half that of continuing smokers. After 5 to

15 years, the risk of stroke is reduced to a rate similar to that of people who are

lifetime nonsmokers, and 10 years after quitting, the chance of dying of lung cancer is

approximately half that of continuing smokers. In addition, the risk of developing

mouth, throat, esophagus, bladder, kidney, or pancreatic cancer is decreased. Finally,

15 years after quitting, the risk of coronary heart disease is reduced to a rate that is

similar to that of people who have never smoked.

25 Similarly, more recent data

suggest that smokers who quit for good over a sustained period have an overall

mortality rate and death rate associated with cardiovascular disease, ischemic heart

disease, and stroke that is similar to individuals who have never smoked. In contrast,

individuals who had successfully quit, but later resumed smoking, had mortality risks

that were significantly higher than lifetime nonsmokers.

26

p. 1905

p. 1906

Table 91-1

Drug Interactions with Tobacco Smoke

Drug/Class Mechanism of Interaction and Effects

Pharmacokinetic Interactions

Alprazolam (Xanax) Conflicting data on significance, but possible ↓ plasma concentrations (up to 50%);

↓ half-life (35%).

Bendamustine

(Treanda)

Metabolized by CYP1A2. Manufacturer recommends using with caution in

smokers due to likely ↓ bendamustine concentrations, with ↑

concentrations of its two active metabolites.

Caffeine ↑ Metabolism (induction of CYP1A2); ↑ clearance (56%).

Caffeine levels likely increase after cessation.

Chlorpromazine

(Thorazine)

↓ AUC (36%) and serum concentrations (24%).

↓ Sedation and hypotension possible in smokers; smokers may require ↑ dosages.

Clopidogrel (Plavix) ↑ Metabolism (induction of CYP1A2) of clopidogrel to its active

metabolite.

Clopidogrel’s effects are enhanced in smokers (≥10 cigarettes/day):

significant ↑ platelet inhibition, ↓ platelet aggregation; although

improved clinical outcomes have been shown, may also ↑ risk of

bleeding.

Clozapine (Clozaril) ↑ Metabolism (induction of CYP1A2); ↓ plasma concentrations (18%).

↑ Levels on cessation may occur; closely monitor drug levels and reduce

dose as required to avoid toxicity.

Erlotinib (Tarceva) ↑ Clearance (24%); ↓ trough serum concentrations (2-fold).

Flecainide (Tambocor) ↑ Clearance (61%); ↓ trough serum concentrations (25%).

Smokers may need ↑ dosages.

Fluvoxamine (Luvox) ↑ Metabolism (induction of CYP1A2); ↑ clearance (24%); ↓ AUC (31%); ↓

Cmax (32%) and Css (by 39%).

Dosage modifications not routinely recommended but smokers may need

↑ dosages.

Haloperidol (Haldol) ↑ Clearance (44%); ↓ serum concentrations (70%); data are inconsistent

therefore clinical significance is unclear.

Heparin Mechanism unknown but ↑ clearance and ↓ half-life are observed. Smoking has

prothrombotic effects.

Smokers may need ↑ dosages because of PK and PD interactions.

Insulin, subcutaneous Possible ↓ insulin absorption secondary to peripheral vasoconstriction; smoking

may cause release of endogenous substances that cause insulin resistance.

PK and PD interactions likely not clinically significant but smokers may need ↑

dosages.

Irinotecan (Camptosar) ↑ Clearance (18%); ↓ serum concentrations of active metabolite SN-38

(∼40%; via induction of glucuronidation); ↓ systemic exposure resulting

in lower hematologic toxicity and may reduce efficacy.

Smokers may need ↑ dosages.

Methadone Possible increase ↑ metabolism (induction of CYP1A2, a minor pathway for

methadone.)- Carefully monitor response upon cessation.

Mexiletine (Mexitil) ↑ Clearance (25%; via oxidation and glucuronidation); ↓ half-life (36%).

Olanzapine (Zyprexa) ↑ Metabolism (induction of CYP1A2): ↑ clearance (98%); ↓ serum

concentrations (12%).

Dosage modifications not routinely recommended but smokers may need

↑ dosages.

Propranolol (Inderal) ↑ Clearance (77%; via side-chain oxidation and glucuronidation).

Riociguat (Adempas) ↓ Plasma concentrations (by 50%–60%)

Smokers may require dosages higher than 2.5 mg 3 times daily; consider

dose reduction upon cessation.

Ropinirole (Requip) ↓ Cmax

(30%) and AUC (38%) in study with patients with restless legs

syndrome.

Smokers may need ↑ dosages.

Tacrine (Cognex) Increase ↑ metabolism (induction of CYP1A2); decrease ↓ half-life (50%); serum

concentrations 3-fold lower. -Smokers may need increased ↑ dosages.

Tasimelteon (Hetlioz) Increased ↑ Metabolism (induction of CYP1A2); drug exposure decreased ↓ by

40%. -Smokers may need increased ↑ dosages.

Theophylline (TheoDur, etc.)

↑ Metabolism (induction of CYP1A2); ↑ clearance (58%–100%); ↓ half-life

(63%).

Levels should be monitored if smoking is initiated, discontinued, or

changed.

↑ Clearance with secondhand smoke exposure.

Maintenance doses are considerably higher in smokers; ↑ Clearance also

with secondhand smoke exposure.

Tizanidine (Zanaflex) ↓ AUC (30%–40%) and ↓ half-life (10%) observed in male smokers.

p. 1906

p. 1907

Tricyclic antidepressants

(e.g., imipramine,

nortriptyline)

Possible interaction with tricyclic antidepressants in the direction of ↓ blood levels,

but the clinicalsignificance is not established.

Warfarin ↑ Metabolism (induction of CYP1A2) of R-enantiomer; however, S-enantiomer is

more potent and effect on INR is inconclusive. Consider monitoring INR on

smoking cessation.

Pharmacodynamic Interactions

Benzodiazepines

(diazepam,

chlordiazepoxide)

↓ Sedation and drowsiness, possibly caused by nicotine stimulation of central

nervous system.

β-Blockers Less effective antihypertensive and heart rate control effects; possibly caused by

nicotine-mediated sympathetic activation.

Smokers may need ↑ dosages.

Corticosteroids, inhaled Smokers with asthma may have less of a response to inhaled

corticosteroids.

Hormonal

contraceptives

(combined)

↑ Risk of cardiovascular adverse effects (e.g., stroke, myocardial

infarction, thromboembolism) in women who smoke and use combined

hormonal contraceptives.

↑ Risk with age and with heavy smoking (15 or more cigarettes/day) and is

quite marked in women aged 35 and older.

Serotonin 5-HT1 receptor

agonists (triptans)

This class of drugs may cause coronary vasospasm; caution for use in smokers

due to possible unrecognized CAD.

Bold rows indicate the most clinically significant interactions.

AUC, area under the curve; Cmax

, maximal concentration; Css, steady state concentration; INR, international

normalized ratio; PD, pharmacodynamics; PK, pharmacokinetics.

Adapted with permission from Rx for Change: Clinician-Assisted Tobacco Cessation. Copyright © 1999–2017.

The Regents of the University of California. All rights reserved.

Quitting at ages 30, 40, 50, and 60 results in 10, 9, 6, and 3 years of life gained,

respectively.

1 On average, cigarette smokers die approximately 10 years younger

than do nonsmokers, and of those who continue smoking, at least half will eventually

die as a result of a tobacco-related disease. Persons who quit before age 35 add 10

years of life and have a life expectancy similar to men who had never smoked.

1 A

reduction in smoking does not equate to a reduction in harm,

27 and even low levels of

smoking (e.g., 1–4 cigarettes/day) have documented risks.

28,29 Therefore, decreasing

the number of cigarettes smoked/day should be viewed as a positive step toward

quitting, but should not be recommended as a targeted end point. For any patient who

uses tobacco, the goal is complete, long-term abstinence from all nicotine-containing

products.

Tobacco Use and Dependence: Treatment Approaches

Most tobacco users attempt to quit without assistance, despite the fact that persons

who receive assistance are more likely to be successful in quitting.

7,30 Given the

complexity of the tobacco dependence syndrome and the constellation of factors that

contribute to tobacco use, treatment requires a multifaceted approach. To assist

clinicians and other specialists in providing cessation treatment to patients who use

tobacco, the US Public Health Service published the Clinical Practice Guideline for

Treating Tobacco Use and Dependence. This document, which represents a

distillation of more than 8,700 published articles,

7 specifies that clinicians can have

an important impact on their patients’ ability to quit. A meta-analysis of 29 studies

7

estimated that compared with patients who do not receive an intervention from a

clinician, patients who receive a tobacco-cessation intervention from a physician

clinician or a nonphysician clinician are 2.2 and 1.7 times, respectively, more likely

to quit (at 5 or more months after cessation). Although even brief advice from a

clinician has been shown to lead to increased odds of quitting,

7 more intensive

counseling yields more dramatic increases in quit rates.

7 Other effective methods for

delivery of counseling include group programs

7,31 and telephone counseling.

32

Internet-based interventions have become more prevalent in recent years, but a metaanalysis of 28 trials revealed inconsistent results.

33

Numerous effective medications are available for tobacco dependence, and

clinicians should encourage their use by all patients attempting to quit smoking—

except when medically contraindicated or with specific populations for which there

is insufficient evidence of effectiveness (i.e., pregnant women, smokeless tobacco

users, light smokers, adolescents).

7 Although both pharmacotherapy and behavioral

counseling are effective independently, patients’ odds of quitting are substantially

increased when the two approaches are used simultaneously.

34 Clinicians can have a

significant impact on a patient’s likelihood of success by recommending

pharmacotherapy agents and by supplementing medication use with behavioral

counseling as described later in this chapter.

p. 1907

p. 1908

Assisting Patients with Quitting

BEHAVIORAL COUNSELING STRATEGIES

According to the Clinical Practice Guideline,

7

five key components constitute

comprehensive counseling for tobacco cessation: (a) asking patients whether they use

tobacco, (b) advising tobacco users to quit, (c) assessing patients’ readiness to quit,

(d) assisting patients with quitting, and (e) arranging follow-up care. These steps are

referred to as the “5 A’s” and are described, in brief, as follows. Figure 91-2 can be

used as a guide for structuring counseling interactions.

Ask: Screening for tobacco use is essential and should be a routine component of

clinical care. The following question can be used to identify tobacco users: “Do

you ever smoke or use any type of tobacco?” At a minimum, tobacco use status

(current, former, never user) and level of use (e.g., number of cigarettes

smoked/day) should be assessed and documented in the medical record. Also,

patients should be asked about exposure to secondhand smoke.

Advise: Tobacco users should be advised to consider quitting; the advice should be

clear and compelling, yet delivered with sensitivity and a tone of voice that

communicates concern and a willingness to assist with quitting. When possible,

messages should be personalized by relating advice to factors such as a patient’s

health status, medication regimen, personal reasons for wanting to quit, or the

impact of tobacco use on others. For example, “I’m concerned because you are on

two different inhalers for your emphysema. Quitting smoking is the single most

important treatment to improve your breathing. I strongly encourage you to quit.

Would you be interested in having me help you with this?”

Assess: Key to the provision of appropriate counseling interventions is the

assessment of a patient’s readiness to quit. Patients should be categorized as

being (a) not ready to quit in the next month; (b) ready to quit in the next month;

(c) a recent quitter, having quit in the past 6 months; or (d) a former user, having

quit more than 6 months ago.

7,35 This classification defines the clinician’s next

step, which is to provide counseling that is tailored to the patient’s level of

readiness to quit. As an example for a current smoker: “Mr. Malkin, what are

your thoughts about quitting, and would you consider quitting sometime in the next

month?” The counseling interventions for patients who are ready to quit will be

different from those for patients who are not considering quitting.

Assist: When counseling tobacco users, it is important that clinicians and patients

view quitting as a process that might take months or even years to achieve. The

goal is to promote forward progress in the process of change, with the target end

point being sustained abstinence from all nicotine-containing products.

When counseling patients who are not ready to quit, an important first step is to

foster motivation. Some patients are not convinced that quitting is important, but most

recognize the need to quit and are simply not ready to make the commitment to do so.

Often, patients have tried to quit multiple times and failed, and thus are too

discouraged to try again. The “5 R’s” can be applied to enhance motivation to quit

7

(Table 91-2) by clinicians offering to work closely with the patient in designing a

treatment plan. Although it might be useful to educate patients about the

pharmacotherapy options, it is inappropriate to prescribe a treatment regimen for

patients who are not ready to quit. For patients who are not ready to quit in the next

30 days, encourage them to seriously consider quitting and ask the following

questions:

Figure 91-2 Tobacco-cessation counseling guide sheet. (Reprinted with permission from Rx for Change:

Clinician-Assisted Tobacco Cessation. Copyright © 1999–2017. The Regents of the University of California. All

rights reserved.)

p. 1908

p. 1909

Table 91-2

Enhancing Motivation to Quit: The “5 R’s” for Tobacco-Cessation Counseling

Relevance—Encourage patients to think about the reasons why quitting is important. Counseling should be

framed such that it relates to the patient’s risk for disease or exacerbation of disease, family or social

situations (e.g., having children with asthma), health concerns, age, or other patient factors, such as prior

experience with quitting.

Risks—Ask patients to identify potential negative health consequences of smoking, such as acute risks

(shortness of breath, asthma exacerbations, harm to pregnancy, infertility), long-term risks (cancer, cardiac,

and pulmonary disease), and environmental risks (promoting smoking among children by being a negative role

1.

2.

3.

model; effects of secondhand smoke on others, including children and pets).

Rewards—Ask patients to identify potential benefits that they anticipate from quitting, such as improved

health, enhanced physical performance, enhanced taste and smell, reduced expenditures for tobacco, less

time wasted or work missed, reduced health risks to others (fetus, children, housemates), and reduced aging

of the skin.

Roadblocks—Help patients identify barriers to quitting and assist in developing coping strategies (Table 91-4)

for addressing each barrier. Common barriers include nicotine withdrawalsymptoms, fear of failure, a need

for socialsupport while quitting, depression, weight gain, and a sense of deprivation or loss.

Repetition—Continue to work with patients who are successful in their quit attempt. Discuss circumstances in

which smoking occurred to identify the trigger(s) for relapse; this is part of the learning process and will be

useful information for the next quit attempt. Repeat interventions when possible.

Reprinted from Fiore MC et al. Treating Tobacco Use and Dependence: 2008 Update. Clinical Practice Guideline.

Rockville, MD: Public Health Service, U.S. Department of Health and Human Services; 2008.

Do you ever plan to quit?

If the patient responds “no,” the clinician should ask, “What would have to change

for you to decide to quit?” If the patient responds “nothing,” then offer to assist, if

or when the patient changes his or her mind. If the patient responds “yes,” the

clinician should continue with question 2.

What might be some benefits of quitting now, instead of later?

The longer a patient smokes, quitting generally becomes more difficult. Most

patients will agree that there is never an ideal time to quit, and procrastinating a

quit date has more negative effects than positive.

What would have to change for you to decide to quit sooner?

This question probes patients’ perceptions of quitting, which reveals some of the

barriers to quitting that can then be discussed.

For patients who are ready to quit (i.e., in the next month), the goal is to work with

the patient in designing an individualized treatment plan, addressing the key issues

listed under the “Assist” component of Figure 91-2.

23 The first steps are to discuss

the patient’s tobacco use history, inquiring about levels of smoking, number of years

smoked, methods used previously for quitting (what worked, what did not work and

why), and reason(s) for previous failed quit attempts. Clinicians should elicit

patients’ opinions about the different medications for quitting and should work with

patients in selecting the quitting methods (e.g., medications, behavioral counseling

programs). Although it is important to recognize that pharmaceutical agents might not

be appropriate, desirable, or affordable for all patients, clinicians should educate

patients that medications, when taken correctly, can substantially increase the

likelihood of success.

Patients should select a quit date, ideally within the next 2 weeks. This allows

sufficient time to prepare for the quit attempt, including mental preparation,

preparation of the environment, removing all tobacco products and ashtrays from the

home, car, and workspace, and soliciting support their family, friends, and

coworkers. Additional strategies for coping with quitting are shown in Table 91-3.

Patients should be counseled about withdrawal symptoms, medication use, and the

importance of receiving behavioral counseling throughout the quit attempt. Finally,

patients should be commended for taking important steps toward improving their

health.

Arrange: Because patients’ ability to quit increases when multiple counseling

interactions are provided, arranging follow-up counseling is an important element

of treatment for tobacco dependence. Follow-up contact should occur soon after

the quit date, preferably during the first week. A second follow-up contact is

recommended within the first month after quitting.

7 Additional follow-up contacts

should occur to monitor patient progress, assess compliance with

pharmacotherapy regimens, and provide additional support.

Relapse prevention counseling should be part of every follow-up contact with

patients who have recently quit smoking. When counseling recent quitters, it is

important to address challenges in countering withdrawal symptoms and cravings or

temptations to use tobacco. A list of strategies for key triggers or temptations for

tobacco use is provided in Table 91-3.

23

Importantly, because tobacco use is a

habitual behavior, patients should be advised to alter their daily routines; this helps

disassociate specific behaviors from the use of tobacco. Patients who slip and smoke

a cigarette (or use any form of tobacco) or experience a full relapse back to habitual

tobacco use should be encouraged to think through the scenario in which tobacco use

first occurred and identify the trigger(s) for relapse. This process provides valuable

information for future quit attempts.

PHARMACOTHERAPY OPTIONS

All smokers who are trying to quit should be encouraged to use one or more US Food

and Drug Administration (FDA)-approved pharmacologic aids for cessation.

Potential exceptions that require special consideration include medical

contraindications or use in specific populations for which there is insufficient

evidence of effectiveness (i.e., pregnant women, smokeless tobacco users, light

smokers, adolescents).

7 Pharmacotherapy should always be combined with

behavioral support and counseling. Currently, the FDA-approved first-line agents

that have been shown to be effective in promoting smoking cessation include five

NRT dosage forms, sustained-release bupropion, and varenicline.

7 The choice of

therapy is dictated by considerations such as patient preference for a given agent,

previous experience with cessation medications, current medical conditions,

previous levels of smoking, medication adherence issues, and the patient’s out-ofpocket costs. Dosing information, precautions, and adverse effects for the first-line

agents are shown in Table 91-4.

p. 1909

p. 1910

Comments

Search This Blog

Archive

Show more

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

علاقة البيبي بالفراولة بالالفا فيتو بروتين

التغيرات الخمس التي تحدث للجسم عند المشي

إحصائيات سنة 2020 | تعداد سكَان دول إفريقيا تنازليا :

ما هو الليمونير للأسنان ؟

ACUPAN 20 MG, Solution injectable

CELEPHI 200 MG, Gélule

الام الظهر

VOXCIB 200 MG, Gélule

ميبستان

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

Popular posts from this blog

TRIPASS XR تري باس

CELEPHI 200 MG, Gélule

ZENOXIA 15 MG, Comprimé

VOXCIB 200 MG, Gélule

Kana Brax Laberax

فومي كايند

بعض الادويه نجد رموز عليها مثل IR ، MR, XR, CR, SR , DS ماذا تعني هذه الرموز

NIFLURIL 700 MG, Suppositoire adulte

Antifongiques مضادات الفطريات

Popular posts from this blog

Kana Brax Laberax

TRIPASS XR تري باس

PARANTAL 100 MG, Suppositoire بارانتال 100 مجم تحاميل

الكبد الدهني Fatty Liver

الم اسفل الظهر (الحاد) الذي يظهر بشكل مفاجئ bal-agrisi

SEDALGIC 37.5 MG / 325 MG, Comprimé pelliculé [P] سيدالجيك 37.5 مجم / 325 مجم ، قرص مغلف [P]

نمـو الدمـاغ والتطـور العقـلي لـدى الطفـل

CELEPHI 200 MG, Gélule

أخطر أنواع المخدرات فى العالم و الشرق الاوسط

Archive

Show more