Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2015 Oct 24.
Published in final edited form as: Tob Control. 2010 Oct;19(5):e1–10. doi: 10.1136/tc.2009.035584

Nicotine Reduction Revisited: Science and Future Directions

Dorothy K Hatsukami 1, Kenneth A Perkins 2, Mark G LeSage 3, David L Ashley 4, Jack E Henningfield 5, Neal L Benowitz 6, Cathy Backinger 7, Mitch Zeller 8
PMCID: PMC4618689  NIHMSID: NIHMS307228  PMID: 20876072

Abstract

Regulation of nicotine levels in cigarettes and other tobacco products is now possible with the passage of the Family Smoking Prevention and Tobacco Control Act (FSPTCA) in 2009 giving the U.S. Food and Drug Administration authority to regulate tobacco products, and with Articles 9-11 of the World Health Organization Framework Convention on Tobacco Control.[1-2] Both regulatory approaches allow establishing product standards for tobacco constituents, including nicotine. The FSPTCA does not allow nicotine levels to be decreased to zero, although FDA has the authority to reduce nicotine yields to very low, presumably non-addicting levels. The proposal to reduce levels of nicotine to a level that is non-addicting was originally suggested in 1994.[3] Reduction of nicotine in tobacco products could potentially have a profound impact on reducing tobacco-related morbidity and mortality. To examine this issue, two meetings were convened in the United States with non-tobacco-industry scientists of varied disciplines, tobacco control policy-makers and representatives of government agencies. This article provides an overview of the current science in the area of reduced nicotine content cigarettes and key conclusions and recommendations for research and policy that emerged from the deliberations of the meeting members.

Keywords: Nicotine reduction, nicotine addiction, tobacco performance standards

INTRODUCTION

The authority of the U.S. Food and Drug Administration (FDA) to regulate tobacco offers an opportunity to reduce cigarette smoking prevalence and tobacco-caused morbidity and mortality.[1, 4] Substantial reductions in smoking prevalence might be achieved by mandated reduction in the cigarette's addictiveness. The primary objectives of this reduction should be cessation, prevention of tobacco use and addiction (by delaying or eliminating progression from experimentation to addiction) and substantially reduced use among smokers who do not quit. Although many factors contribute to initiation of tobacco use, there is widespread agreement that nicotine addiction sustains tobacco use.[5-9] Thus, reducing the nicotine in cigarettes to a level that is non-addicting may have an impact on reducing morbidity and mortality and improving public health.

In 1994, Benowitz and Henningfield proposed gradually reducing nicotine levels of all cigarettes over 10-15 years.[3] Computer simulation modeling the effects of reducing nicotine in cigarettes to nonaddictive levels projected a decline in smoking prevalence from 23% to 5%.[10] However, few studies have addressed whether or not reducing nicotine in cigarettes is a viable public health policy measure.

Two meetings were held (in 2007 and 2009) in the United States to revisit this topic.1 Using seminal articles [3, 11] as a starting point, these meetings sought to: 1) Present current science on topics related to nicotine reduction; 2) Based on this science, discuss the potential feasibility and approaches to reducing nicotine in tobacco products; and 3) Establish a research agenda to determine the viability and practicality of nicotine reduction as a policy measure. Topics for the scientific overview were determined by a steering committee. This paper provides an overview of the science on each of the topics presented at the meetings and an outline of the most pressing research questions proposed by participants. This paper is not intended to be a comprehensive review of the extant literature on all aspects of nicotine reduction. This paper is focused primarily on the contribution of nicotine per se to tobacco dependence and does not address other factors that may enhance the addiction potential or appeal of a tobacco product, such as cultural, social, physical and economic aspects associated with its use.

CURRENT SCIENCE

We review new science in two categories: a) human laboratory and clinical studies, and animal studies relevant to understanding the effects of a reduced nicotine policy, including studies that examine factors that may modulate nicotine reinforcement, and b) contributions of product design and non-nicotine constituents that may moderate the influence of low nicotine content on the addictiveness or abuse liability of a cigarette product.

Human research relevant to reduction in cigarette nicotine delivery

Reducing nicotine delivery in cigarettes would be aimed at preventing intake of nicotine that reaches the threshold dose for nicotine reinforcement (i.e., the level at which nicotine leads to persistent self-administration), which is believed essential for onset of addiction. The few human studies on reduced nicotine (or denicotinized) cigarettes [12-32,35-36,40] have examined: 1) positive and negative reinforcing effects of denicotinized cigarettes; 2) effects of reduced nicotine cigarettes on toxicant exposure and compensatory smoking (i.e. greater smoking in an effort to maintain a desired nicotine intake); and 3) use of reduced nicotine cigarettes as a cessation tool. In these studies, the nicotine delivery in most of the reduced nicotine or denicotinized cigarettes is relatively low because they contain smaller amounts of nicotine (5-10% of the yield of standard commercial brands) in the tobacco itself. (These are different from the widely-available, so-called “light” brands, which contain amounts of nicotine similar to standard brands but are engineered to dilute smoke intake with air.) There are also human studies describing the amount of smoking associated with onset of dependence symptoms. Although studies of the threshold for nicotine discrimination have been conducted, to date no systematic human study has examined the threshold dose for the development or maintenance of nicotine addiction nor directly examined the best approach for reducing levels of nicotine in cigarettes to maximize public health benefits.2

Laboratory and residential human studies

Laboratory studies show that denicotinized cigarettes produce acute subjective effects similar to those of nicotine cigarettes. For example, denicotinized cigarettes have been shown to reduce craving and negative affect due to withdrawal during short-term abstinence periods from usual brand cigarettes.[12-21] The acute withdrawal relieving effects are found not to be due to expectancies for nicotine or the simple motor aspects of smoking (e.g. handling), highlighting the importance of smoke inhalation per se.[22] Denicotinized cigarettes and standard nicotine cigarettes can produce similar self-reported liking and satisfaction in smokers [23, 24] although another study found results to the contrary [35], and can produce similar delays in the latency to smoke (i.e., the time to smoke a cigarette) or reductions in the amount of subsequent smoking of nicotine cigarettes.[17, 25] Denicotinized cigarettes may also be as acutely reinforcing as nicotine cigarettes in dependent smokers[26], suggesting that denicotinized cigarettes may serve as an effective short term substitute for nicotine-containing cigarettes when the latter are unavailable.[27-28]

Most of these studies were conducted in the laboratory and over a limited period of time (e.g. a few hours) and may not reflect outcomes observed over longer time periods. In an 11-day inpatient study, the reinforcing efficacy of denicotinized cigarettes decreased over time (as measured by number of cigarettes smoked and how hard subjects worked to earn puffs on the cigarette), but denicotinized cigarettes continued to effectively reduce craving from cessation of usual brand cigarettes [29], which is consistent with results from a similarly designed outpatient study.[36]

The responses observed with denicotinized cigarettes may be because non-nicotine sensory aspects have acquired reinforcing effects, non-nicotine constituents other than nicotine are reinforcing, or that low levels of nicotine are sufficient to maintain smoking behavior because these levels can produce effects of physiological significance, at least acutely. For example, recent brain imaging studies show that smoking a single very-low-nicotine cigarette results in significant (23%) occupancy of α4β2 nicotinic receptors, which are considered the primary receptor subtype mediating nicotine's reinforcing and other behavioral effects.[37] Thus, the reinforcing and mood effects of very-low-nicotine cigarettes may be attributable, in part, to nicotine's pharmacological effects. Other evidence also suggests that very low-level nicotine exposure may have important pharmacological effects. This includes in vitro studies showing that significant nicotinic receptor desensitization, a potential contributor to nicotine addiction,[38] can occur with nicotine doses below a threshold for activating receptors, which mediates nicotine's acute reinforcing effects.[38-39]

In summary, abrupt switching to denicotinized cigarettes does not appear to result in significant withdrawal symptoms and may maintain similar levels of smoking reward and reinforcement in the short-term. Over more extended exposure, the positive reinforcing properties of denicotinized cigarettes appear to decrease. Longer-term clinical studies, presented next, address the time course of these effects while smoking reduced nicotine cigarettes, as well as their impact while smokers are in the natural environment.

Longer-term clinical studies: Gradual nicotine reduction effects

One of the major concerns regarding reduced nicotine cigarettes is whether they would lead to “compensatory” (i.e. increased) smoking behavior to maintain level of nicotine intake, which may result in greater exposure to toxic smoke emissions. In preparation for a series of clinical studies, Benowitz et al.[30] examined the acute effects of cigarettes containing five levels of nicotine (1 to 12 mg) on plasma nicotine and blood carboxyhemoglobin concentrations in a within-subject, cross-over study. Systemic nicotine intake was related linearly to the nicotine content of the cigarettes, and compensation after smoking a single low-nicotine content cigarette was relatively small. Carbon monoxide exposure was similar across the nicotine doses, further supporting the idea that compensation was minimal. Similar results were found when CO boost of an even lower, 0.05 mg nicotine yield cigarette was compared with preferred brand.[35]

A subsequent clinical study [31] examined the effects of reduced nicotine content cigarettes on nicotine and carcinogen exposure when smokers were asked to smoke progressively lower nicotine content cigarettes. Over the weeks of nicotine content tapering (5 levels of nicotine from 12 mg to 1 mg, or 0.8mg to 0.1 mg nicotine based on machine determined yields; each level smoked for one week) plasma cotinine levels declined to 30% of that seen when subjects were smoking their usual cigarettes. There was little evidence of increased smoking (no change in number of cigarettes or CO), no change in cardiovascular risk factors (e.g., HDL cholesterol, c-reactive protein, fibrinogen, P-selectin), and a decrease in 4-(methlynitrosamino)-1-(3-pryridyl)-1-butanol and its glucuronides (or total NNAL), a metabolite of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK), a potent lung carcinogen. Despite the sharp reduction in nicotine intake, there was no increase in craving and no change in self-reported depression. However, there was an increase in self-reported irritability and eating. At 4 weeks after the end of the study (when subjects were allowed to return to their usual brand cigarettes or to quit), 25% of the subjects attained abstinence, a surprisingly high rate given that these smokers did not enroll in the study in order to quit.

In a similarly designed study [32], researchers examined commercial low yield cigarettes engineered to increase smoke dilution to provide progressively lower yields, as assessed by smoking machine testing (0.9 mg to 0.1 mg based on machine determined yields). Little change was seen in plasma cotinine concentration from 0.9 to the 0.4 mg nicotine yield cigarettes, suggesting compensation in smoking behavior. However, significant decreases in this measure and carcinogen exposure biomarker levels (NNAL and metabolites of some polycyclic aromatic hydrocarbons) were observed when smokers were switched to 0.2 mg and/or to 0.1 mg nicotine yield cigarettes, most likely because of the extensive filter ventilation of these “ultralow yield” cigarettes, too much to be overcome by compensation. The 0.1 mg nicotine yield cigarette also produced non-significantly greater withdrawal. Two of 20 subjects quit smoking 4 weeks after tapering to the lowest yield cigarette. This study also suggests that substantial reduction in the nicotine yield of cigarettes may not lead to compensation, may reduce toxicant exposure, and may facilitate abstinence, if nicotine yields are sufficiently low.

Longer term clinical trials: Reduced nicotine cigarettes as a cessation tool

Recent clinical trials support the idea that low nicotine cigarettes may be useful as an aid for cessation. One study compared 6 weeks of use of 0.3 mg and 0.05 mg nicotine yield cigarettes with medicinal nicotine (lozenge) as methods for cessation. Seven-day point prevalence success rates at the end of 12 weeks were 14% with 0.3 mg cigarette, 36% with 0.05 mg cigarette, and 20% with nicotine lozenge [p= 0.02;40]. Number of cigarettes per day and level of carbon monoxide (CO) increased with 0.3 mg cigarette, but decreased with 0.05 mg cigarette; yet, in both conditions, there was significant reduction in biomarkers of carcinogen exposure, presumably because these cigarettes contain low nitrosamine levels [33].

Another relevant area of research includes examining methods that may maximize extinction of smoking behavior by separating nicotine intake from cigarette use. To date, four clinical trials have examined combined use of nicotine patch with denicotinized cigarettes over brief periods (2 weeks or less for denicotinized cigarettes).[36, 41-43] Findings suggest that the use of nicotine patch with denicotinized cigarettes, compared to nicotine patch or denicotinized cigarettes alone, may lead to greater satiation and craving relief.[42] On the other hand, another study showed no differences between denicotinized cigarettes with or without the nicotine patch on cigarette satisfaction and liking, but a trend towards greater withdrawal relief.[36] This study also found addition of the patch reduced total puff volume, CO boost and number of denicotinized cigarettes smoked.[36] However, nicotine patch did not appear to facilitate the process of smoking extinction when observed under short term study conditions.[36]

These clinical studies suggest that reduced smoking behavior and increased cessation could occur with cigarettes of no more than 0.1 mg in nicotine yield. Addition of nicotine patch may promote smoking cessation, although the evidence is limited. Some study populations have consisted of smokers who wanted to quit, and quit motivation can strongly influence treatment effects.[44] In a 1940s study [45], “inveterate” smokers who were switched to a very low nicotine cigarette for about one month reported significant withdrawal symptoms. Therefore, research is needed on the impact of reduced nicotine cigarettes in a more representative population of smokers with varying levels of interest in quitting (i.e. the majority of smokers who would be confronted with a marketplace of low nicotine cigarettes).

Development of nicotine dependence

The dose of nicotine that will lead to extinction of smoking may not be the dose that is associated with the onset of dependence symptoms or nicotine addiction. Studies conducted with adolescent smokers [46-51] suggest that the potential threshold for onset of nicotine addiction is likely to be substantially lower than the 5 standard nicotine cigarettes per day suggested by earlier research.[3] Several cross-sectional and longitudinal studies have shown that youth smoking on a less than daily basis nevertheless report onset of dependence symptoms.[46-49] About half the youth smokers who reported one or more symptoms reflective of a loss of autonomy over smoking had smoked on average two cigarettes one day per week, and half of those who met ICD-10 defined dependence reported smoking 46 cigarettes a month, or 1-2 cigarettes per day.[50-51] That symptoms of dependence can develop with low rates of smoking is consistent with results from a small study of adults demonstrating about 50% occupancy of α4β2 nAChRs for 3 hours after just 1-2 puffs on a 1.2-1.4 mg nicotine yield cigarette.[52] Similarly, other prolonged brain effects (long-term potentiation of the excitatory transmission to the brain reward centers) have been observed after brief application of low concentrations of nicotine [0.5-1.0 μM; 53].

Both human and animal studies have shown that the adolescent brain is more vulnerable and sensitive to nicotine's effects.[51] For example, adult smokers who initiated smoking during adolescence exhibit greater cigarette consumption, lower likelihood of trying to quit, and increased risk of relapse compared to those who started smoking later in life.[54-56] Adolescent rats and mice might also be more sensitive than adults to the rewarding and reinforcing effects of nicotine, as indexed by greater conditioned place preference (CPP),[57-62] faster acquisition of nicotine self-administration (NSA), and higher baseline NSA rates compared to adults [63-66; but see, 67-69]. What remains unknown are the effects of low dose nicotine cigarettes in adolescents and whether there is a dose that reduces the probability of sustained cigarette use.

Animal research

Given the similarity across species in intravenous nicotine doses that are self-administered[70-71], animal studies may be useful for identifying a range of doses that may encompass the nicotine reinforcement threshold in humans. Numerous studies have shown that acquisition and maintenance of intravenous NSA in adult animals (e.g., rats, monkeys) is dependent upon unit dose, with the peak of the dose-response curve at unit doses of 0.01 to 0.03 mg/kg under small fixed-ratio (FR, fixed number of lever presses for a dose of nicotine) schedules [see 70 for review ]. While NSA in animals typically decreases at unit doses below 0.01 mg/kg, unit doses as low as 0.003 mg/kg have been shown to maintain NSA in rats above saline extinction levels when substituted for a higher training dose (e.g., 0.03 mg/kg), though variability between subjects is apparent [72-76; but see 77, 78]. No animal studies have specifically characterized the reinforcement threshold dose of nicotine during acquisition of NSA in adolescents or in the context of progressively reducing the unit nicotine dose during maintenance of NSA in adults.

Individual differences and environmental moderators

Animal or human data suggest that a variety of factors could modulate nicotine reinforcement thresholds. The same low dose of nicotine may or may not be reinforcing, depending on individual differences and environmental moderators of nicotine reinforcement (i.e. self-administration).

However, the focus of most research—differences between groups in their mean response to a fixed dose—is of uncertain utility in informing policy. Rather than a group's mean response at a particular low dose, the distribution of the minimum doses eliciting a response from the individuals within the group will be of greater relevance in identifying a nicotine dose below the threshold for nicotine reinforcement. Such a distribution identifies how many group members will be at risk at a given low dose below the group's mean threshold (e.g. if FDA set a maximum dose for cigarettes).

Although nicotine discrimination is not directly related to reinforcement, human studies of the threshold dose for nicotine discrimination provide an example of the potentially wide variability within groups of humans in reinforcement threshold doses. In a study involving nicotine nasal spray, the median threshold dose for discrimination from placebo was quite low and similar between 19 nonsmokers and 18 smokers, 0.002 mg/kg (approx. 0.14 mg for 70 kg human) and 0.003 mg/kg (approx. 0.21 mg/70 kg), respectively.[79] However, threshold doses in this study varied by over 100-fold within both smokers and nonsmokers, from 0.00013 mg/kg up to 0.020 mg/kg. Whether this within-group variability generalizes to nicotine via smoking would be important to determine. With these caveats in mind, the following brief overview of human research examines mean differences between groups in the reinforcing and rewarding effects of fixed nicotine doses.

Individual differences

Sex

A considerable body of research indicates that adult women smokers may be less sensitive than men to the reinforcing and rewarding effects of nicotine per se, but more sensitive to the reinforcing and rewarding effects of non-nicotine aspects of smoking, such as cues.[80] This sex difference has also been observed among adult nonsmokers [81] suggesting an innate sex difference prior to onset of dependence.

However, adolescent girls are more likely than boys to report experiencing nicotine dependence symptoms[46-47, 49, 51] and experiencing the onset of nicotine dependence symptoms faster[51], although another study showed no difference in latency to dependence symptoms.[50] These results suggest a greater, rather than less, sensitivity to tobacco use in girls compared to boys, although the relative contributions of nicotine versus non-nicotine factors cannot be disentangled in this research. Animal studies also show greater sensitivity to nicotine in female rats compared to male rats.[82-83] However, these studies involved nicotine delivery accompanied by cues, and consistent with adult human differences [80], female rats also appear more sensitive to conditioned reinforcement from cues associated with nicotine intake.[84]

Comorbid disorders

Because smoking prevalence is much higher in those with other psychiatric or substance abuse disorders, nicotine may be more reinforcing in smokers with such comorbidities than in smokers without such disorders. These disorders include schizophrenia, major depression, attention deficit hyperactivity disorder, and alcohol, cocaine, opiate, and stimulant dependence.[86] Little research has directly examined this notion, but Hughes et al.[87] found that nicotine via gum was more reinforcing in smokers who were former alcoholics compared to smokers who had never been alcoholic. However, chronic alcohol exposure may lead to tolerance to many effects of nicotine,[88] suggesting reduced sensitivity to those effects and perhaps to nicotine threshold dose.

Individual differences: nicotine pharmacokinetics

Human studies have shown a relationship between nicotine pharmacokinetics and smoking. Populations with slower nicotine metabolism smoke on average fewer cigarettes per day than those with more rapid elimination,[89] and greater smoking and lower quit rates are associated with faster nicotine metabolism.[90] This relationship can be demonstrated within subjects, as inhibition of nicotine metabolism in smokers by methoxsalen is accompanied by smoking reduction.[91] While faster metabolism is associated with greater dependence in adult smokers, slower metabolism may be associated with a higher likelihood of developing dependence during smoking initiation ([92] but see[93]). In the context of initiation, slower metabolism may be associated with greater vulnerability to addiction, perhaps because it results in prolonged presence of higher brain nicotine concentrations, producing a higher magnitude (or longer duration) of reinforcement. For the same reason, slower metabolism could also enhance the reinforcing effects of smoking cigarettes with low nicotine content in both adults and adolescents. Slow metabolism could therefore lower the nicotine reinforcement threshold in smoking initiation and maintenance.

Ethnic/racial groups

Smoking prevalence and daily smoking rate vary by ethnicity, as does nicotine kinetics,[94] suggesting some possible influence of ethnicity/race on nicotine reinforcement. Some populations demonstrate slower nicotine metabolism (e.g. Asian Americans), which may account for their lower smoking rate compared to populations with more rapid elimination.[89] Few differences in pharmacodynamic effects of nicotine as a function of ethnicity or racial identity have been shown, although little research has examined these differences.

Genetic factors

Genetic factors have been associated with risk of onset and persistence of nicotine dependence in clinical research,[95] but it is not clear that these associations are due to differences between genotypes in sensitivity to nicotine reinforcement, either prior to or after dependence onset. However, among nonsmokers in one study, nicotine choice (i.e. self-administration) via nasal spray was greater in those without the DRD4-7 allele compared to those with the 7 allele, but other dopamine-related genetic factors, including DRD2 and DAT, were not related to reinforcement.[96] Research with dependent smokers shows that presence of the G allele of OPRM1, the mu opioid receptor gene, is associated with reduced nicotine choice, but only in women and not men.[97] Together with the literature on genetic influences on nicotine metabolism,[90] the possible influence of genetic factors on nicotine reinforcement threshold dose remains likely, but more research is needed to clarify which factors are involved.

Other differences

Obese smokers may be less sensitive to nicotine reinforcement and reward,[98] suggesting that their threshold dose for nicotine reinforcement may be higher. Parental history of smoking is associated with greater risk of dependence in offspring, but greater sensitivity to nicotine reinforcement may not explain the association.[99] Impulsive personality factors are associated with greater risk of onset of dependence and may increase choice of nicotine nasal spray in nonsmokers, particularly in males,[100] suggesting that more impulsive individuals may be more sensitive to nicotine reinforcement threshold.

Environmental moderators

The reinforcing effects of nicotine are not an immutable property of the drug but may be acutely influenced by environmental factors. Environmental moderators of nicotine reinforcement may be numerous; only a few will be discussed.

Nicotine formulation (including expectancy and cues)

Although differences between formulations in nicotine pharmacokinetics play a key role In abuse liability and possibly threshold dose for reinforcement,[101] differences in expectancy for effects and other stimuli accompanying nicotine intake may also be involved. Smokers have expectancies for pleasurable effects from smoking but usually little expectancy of positive effects from less familiar formulations.[102] Expectancy for nicotine can speed the latency to smoke and increase reward, especially in women.[103] Similarly, smoking-associated stimuli or cues enhance nicotine reinforcement and reward from smoking.[80] This suggests that the threshold dose for reinforcement in dependent adults may be lower with smoking than with other forms of nicotine.

Stress or mood

Acute stress and negative mood commonly increase risk of relapse in quitting smokers, and laboratory studies show that smoking reinforcement is greater under such conditions,[104-105] perhaps suggesting enhanced sensitivity to nicotine reinforcement threshold dose. Similar effects of stress have been reported with animal NSA models.[106] Older research suggests small changes in nicotine pharmacokinetics due to stress,[107] but this effect is unlikely to have a substantial influence by itself on threshold dose for reinforcement. One study of dependent smokers found greater acute smoking reinforcement due to negative mood as a function of DRD4, DRD2 and DAT genotypes, but only DRD4 was associated with mood-induced increase in nicotine reinforcement per se.[108]

Acute alcohol intake and other drug use

Dependence on other drugs increases risk of smoking onset and persistence, and vice versa, but it is not clear that these associations are due to acute effects of those other drugs on sensitivity to nicotine reinforcement. Several, but not all, studies have shown increased acute smoking reinforcement following alcohol consumption [109] and stimulant use, such as methylphenidate[110] and cocaine.[111] Other research showed increased acute alcohol reinforcement following smoking, at least in men.[112] However, none of this research has clarified that the increases in smoking or other drug use are due to nicotine per se. Some animal studies have shown that caffeine and methylphenidate can increase NSA and nicotine discrimination in rats.[e.g., 113, 114]

Additional observations

In both the human and animal literature, there is a scarcity of data not only on the effects of reduced nicotine doses on smoking or nicotine intake but also on other responses. Even if a threshold reinforcing nicotine dose is identified and a non-addictive cigarette can be produced, it will be important to determine whether there are other adverse effects from the nicotine exposure that occurs in adolescents who nonetheless experiment with such cigarettes. The threshold for nicotine's reinforcing effects may be higher than the threshold for nicotine's other potentially adverse effects, including enhancing vulnerability to other drug use. In animal studies, very brief exposure to i.v. nicotine doses (only two infusions of 0.03 mg/kg daily for 4 days) in adolescent rats sensitized them to the reinforcing effects of cocaine, suggesting a potential adverse consequence of very limited nicotine exposure in adolescence.[115] This daily dose (0.06 mg/kg/day or 4.2 mg for a 70 kg smoker) is comparable in humans to the nicotine intake from 4 standard cigarettes. Therefore, low-level nicotine exposure in adolescents experimenting with cigarettes designed to prevent nicotine addiction could potentially produce risk of addiction to other drugs of abuse.

PRODUCT DESIGN AND CONSTITUENTS

Reducing dependence risk due to nicotine from tobacco use requires more than simply mandating lower amounts of nicotine in products. Although the addiction potential of a product is critically determined by its active pharmacological entities (e.g., nicotine), the real world risk of addiction is determined by many other aspects of the drug formulation (e.g., oral, nasal, smoked), which can influence the biological impact of a given drug dose. For example, oral smokeless tobacco products and cigars appear to carry a somewhat lower addiction risk than cigarettes,[116-118] and the risk of developing addiction to nicotine replacement medications appears to be very small [3, 5, 9, 119], even though the absolute nicotine delivery from these formulations may not differ much.

A variety of physical parameters, including amount of tobacco, its nicotine content, length and circumference of the tobacco column, filter length, filter ventilation, filter composition, and other ingredients, influence delivery of nicotine, nicotine-free dry particulate matter (NFDPM, commonly known as tar) and various gases.[6-7, 120] A wide range of chemical parameters (such as moisture content of tobacco, specific tobacco composition parameters, specific additives in paper and tobacco) change the response of tobacco to heating under combustion or pyrolysis conditions. Interactions of physical and chemical characteristics of cigarettes may also change the size distribution of aerosol particles that determine deposition and absorption of nicotine and other constituents.[7]

The complexity of this issue is illustrated by a study that described how physical parameter differences of cigarette (e.g., non-filter cigarettes, cellulose acetate filter cigarette with and or without perforation, cellulose acetate filter cigarette with perforation and highly porous paper), despite the same tobacco composition, can dramatically alter the amount of machine-determined yields of nicotine and various other toxicants.[121] Formaldehyde varied among cigarettes from 21 μg to 36 μg, phenol from 62 μg to 161 μg, and acetaldehyde from 550 μg to 1290 μg. Nicotine delivery varied from 0.94 mg/cigarette to 1.5 mg/cigarette within the filter types. Changes in filtration, however, made very little difference for other toxicants such as the polycyclic aromatic hydrocarbons (e.g., benz(a)anthracene, benzo(a)pyrene), showing how limited and variable filtration can be in reducing some constituents.

Constituents within tobacco products also augment levels of free base nicotine and thereby may contribute to tobacco's reinforcing effects. For example, ammonia, diammonium phosphate, or urea have been identified in tobacco industry documents as capable of enhancing the bioavailability of nicotine by delivering increased levels of free-base nicotine in the smoke.[122-123] Pankow et al.[124-125] described changes that occur in the balance between nicotine in the particulate phase and the gas phase as chemical additives alter the pH of smoke. Since the rate of transfer of nicotine from product to smoke and from smoke to the nicotine-receptors in the back of the throat and lungs is driven by the fraction of free nicotine, addition of chemicals or design features altering either the pH of emissions or enhancing delivery from tobacco products might also contribute to the cigarette's high addiction potential.[34, 126]

Some of the more than 4,000 tobacco smoke constituents other than nicotine may also contribute to engendering and maintaining smoking behavior. For example, nornicotine, a minor alkaloid constituent of tobacco and a brain metabolite of nicotine,[127-128] accumulates in rat brain with repeated nicotine administration and exhibits a brain half-life three-times longer than nicotine,[129-130] evokes dopamine release in rat striatum in a mecamylamine-dependent manner,[131] elicits discriminative stimulus (i.e., subjective) effects similar to nicotine,[132] and produces reinforcing effects in its own right [i.e., is self-administered, 133].

Anabasine, another minor tobacco alkaloid, produces nicotine-like discriminative stimulus effects,[134-136] and the combination of nicotine with a smoke-relevant cocktail of minor alkaloids (anabasine, nornicotine, anatabine, cotinine, and myosmine) is more reinforcing than nicotine alone.[137] It is likely that other compounds in cigarette smoke contribute to addiction risk. For example, acetaldehyde, a combustion product, increases the firing rate of dopamine neurons in the ventral tegmental area,[138] potentiates the locomotor effects of nicotine,[139] is self administered in animals,[140-142] and increases acquisition of nicotine self-administration (NSA) in adolescent rats at unit nicotine doses that alone are not self-administered.[57]

Monoamine oxidase inhibitors (MAO) inhibitors, such as harman, are present in tobacco and tobacco smoke,[143-144] and the brains of smokers show 28% lower MAO-A and 40% lower MAO-B activity in PET-imaging studies.[145] Harman is also a condensation product of acetaldehyde.[143] MAO catalyses intraneuronal deamination of biogenic amines, such as dopamine, serotonin and noradrenaline. Consequently, MAO inhibition increases extracellular brain levels of these amines and may potentiate effects of nicotine, since nicotine stimulates amine release. Studies in rats show that the nonspecific MAO inhibitor tranylcypromine (TNCP) or various MAO-A selective drugs enhance nicotine-induced dopamine release in the nucleus accumbens,[146] locomotor sensitization to nicotine,[147] nicotine discrimination,[148] and nicotine self administration.[85, 146, 149] These findings provide evidence that non-nicotine tobacco constituents could play a role in moderating nicotine reinforcement thresholds.

In summary, product design, pH, and constituents other than nicotine may play a significant role in addictiveness of cigarettes.[150-151] Additives, such as menthol may also increase reinforcing efficacy by making tobacco more palatable, enhancing taste and smoothness, and facilitating self-administration.[152-153]

RESEARCH RECOMMENDATIONS

Based on the existing science, it appears reasonable to propose that a threshold dose for nicotine reinforcement will eventually be identified such that cigarettes containing doses below this threshold should not produce dependence in the majority of individuals. Identifying this threshold will require different lines of research, including assessment of smoking reinforcement in adult smokers as well as naïve adolescents as a function of lowering cigarette nicotine dose. Studies of nicotine threshold dose with naïve adolescents, however, will be particularly challenging and perhaps should be studied primarily in animal models.

The best approach to reduce nicotine in cigarettes is not certain, but two alternatives may be considered: 1) Over time, a mandated, stepped reduction of nicotine content in combustible products; or 2) Immediate, significant reduction of nicotine content in cigarettes to a dose where no significant compensatory smoking behavior would occur. The primary outcomes to be measured in clinical research would be: a) number of quit attempts; b) sustained cessation of tobacco use; c) toxicant exposure; and d) drop-out rate and other non-compliance measures, including uptake of other products. Attendant to this general research question are a number of specific research questions, listed in Table 1.

Table 1.

Nicotine Reduction Research Questions

Short-Term Needs
• What is the nicotine threshold dose(s) for addiction and the distribution of thresholds across subjects? What are the moderating influences?
• What are the effects of reduced nicotine cigarettes on the brain, both in adult smokers and in adolescents experimenting with these cigarettes? Are there changes in the receptor number or binding potential and do they change over time? Does receptor sensitization change over time?
• What is the extent of compensatory smoking across the two nicotine reduction approaches (immediate vs. gradual reductions in nicotine content) and what interventions can be used to minimize compensatory smoking, such as making nicotine available through less hazardous delivery systems (e.g, NRT) or through non-nicotine pharmacotherapies?
• What are the effects of reduced nicotine cigarettes in subpopulations (consumers who smoke for self-medication purposes such as those with co-morbidity or who are severely addicted to tobacco products) and how can negative consequences be mitigated?
• What product contents and design features affect product addictiveness (such as the identification and balance of acids and bases, the relationship between smoke chemistry and aerosol physics and respiratory tract deposition)? Should we be focusing more attention beyond nicotine to reduce addictiveness? What would be the effect be of merely banning ventilated filters or specific additives in cigarettes?
Long-Term Needs
• What would be the public's reaction to and perception of a marketplace containing only reduced nicotine cigarettes? How could we frame the message and educate the public about reduced nicotine cigarettes to produce the greatest public health benefit?
• What are the potential unintended consequences from reduced nicotine cigarettes, how can they be determined and monitored, and what needs to be done to mitigate against negative consequences?

Unintended consequences may be particularly difficult to study.[11, 154] Among potential concerns are: 1) A switch to other drugs of abuse, particularly among populations smoking for social reinforcement, self-identity, or self-medication purposes; 2) Dual use of tobacco products, such as reduced nicotine cigarettes with oral tobacco or small cigars, which may lead to greater exposure to toxicants, especially if these other tobacco products continue to contain higher nicotine levels; 3) Use of reduced nicotine cigarettes as starter products. Just as low free-base nicotine smokeless tobacco products served as starter products for higher nicotine and more toxic smokeless tobacco products,[155] these reduced nicotine cigarettes may lead to the use of other tobacco products with higher levels of nicotine, unless these other products also contain low nicotine levels; 4) Illicit cigarette marketing and smuggling[156] including through the internet and through territories that do not require reduced nicotine content of cigarettes; 5) Product tampering or manipulation (such as adding nicotine to the product); and 6) Industry manipulations (e.g., nicotine analogues, companion products to increase nicotinic effects).

Conclusions

Developing practical, scientifically-supported recommendations about nicotine levels in tobacco products involves filling gaps in knowledge in diverse areas including tobacco product design, content, and emissions; biomarkers of exposure; addiction; sensory perception; motivational factors; withdrawal and craving; animal studies; human clinical research; genetics; physiology, pharmacokinetics and metabolism; population studies; economics; and communications and messaging. Consequently, an organized and multidisciplinary effort should be established to set priorities and goals (the desired end results); engage appropriate scientific, research, and government communities/organizations; shape the direction of research; and ensure that efforts stay focused on the ultimate goal of understanding how nicotine reduction could impact the morbidity and mortality of tobacco use. Access to tobacco industry knowledge and research conducted in the areas of nicotine manipulation, nicotine analogues and abuse liability testing of tobacco products could advance the understanding of this issue.

If nicotine reduction is enacted, then large-scale surveillance is needed in order to understand the population-level impact of such changes. Marketplace monitoring and assessing unintended consequences (smuggling, nicotine spiking, new product introductions, etc) will also involve broad surveys.

We are in an unprecedented time in tobacco control where the possibilities of reducing tobacco-related morbidity and mortality by reducing nicotine in tobacco products could be a reality. Because this measure has the potential to have a profound impact on reducing smoking prevalence and thereby smoking-related death and disease, the sooner we are able to address the research questions described in this article, the sooner we can reduce the number of lives lost to tobacco.

WHAT THIS PAPER ADDS.

This paper provides a scientific exploration of the issue of reduction of nicotine content in cigarettes on tobacco addiction. This topic is of particular significance because of the Family Smoking Prevention and Tobacco Control Act, providing the U.S. Food and Drug Administration jurisdiction over the regulation of tobacco products, and Articles 9-11 of the World Health Organization Framework Convention on Tobacco Control. An updated overview of the scientific literature on reduced nicotine cigarettes is provided. In addition, recommendations for future directions and research agenda from tobacco control researchers, policymakers and representatives of governmental agencies developed at meetings convened in the U.S. are described.

ACKNOWLEGEMENTS

Funded by National Cancer Institute, National Institute on Drug Abuse, American Legacy Foundation and University of Minnesota Transdisciplinary Tobacco Use Research Center, DA/CA P50-013333.

Footnotes

1

Meetings were held in November 2007 and January 2009 with experts from different scientific disciplines, tobacco control policy advocates, and representatives of different federal government agencies (see Appendix 1). The meetings were sponsored by the National Cancer Institute's Tobacco Harm Reduction Network and the University of Minnesota Transdisciplinary Tobacco Use Research Center. The meetings were funded by the National Cancer Institute, National Institute on Drug Abuse and the American Legacy Foundation.

2

The tobacco industry itself conducted extensive research to determine the threshold for targeting nicotine dosing by cigarettes, concluding, for example that “to lower nicotine too much might end up destroying the nicotine habit in a large number of consumers and prevent it from ever being acquired by new smokers.” (U.S. Dept. Justice Ex 20,112, British American Tobacco Company, 1959, cf [34] However, the tobacco industry research appeared more focused on establishing the threshold dose that would r eadily sustain addiction and not necessarily the dose, below which addiction would be unlikely to occur.

OTHER DECLARATIONS

Dorothy Hatsukami has received grant funding from Nabi Biopharmaceuticals to conduct nicotine vaccine clinical trials. Jack Henningfield provides consulting support for GlaxoSmithKline Consumer Health through Pinney Associates on an exclusive basis on issues related to tobacco dependence treatment, has financial interest in a potential new oral nicotine replacement product, and serves as an expert witness in litigation against tobacco companies. Neal Benowitz serves as a consultant for Pfizer and as an expert witness in litigation against tobacco companies. Mitch Zeller provides consulting support to GlaxoSmithKline Consumer Health through Pinney Associates on an exclusive basis on issues related to tobacco dependence treatment. Kenneth Perkins has served as a consultant to Cypress Bioscience.

The Corresponding Author has the right to grant on behalf of all authors and does grant on behalf of all authors, an exclusive license (or non exclusive for government employees) on a worldwide basis to the BMJ Publishing Group Ltd and its Licensees to permit this article (if accepted) to be published in Journal (insert name) editions and any other BMJPGL products to exploit all subsidiary rights, as set out in our license.

REFERENCES

  • 1.U.S. Congress . In: Family Smoking Prevention and Tobacco Control and Federal Retirement Reform. Congress US, editor. U. S. Government Printing Office; Washington, D.C.: 2009. Public Law 111-31 - June 22, 2009. [Google Scholar]
  • 2.World Health Organization The World Health Organization Framework Convention on Tobacco Control. 2003 www.who.int/tobacco/framework/final_text/en/print.html.
  • 3.Benowitz NL, Henningfield JE. Establishing a nicotine threshold for addiction. The implications for tobacco regulation. N Engl J Med. 1994;331(2):123–5. doi: 10.1056/NEJM199407143310212. [DOI] [PubMed] [Google Scholar]
  • 4.Redhead CS, Burrows VK. FDA Tobacco Regulation: The Family Smoking Prevention and Tobacco Control Act of 2009. CRS Report for Congress. Congressional Research Service R40475, May 28, 7-5700. 2009 [Google Scholar]
  • 5.U.S. Department of Health & Human Services . A report of the Surgeon General. U.S. Department of Health & Human Services; 1988. The health consequences of smoking: nicotine and addiction. [Google Scholar]
  • 6.Food and Drug Administration Regulations restricting the sale and distribution of cigarettes and smokeless tobacco products to protect children and adolescents; proposed reule analysis regarding FDA's jurisdiction over nicotine-containing cigarettes and smokeless tobacco products; notice. Federal Register. 1995;60:41314–792. [Google Scholar]
  • 7.Food and Drug Administration Regulations restricting the sale and distribution of cigarettes and smokeless tobacco to protect children and adolescents; final rule. Federal Register. 1996;61:44396–5318. [PubMed] [Google Scholar]
  • 8.World Health Organization . Advancing knowledge on regulating tobacco products (monograph) World Health Organization; Geneva, Switzerland: 2001. [Google Scholar]
  • 9.Royal College of Physicians of London . A report of the Tobacco Advisory Group. Royal College of Physicians of London; London: 2000. Nicotine Addiction in Britain. [Google Scholar]
  • 10.Tengs TO, Ahmad S, Savage JM, et al. The AMA proposal to mandate nicotine reduction in cigarettes: a simulation of the population health impacts. Prev Med. 2005;40(2):170–80. doi: 10.1016/j.ypmed.2004.05.017. [DOI] [PubMed] [Google Scholar]
  • 11.Henningfield JE, Benowitz NL, Slade J, et al. Reducing the addictiveness of cigarettes. Council on Scientific Affairs, American Medical Association. Tobacco Control. 1998;7(3):281–93. doi: 10.1136/tc.7.3.281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Baldinger B, Hasenfratz M, Battig K. Effects of smoking abstinence and nicotine abstinence on heart rate, activity and cigarette craving under field conditions. Human Psychopharmacology: Clinical and Experimental. 1995;10(2):127–36. [Google Scholar]
  • 13.Brauer LH, Behm FM, Lane JD, et al. Individual differences in smoking reward from de-nicotinized cigarettes. Nicotine Tob Res. 2001;3(2):101–9. doi: 10.1080/14622200123249. [DOI] [PubMed] [Google Scholar]
  • 14.Breland AB, Buchhalter AR, Evans SE, et al. Evaluating acute effects of potential reduced-exposure products for smokers: clinical laboratory methodology. Nicotine Tob Res. 2002;4(Suppl 2):S131–40. doi: 10.1080/1462220021000032780. [DOI] [PubMed] [Google Scholar]
  • 15.Buchhalter AR, Schrinel L, Eissenberg T. Withdrawal-suppressing effects of a novel smoking system: comparison with own brand, not own brand, and denicotinized cigarettes. Nicotine Tob Res. 2001;3(2):111–8. doi: 10.1080/14622200110042636. [DOI] [PubMed] [Google Scholar]
  • 16.Butschky MF, Bailey D, Henningfield JE, et al. Smoking without nicotine delivery decreases withdrawal in 12-hour abstinent smokers. Pharmacol Biochem Behav. 1995;50(1):91–6. doi: 10.1016/0091-3057(94)00269-o. [DOI] [PubMed] [Google Scholar]
  • 17.Dallery J, Houtsmuller EJ, Pickworth WB, et al. Effects of cigarette nicotine content and smoking pace on subsequent craving and smoking. Psychopharmacology (Berl) 2003;165(2):172–80. doi: 10.1007/s00213-002-1242-8. [DOI] [PubMed] [Google Scholar]
  • 18.Eid NC, Fant RV, Moolchan ET, et al. Placebo cigarettes in a spaced smoking paradigm. Pharmacol Biochem Behav. 2005;81(1):158–64. doi: 10.1016/j.pbb.2005.03.007. [DOI] [PubMed] [Google Scholar]
  • 19.Gross J, Lee J, Stitzer ML. Nicotine-containing versus de-nicotinized cigarettes: effects on craving and withdrawal. Pharmacol Biochem Behav. 1997;57(1-2):159–65. doi: 10.1016/s0091-3057(96)00309-7. [DOI] [PubMed] [Google Scholar]
  • 20.Rose J, Behm FM, Westman EC, et al. Pharmacologic and sensorimotor components of satiation in cigarette smoking. Pharmacol Biochem Behav. 2003;76(2):243–50. doi: 10.1016/j.pbb.2003.07.002. [DOI] [PubMed] [Google Scholar]
  • 21.Rose J, Behm FM, Westman EC, et al. Dissociating nicotine and nonnicotine components of cigarette smoking. Pharmacol Biochem Behav. 2000;67(1):71–81. doi: 10.1016/s0091-3057(00)00301-4. [DOI] [PubMed] [Google Scholar]
  • 22.Perkins KA, Ciccocioppo M, Conklin CA, et al. Mood influences on acute smoking responses are independent of nicotine intake and dose expectancy. J Abnorm Psychol. 2008;117(1):79–93. doi: 10.1037/0021-843X.117.1.79. [DOI] [PubMed] [Google Scholar]
  • 23.Westman EC, Behm FM, Rose JE. Dissociating the nicotine and airway sensory effects of smoking. Pharmacology, Biochemistry & Behavior. 1996;53(2):309–15. doi: 10.1016/0091-3057(95)02027-6. [DOI] [PubMed] [Google Scholar]
  • 24.Westman EC, Levin ED, Rose JE. Smoking while wearing the nicotine patch: Is smoking satisfying or harmful? Clinical Research. 1992;40:871A. [Google Scholar]
  • 25.Pickworth WB, Fant RV, Nelson RA, et al. Pharmacodynamic effects of new de-nicotinized cigarettes. Nicotine Tob Res. 1999;1(4):357–64. doi: 10.1080/14622299050011491. [DOI] [PubMed] [Google Scholar]
  • 26.Rusted JM, Mackee A, Williams R, et al. Deprivation state but not nicotine content of the cigarette affects responding by smokers on a progressive ratio task. Psychopharmacology (Berl) 1998;140(4):411–7. doi: 10.1007/s002130050783. [DOI] [PubMed] [Google Scholar]
  • 27.Shahan TA, Bickel WK, Badger GJ, et al. Sensitivity of nicotine-containing and de-nicotinized cigarette consumption to alternative non-drug reinforcement: a behavioral economic analysis. Behav Pharmacol. 2001;12(4):277–84. doi: 10.1097/00008877-200107000-00006. [DOI] [PubMed] [Google Scholar]
  • 28.Shahan TA, Bickel WK, Madden GJ, et al. Comparing the reinforcing efficacy of nicotine containing and de-nicotinized cigarettes: a behavioral economic analysis. Psychopharmacology (Berl) 1999;147(2):210–6. doi: 10.1007/s002130051162. [DOI] [PubMed] [Google Scholar]
  • 29.Donny EC, Houtsmuller E, Stitzer ML. Smoking in the absence of nicotine: behavioral, subjective and physiological effects over 11 days. Addiction. 2007;102(2):324–34. doi: 10.1111/j.1360-0443.2006.01670.x. [DOI] [PubMed] [Google Scholar]
  • 30.Benowitz NL, Jacob P, 3rd, Herrera B. Nicotine intake and dose response when smoking reduced-nicotine content cigarettes. Clin Pharmacol Ther. 2006;80(6):703–14. doi: 10.1016/j.clpt.2006.09.007. [DOI] [PubMed] [Google Scholar]
  • 31.Benowitz NL, Hall SM, Stewart S, et al. Nicotine and carcinogen exposure with smoking of progressively reduced nicotine content cigarette. Cancer Epidemiol Biomarkers Prev. 2007;16(11):2479–85. doi: 10.1158/1055-9965.EPI-07-0393. [DOI] [PubMed] [Google Scholar]
  • 32.Benowitz NL, Dains KM, Hall SM, et al. Progressive commercial cigarette yield reduction: biochemical exposure and behavioral assessment. Cancer Epidemiol Biomarkers Prev. 2009;18(3):876–83. doi: 10.1158/1055-9965.EPI-08-0731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Stepanov I, Jensen J, Hatsukami D, et al. Tobacco-specific nitrosamines in new tobacco products. Nicotine Tob Res. 2006;8(2):309–13. doi: 10.1080/14622200500490151. [DOI] [PubMed] [Google Scholar]
  • 34.Henningfield JE, Pankow J, Garrett B. Ammonia and other chemical base tobacco additives and cigarette nicotine delivery: issues and research needs. Nicotine Tob Res. 2004;6(2):199–205. doi: 10.1080/1462220042000202472. [DOI] [PubMed] [Google Scholar]
  • 35.Strasser AA, Lerman C, Sanborn PM, et al. New lower nicotine cigarettes can produce compensatory smoking and increased carbon monoxide exposure. Drug Alcohol Depend. 2007;86(2-3):294–300. doi: 10.1016/j.drugalcdep.2006.06.017. [DOI] [PubMed] [Google Scholar]
  • 36.Donny EC, Jones M. Prolonged exposure to denicotinized cigarettes with or without transdermal nicotine. Drug Alcohol Depend. 2009;104(1-2):23–33. doi: 10.1016/j.drugalcdep.2009.01.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Brody AL, Mandelkern MA, Costello MR, et al. Brain nicotinic acetylcholine receptor occupancy: effect of smoking a denicotinized cigarette. Int J Neuropsychopharmacol. 2009;12(3):305–16. doi: 10.1017/S146114570800922X. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Picciotto MR, Addy NA, Mineur YS, et al. It is not “either/or”: activation and desensitization of nicotinic acetylcholine receptors both contribute to behaviors related to nicotine addiction and mood. Prog Neurobiol. 2008;84(4):329–42. doi: 10.1016/j.pneurobio.2007.12.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Lu Y, Marks MJ, Collins AC. Desensitization of nicotinic agonist-induced [3H]gamma-aminobutyric acid release from mouse brain synaptosomes is produced by subactivating concentrations of agonists. J Pharmacol Exp Ther. 1999;291(3):1127–34. [PubMed] [Google Scholar]
  • 40.Hatsukami D, Kotlyar M, Hertsgaard LA, et al. Reduced nicotine content cigarettes: effects on toxicant exposure, dependence and cessation. Addiction. 2010;105:343–55. doi: 10.1111/j.1360-0443.2009.02780.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Becker KM, Rose JE, Albino AP. A randomized trial of nicotine replacement therapy in combination with reduced-nicotine cigarettes for smoking cessation. Nicotine Tob Res. 2008;10(7):1139–48. doi: 10.1080/14622200802123294. [DOI] [PubMed] [Google Scholar]
  • 42.Rezaishiraz H, Hyland A, Mahoney MC, et al. Treating smokers before the quit date: Can nicotine patches and denicotinized cigarettes reduce cravings? Nicotine Tob Res. 2007;9(11):1139–46. doi: 10.1080/14622200701684172. [DOI] [PubMed] [Google Scholar]
  • 43.Rose J, Behm FM, Westman EC, et al. Precessation treatment with nicotine skin patch facilitates smoking cessation. Nicotine & Tobacco Research. 2006;8(1):89–101. doi: 10.1080/14622200500431866. [DOI] [PubMed] [Google Scholar]
  • 44.Perkins KA, Stitzer M, Lerman C. Medication screening for smoking cessation: a proposal for new methodologies. Psychopharmacology (Berl) 2006;184(3-4):628–36. doi: 10.1007/s00213-005-0105-5. [DOI] [PubMed] [Google Scholar]
  • 45.Finnegan JK, Larson PS, Haag HB. The Role of Nicotine in the Cigarette Habit. Science. 1945;102(2639):94–96. doi: 10.1126/science.102.2639.94. [DOI] [PubMed] [Google Scholar]
  • 46.O'Loughlin J, DiFranza J, Tyndale RF, et al. Nicotine-dependence symptoms are associated with smoking frequency in adolescents. Am J Prev Med. 2003;25(3):219–25. doi: 10.1016/s0749-3797(03)00198-3. [DOI] [PubMed] [Google Scholar]
  • 47.Gervais A, O'Loughlin J, Meshefedjian G, et al. Milestones in the natural course of onset of cigarette use among adolescents. CMAJ. 2006;175(3):255–61. doi: 10.1503/cmaj.051235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kandel DB, Hu MC, Griesler PC, et al. On the development of nicotine dependence in adolescence. Drug Alcohol Depend. 2007;91(1):26–39. doi: 10.1016/j.drugalcdep.2007.04.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Caraballo RS, Novak SP, Asman K. Linking quantity and frequency profiles of cigarette smoking to the presence of nicotine dependence symptoms among adolescent smokers: findings from the 2004 National Youth Tobacco Survey. Nicotine Tob Res. 2009;11(1):49–57. doi: 10.1093/ntr/ntn008. [DOI] [PubMed] [Google Scholar]
  • 50.DiFranza JR, Savageau JA, Fletcher K, et al. Symptoms of tobacco dependence after brief intermittent use: the Development and Assessment of Nicotine Dependence in Youth-2 study. Arch Pediatr Adolesc Med. 2007b;161(7):704–10. doi: 10.1001/archpedi.161.7.704. [DOI] [PubMed] [Google Scholar]
  • 51.DiFranza JR, Savageau JA, Rigotti NA, et al. Development of symptoms of tobacco dependence in youths: 30 month follow up data from the DANDY study. Tob Control. 2002b;11(3):228–35. doi: 10.1136/tc.11.3.228. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Brody AL, Mandelkern MA, London ED, et al. Cigarette smoking saturates brain alpha 4 beta 2 nicotinic acetylcholine receptors. Arch Gen Psychiatry. 2006;63(8):907–15. doi: 10.1001/archpsyc.63.8.907. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mansvelder HD, McGehee DS. Long-term potentiation of excitatory inputs to brain reward areas by nicotine. Neuron. 2000;27(2):349–57. doi: 10.1016/s0896-6273(00)00042-8. [DOI] [PubMed] [Google Scholar]
  • 54.Breslau N, Peterson EL. Smoking cessation in young adults: age at initiation of cigarette smoking and other suspected influences. Am J Public Health. 1996;86(2):214–20. doi: 10.2105/ajph.86.2.214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Chen J, Millar WJ. Age of smoking initiation: implications for quitting. Health Rep. 1998;9(4):39–46(Eng). 39–48(Fre). [PubMed] [Google Scholar]
  • 56.Cui Y, Wen W, Moriarty CJ, et al. Risk factors and their effects on the dynamic process of smoking relapse among veteran smokers. Behav Res Ther. 2006;44(7):967–81. doi: 10.1016/j.brat.2005.07.006. [DOI] [PubMed] [Google Scholar]
  • 57.Belluzzi JD, Lee AG, Oliff HS, et al. Age-dependent effects of nicotine on locomotor activity and conditioned place preference in rats. Psychopharmacology (Berl) 2004;174(3):389–95. doi: 10.1007/s00213-003-1758-6. [DOI] [PubMed] [Google Scholar]
  • 58.Brielmaier JM, McDonald CG, Smith RF. Immediate and long-term behavioral effects of a single nicotine injection in adolescent and adult rats. Neurotoxicol Teratol. 2007;29(1):74–80. doi: 10.1016/j.ntt.2006.09.023. [DOI] [PubMed] [Google Scholar]
  • 59.Kota D, Martin BR, Damaj MI. Age-dependent differences in nicotine reward and withdrawal in female mice. Psychopharmacology (Berl) 2008;198(2):201–10. doi: 10.1007/s00213-008-1117-8. [DOI] [PubMed] [Google Scholar]
  • 60.Kota D, Martin BR, Robinson SE, et al. Nicotine dependence and reward differ between adolescent and adult male mice. J Pharmacol Exp Ther. 2007;322(1):399–407. doi: 10.1124/jpet.107.121616. [DOI] [PubMed] [Google Scholar]
  • 61.Shram MJ, Funk D, Li Z, et al. Periadolescent and adult rats respond differently in tests measuring the rewarding and aversive effects of nicotine. Psychopharmacology (Berl) 2006;186(2):201–8. doi: 10.1007/s00213-006-0373-8. [DOI] [PubMed] [Google Scholar]
  • 62.Vastola BJ, Douglas LA, Varlinskaya EI, et al. Nicotine-induced conditioned place preference in adolescent and adult rats. Physiol Behav. 2002;77(1):107–14. doi: 10.1016/s0031-9384(02)00818-1. [DOI] [PubMed] [Google Scholar]
  • 63.Adriani W, Macri S, Pacifici R, et al. Peculiar vulnerability to nicotine oral self-administration in mice during early adolescence. Neuropsychopharmacology. 2002;27(2):212–24. doi: 10.1016/S0893-133X(02)00295-6. [DOI] [PubMed] [Google Scholar]
  • 64.Chen H, Matta SG, Sharp BM. Acquisition of nicotine self-administration in adolescent rats given prolonged access to the drug. Neuropsychopharmacology. 2007;32(3):700–9. doi: 10.1038/sj.npp.1301135. [DOI] [PubMed] [Google Scholar]
  • 65.Levin ED, Lawrence SS, Petro A, et al. Adolescent vs. adult-onset nicotine self-administration in male rats: duration of effect and differential nicotinic receptor correlates. Neurotoxicol Teratol. 2007;29(4):458–65. doi: 10.1016/j.ntt.2007.02.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Levin ED, Rezvani AH, Montoya D, et al. Adolescent-onset nicotine self-administration modeled in female rats. Psychopharmacology (Berl) 2003;169(2):141–9. doi: 10.1007/s00213-003-1486-y. [DOI] [PubMed] [Google Scholar]
  • 67.Shram MJ, Funk D, Li Z, et al. Nicotine self-administration, extinction responding and reinstatement in adolescent and adult male rats: evidence against a biological vulnerability to nicotine addiction during adolescence. Neuropsychopharmacology. 2008b;33(4):739–48. doi: 10.1038/sj.npp.1301454. [DOI] [PubMed] [Google Scholar]
  • 68.Shram MJ, Li Z, Le AD. Age differences in the spontaneous acquisition of nicotine self-administration in male Wistar and Long-Evans rats. Psychopharmacology (Berl) 2008c;197(1):45–58. doi: 10.1007/s00213-007-1003-9. [DOI] [PubMed] [Google Scholar]
  • 69.Shram MJ, Siu EC, Li Z, et al. Interactions between age and the aversive effects of nicotine withdrawal under mecamylamine-precipitated and spontaneous conditions in male Wistar rats. Psychopharmacology (Berl) 2008a;198(2):181–90. doi: 10.1007/s00213-008-1115-x. [DOI] [PubMed] [Google Scholar]
  • 70.Matta SG, Balfour DJ, Benowitz NL, et al. Guidelines on nicotine dose selection for in vivo research. Psychopharmacology (Berl) 2007;190(3):269–319. doi: 10.1007/s00213-006-0441-0. [DOI] [PubMed] [Google Scholar]
  • 71.Harvey DM, Yasar S, Heishman SJ, et al. Nicotine serves as an effective reinforcer of intravenous drug-taking behavior in human cigarette smokers. Psychopharmacology (Berl) 2004;175(2):134–42. doi: 10.1007/s00213-004-1818-6. [DOI] [PubMed] [Google Scholar]
  • 72.Shoaib M, Schindler CW, Goldberg SR. Nicotine self-administration in rats: strain and nicotine pre-exposure effects on acquisition. Psychopharmacology (Berl) 1997;129(1):35–43. doi: 10.1007/s002130050159. [DOI] [PubMed] [Google Scholar]
  • 73.Watkins SS, Epping-Jordan MP, Koob GF, et al. Blockade of nicotine self-administration with nicotinic antagonists in rats. Pharmacol Biochem Behav. 1999;62(4):743–51. doi: 10.1016/s0091-3057(98)00226-3. [DOI] [PubMed] [Google Scholar]
  • 74.Brower VG, Fu Y, Matta SG, et al. Rat strain differences in nicotine self-administration using an unlimited access paradigm. Brain Res. 2002;930(1-2):12–20. doi: 10.1016/s0006-8993(01)03375-3. [DOI] [PubMed] [Google Scholar]
  • 75.Corrigall WA, Coen KM. Nicotine maintains robust self-administration in rats on a limited-access schedule. Psychopharmacology (Berl) 1989;99(4):473–8. doi: 10.1007/BF00589894. [DOI] [PubMed] [Google Scholar]
  • 76.Cox BM, Goldstein A, Nelson WT. Nicotine self-administration in rats. Br J Pharmacol. 1984;83(1):49–55. doi: 10.1111/j.1476-5381.1984.tb10118.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.DeNoble VJ, Mele PC. Intravenous nicotine self-administration in rats: effects of mecamylamine, hexamethonium and naloxone. Psychopharmacology (Berl) 2006;184(3-4):266–72. doi: 10.1007/s00213-005-0054-z. [DOI] [PubMed] [Google Scholar]
  • 78.Donny EC, Caggiula AR, Knopf S, et al. Nicotine self-administration in rats. Psychopharmacology (Berl) 1995;122(4):390–94. doi: 10.1007/BF02246272. [DOI] [PubMed] [Google Scholar]
  • 79.Perkins KA, Fonte C, Sanders M, et al. Threshold doses for nicotine discrimination in smokers and nonsmokers. Psychopharmacology (Berl) 2001;155:163–70. doi: 10.1007/s002130000660. [DOI] [PubMed] [Google Scholar]
  • 80.Perkins KA. Sex differences in nicotine reinforcement and reward: influences on the persistence of tobacco smoking. In: Bevins R, Caggiula AR, editors. The Motivational Impact of Nicotine and Its Role in Tobacco Use. Springer-Verlag; New York: 2009. pp. 143–69. [DOI] [PubMed] [Google Scholar]
  • 81.Perkins KA, Sanders M, D'Amico D, et al. Nicotine discrimination and self-administration as a function of smoking status. Psychopharmacology (Berl) 1997;131:361–70. doi: 10.1007/s002130050304. [DOI] [PubMed] [Google Scholar]
  • 82.Donny EC, Caggiula AR, Rowell PP, et al. Nicotine self-administration in rats: estrous cycle effects, sex differences and nicotinic receptor binding. Psychopharmacology (Berl) 2000;151(4):392–405. doi: 10.1007/s002130000497. [DOI] [PubMed] [Google Scholar]
  • 83.Lynch WJ. Sex and ovarian hormones influence vulnerability and motivation for nicotine during adolescence in rats. Pharmacol Biochem Behav. 2009;94(1):43–50. doi: 10.1016/j.pbb.2009.07.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Chaudhri N, Caggiula AR, Donny EC, et al. Sex differences in the contribution of nicotine and nonpharmacological stimuli to nicotine self-administration in rats. Psychopharmacology (Berl) 2005 doi: 10.1007/s00213-005-2152-3. [DOI] [PubMed] [Google Scholar]
  • 85.Guillem K, Vouillac C, Azar MR, et al. Monoamine oxidase inhibition dramatically increases the motivation to self-administer nicotine in rats. J Neurosci. 2005;25(38):8593–600. doi: 10.1523/JNEUROSCI.2139-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Dani JA, Harris RA. Nicotine addiction and comorbidity with alcohol abuse and mental illness. Nat Neurosci. 2005;8:1465–70. doi: 10.1038/nn1580. [DOI] [PubMed] [Google Scholar]
  • 87.Hughes JR, Rose GL, Callas PW. Nicotine is more inforcing in smokers with a past history of alcoholism than in smokers without this history. Alcohol: Clin Exper Res. 2000;24:1633–38. [PubMed] [Google Scholar]
  • 88.Luo Y, Marks MJ, Collins AC. Genotype regulates the development of tolerance to ethanol and cross-tolerance to nicotine. Alcohol. 1994;11:167–76. doi: 10.1016/0741-8329(94)90059-0. [DOI] [PubMed] [Google Scholar]
  • 89.Benowitz NL, Perez-Stable EJ, Herrera B, et al. Slower metabolism and reduced intake of nicotine from cigarette smoking in Chinese-Americans. J Natl Cancer Inst. 2002;94(2):108–15. doi: 10.1093/jnci/94.2.108. [DOI] [PubMed] [Google Scholar]
  • 90.Benowitz NL. Clinical pharmacology of nicotine: implications for understanding, preventing, and treating tobacco addiction. Clin Pharmacol Ther. 2008;83(4):531–41. doi: 10.1038/clpt.2008.3. [DOI] [PubMed] [Google Scholar]
  • 91.Sellers EM, Kaplan HL, Tyndale RF. Inhibition of cytochrome P450 2A6 increases nicotine's oral bioavailability and decreases smoking. Clin Pharmacol Ther. 2000;68(1):35–43. doi: 10.1067/mcp.2000.107651. [DOI] [PubMed] [Google Scholar]
  • 92.Karp I, O'Loughlin J, Hanley J, et al. Risk factors for tobacco dependence in adolescent smokers. Tob Control. 2006;15(3):199–204. doi: 10.1136/tc.2005.014118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Audrain-McGovern J, Al Koudsi N, Rodriguez D, et al. The role of CYP2A6 in the emergence of nicotine dependence in adolescents. Pediatrics. 2007;119(1):e264–74. doi: 10.1542/peds.2006-1583. [DOI] [PubMed] [Google Scholar]
  • 94.Derby KS, Cuthrell K, Caberto C, et al. Nicotine metabolism in three ethnic/racial groups with different risks of lung cancer. Cancer Epidemiol Biomarkers Prev. 2008;17:3526–35. doi: 10.1158/1055-9965.EPI-08-0424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Li MD. The genetics of nicotine dependence. Current Psychiatry Reports. 2006;8:158–64. doi: 10.1007/s11920-006-0016-0. [DOI] [PubMed] [Google Scholar]
  • 96.Perkins KA, Lerman C, Coddington SB, et al. Gene and gene by sex associations with initial nicotine sensitivity in nonsmokers. Behav Pharmacol. 2008;19:630–40. doi: 10.1097/FBP.0b013e32830c3621. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Ray R, Jepson C, Patterson F, et al. Association of OPRM1 Asn40Asp variant with the relative reinforcing value of nicotine in female smokers. Psychopharmacology (Berl) 2006;188:355–63. doi: 10.1007/s00213-006-0504-2. [DOI] [PubMed] [Google Scholar]
  • 98.Blendy JA, Strasser A, Walters CL, et al. Reduced nicotine reward in obesity: cross comparison in human and mouse. Psychopharmacology (Berl) 2005;180:306–15. doi: 10.1007/s00213-005-2167-9. [DOI] [PubMed] [Google Scholar]
  • 99.Perkins KA, Coddington SB, Karelitz JL, et al. Variability in initial nicotine sensitivity due to sex, history of other drug use, and parental smoking. Drug Alcohol Depend. 2009;99:47–57. doi: 10.1016/j.drugalcdep.2008.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Perkins KA, Lerman C, Coddington SB, et al. Initial nicotine sensitivity in humans as a function of impulsivity. Psychopharmacology (Berl) 2008;200:529–44. doi: 10.1007/s00213-008-1231-7. [DOI] [PubMed] [Google Scholar]
  • 101.Henningfield JE, Keenan R. Nicotine delivery kinetics and abuse liability. J Consult Clin Psychol. 1993;61:743–50. doi: 10.1037//0022-006x.61.5.743. [DOI] [PubMed] [Google Scholar]
  • 102.Perkins KA, Sayette M, Conklin CA, et al. Placebo effects of tobacco smoking and other nicotine intake. Nicotine & Tobacco Research. 2003;5:695–709. doi: 10.1080/1462220031000158636. [DOI] [PubMed] [Google Scholar]
  • 103.Perkins KA, Doyle T, Ciccocioppo M, et al. Sex differences in the influence of nicotine and dose instructions on subjective and reinforcing effects of smoking. Psychopharmacology (Berl) 2006;184:600–07. doi: 10.1007/s00213-005-0103-7. [DOI] [PubMed] [Google Scholar]
  • 104.Rose J, Ananda S, Jarvik ME. Cigarette smoking during anxiety-provoking and monotonous tasks. Addict Behav. 1983;8:353–59. doi: 10.1016/0306-4603(83)90035-7. [DOI] [PubMed] [Google Scholar]
  • 105.Conklin CA, Perkins KA. Subjective and reinforcing effects of smoking during negative mood induction. J Abnorm Psychol. 2005;114:153–64. doi: 10.1037/0021-843X.114.1.153. [DOI] [PubMed] [Google Scholar]
  • 106.O'Dell LE, Khroyan TV. Rodent models of nicotine reward: What do they tell us about tobacco abuse in humans? Pharmacol Biochem Behav. 2009;91(4):481–8. doi: 10.1016/j.pbb.2008.12.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Schachter S. Pharmacological and psychological determinants of smoking. Ann Intern Med. 1978;88:104–14. doi: 10.7326/0003-4819-88-1-104. [DOI] [PubMed] [Google Scholar]
  • 108.Perkins KA, Lerman C, Grottenthaler A, et al. Dopamine and opioid gene variants are associated with increased smoking reward and reinforcement due to negative mood. Behav Pharmacol. 2008;19:641–49. doi: 10.1097/FBP.0b013e32830c367c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Mitchell SH, de Wit H, Zacny JP. Effects of varying ethanol dose on cigarette consumption in healthy normal volunteers. Behav Pharmacol. 1995;6:359–65. [PubMed] [Google Scholar]
  • 110.Rush CR, Higgins ST, Vansickel AR, et al. Methylphenidate increases cigarette smoking. Psychopharmacology (Berl) 2005;181:781–89. doi: 10.1007/s00213-005-0021-8. [DOI] [PubMed] [Google Scholar]
  • 111.Roll JM, Higgins ST, Tidey J. Cocaine use can increase cigarette smoking: evidence from laboratory and naturalistic settings. Exper Clin Psychopharmacol. 1997;5:263–68. doi: 10.1037//1064-1297.5.3.263. [DOI] [PubMed] [Google Scholar]
  • 112.Perkins KA, Fonte C, Grobe JE. Sex differences in the acute effects of cigarette smoking on the reinforcing value of alcohol. Behav Pharmacol. 2000;11(1):63–70. doi: 10.1097/00008877-200002000-00007. [DOI] [PubMed] [Google Scholar]
  • 113.Shoaib M, Swanner LS, Yasar S, et al. Chronic caffeine exposure potentiates nicotine self-administration in rats. Psychopharmacology (Berl) 1999;142(4):327–33. doi: 10.1007/s002130050896. [DOI] [PubMed] [Google Scholar]
  • 114.Wooters TE, Neugebauer NM, Rush CR, et al. Methylphenidate enhances the abuse-related behavioral effects of nicotine in rats: intravenous self-administration, drug discrimination, and locomotor cross-sensitization. Neuropsychopharmacology. 2008;33(5):1137–48. doi: 10.1038/sj.npp.1301477. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.McQuown SC, Belluzzi JD, Leslie FM. Low dose nicotine treatment during early adolescence increases subsequent cocaine reward. Neurotoxicol Teratol. 2007;29(1):66–73. doi: 10.1016/j.ntt.2006.10.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Henningfield JE, Fant RV, Tomar SL. Smokeless tobacco: an addicting drug. Adv Dent Res. 1997;11(3):330–5. doi: 10.1177/08959374970110030401. [DOI] [PubMed] [Google Scholar]
  • 117.Hatsukami D, Lemmonds C, Tomar SL. Smokeless tobacco use: harm reduction or induction approach? Prev Med. 2004;38(3):309–17. doi: 10.1016/j.ypmed.2003.10.006. [DOI] [PubMed] [Google Scholar]
  • 118.National Cancer Institute . Cigars: Health Effects and Trends. Smoking and Tobacco Control Monograph No. 9. National Institutes of Health; Rockville, MD: 1998. [Google Scholar]
  • 119.Henningfield JE, Shiffman S, Ferguson SG, et al. Tobacco dependence and withdrawal: science base, challenges and opportunities for pharmacotherapy. Pharmacol Ther. 2009;123(1):1–16. doi: 10.1016/j.pharmthera.2009.03.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.World Health Organization Article 9 The World Health Organization Framework Convention on Tobacco Control. 2003 www.who.int/tobacco/framework/final_text/en/print.html.
  • 121.National Cancer Institute . Risks Associated with Smoking Cigarettes with Low Machine-Measured Yields of Tar and Nicotine. Smoking and Tobacco Control Monograph No 13. U.S. Department of Health and Human Servie, National Institutes of Health, National Cancer Institute; Bethesda, MD: 2001. [Google Scholar]
  • 122.Crellin RA. Project SHIP (Examination of branded and experimental products from the U.S.A.) 1985 [Google Scholar]
  • 123.Gordon DL. PM's global strategy: Marlboro product technology. 1992 [Google Scholar]
  • 124.Pankow JF. A consideration of the role of gas/particle partitioning in the deposition of nicotine and other tobacco smoke compounds in the respiratory tract. Chem Res Toxicol. 2001;14:1465–81. doi: 10.1021/tx0100901. [DOI] [PubMed] [Google Scholar]
  • 125.Pankow JF, Mader BT, Isabelle LM, et al. Conversion of nicotine in tobacco smoke to its volatile and available unprotonated form through the action of gaseous ammonia. Environmental Science and Technology. 1997;31:2428–33. [Google Scholar]
  • 126.Ashley DL, Pankow JF, Tavakoli AD, et al. Approaches, challenges, and experience in assessing free nicotine. In: Henningfield JE, London ED, Pogun S, editors. Nicotine Psychopharmacology Handbook of Experimental Pharmacologoy 192. Springer-Verlag; Berlin Heidelberg: 2009. pp. 437–56. [DOI] [PubMed] [Google Scholar]
  • 127.Jacob P, 3rd, Yu L, Shulgin AT, et al. Minor tobacco alkaloids as biomarkers for tobacco use: comparison of users of cigarettes, smokeless tobacco, cigars, and pipes. Am J Public Health. 1999;89(5):731–6. doi: 10.2105/ajph.89.5.731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Crooks PA, Li M, Dwoskin LP. Metabolites of nicotine in rat brain after peripheral nicotine administration. Cotinine, nornicotine, and norcotinine. Drug Metab Dispos. 1997;25(1):47–54. [PubMed] [Google Scholar]
  • 129.Ghosheh O, Dwoskin LP, Li WK, et al. Residence times and half-lives of nicotine metabolites in rat brain after acute peripheral administration of [2′-(14)C]nicotine. Drug Metab Dispos. 1999;27(12):1448–55. [PubMed] [Google Scholar]
  • 130.Ghosheh O, Dwoskin LP, Miller DK, et al. Accumulation of nicotine and its metabolites in rat brain after intermittent or continuous peripheral administration of [2′-(14)C]nicotine. Drug Metab Dispos. 2001;29(5):645–51. [PubMed] [Google Scholar]
  • 131.Dwoskin LP, Buxton ST, Jewell AL, et al. S(−)-nornicotine increases dopamine release in a calcium-dependent manner from superfused rat striatal slices. J Neurochem. 1993;60(6):2167–74. doi: 10.1111/j.1471-4159.1993.tb03502.x. [DOI] [PubMed] [Google Scholar]
  • 132.Goldberg SR, Risner ME, Stolerman IP, et al. Nicotine and some related compounds: effects on schedule-controlled behaviour and discriminative properties in rats. Psychopharmacology (Berl) 1989;97(3):295–302. doi: 10.1007/BF00439441. [DOI] [PubMed] [Google Scholar]
  • 133.Bardo MT, Green TA, Crooks PA, et al. Nornicotine is self-administered intravenously by rats. Psychopharmacology (Berl) 1999;146(3):290–6. doi: 10.1007/s002130051119. [DOI] [PubMed] [Google Scholar]
  • 134.Pratt JA, Stolerman IP, Garcha HS, et al. Discriminative stimulus properties of nicotine: further evidence for mediation at a cholinergic receptor. Psychopharmacology (Berl) 1983;81(1):54–60. doi: 10.1007/BF00439274. [DOI] [PubMed] [Google Scholar]
  • 135.Romano C, Goldstein A, Jewell NP. Characterization of the receptor mediating the nicotine discriminative stimulus. Psychopharmacology (Berl) 1981;74(4):310–5. doi: 10.1007/BF00432737. [DOI] [PubMed] [Google Scholar]
  • 136.Brioni JD, Kim DJ, O'Neill AB, et al. Clozapine attenuates the discriminative stimulus properties of (−)-nicotine. Brain Res. 1994;643(1-2):1–9. doi: 10.1016/0006-8993(94)90002-7. [DOI] [PubMed] [Google Scholar]
  • 137.Clemens KJ, Caille S, Stinus L, et al. The addition of five minor tobacco alkaloids increases nicotine-induced hyperactivity, sensitization and intravenous self-administration in rats. Int J Neuropsychopharmacol. 2009:1–12. doi: 10.1017/S1461145709000273. [DOI] [PubMed] [Google Scholar]
  • 138.Foddai M, Dosia G, Spiga S, et al. Acetaldehyde increases dopaminergic neuronal activity in the VTA. Neuropsychopharmacology. 2004;29(3):530–6. doi: 10.1038/sj.npp.1300326. [DOI] [PubMed] [Google Scholar]
  • 139.Cao J, Belluzzi JD, Loughlin SE, et al. Acetaldehyde, a major constituent of tobacco smoke, enhances behavioral, endocrine, and neuronal responses to nicotine in adolescent and adult rats. Neuropsychopharmacology. 2007;32(9):2025–35. doi: 10.1038/sj.npp.1301327. [DOI] [PubMed] [Google Scholar]
  • 140.Myers WD, Ng KT, Singer G. Intravenous self-administration of acetaldehyde in the rat as a function of schedule, food deprivation and photoperiod. Pharmacol Biochem Behav. 1982;17(4):807–11. doi: 10.1016/0091-3057(82)90364-1. [DOI] [PubMed] [Google Scholar]
  • 141.Amit Z, Smith BR. A multi-dimensional examination of the positive reinforcing properties of acetaldehyde. Alcohol. 1985;2(2):367–70. doi: 10.1016/0741-8329(85)90077-1. [DOI] [PubMed] [Google Scholar]
  • 142.Takayama S, Uyeno ET. Intravenous self-administration of ethanol and acetaldehyde by rats. Yakubutsu Seishin Kodo. 1985;5(4):329–34. [PubMed] [Google Scholar]
  • 143.Talhout R, Opperhuizen A, van Amsterdam JG. Role of acetaldehyde in tobacco smoke addiction. Eur Neuropsychopharmacol. 2007;17(10):627–36. doi: 10.1016/j.euroneuro.2007.02.013. [DOI] [PubMed] [Google Scholar]
  • 144.Khalil AA, Steyn S, Castagnoli N., Jr. Isolation and characterization of a monoamine oxidase inhibitor from tobacco leaves. Chem Res Toxicol. 2000;13(1):31–5. doi: 10.1021/tx990146f. [DOI] [PubMed] [Google Scholar]
  • 145.Fowler JS, Logan J, Wang GJ, et al. Monoamine oxidase and cigarette smoking. Neurotoxicology. 2003;24(1):75–82. doi: 10.1016/s0161-813x(02)00109-2. [DOI] [PubMed] [Google Scholar]
  • 146.Villegier AS, Lotfipour S, McQuown SC, et al. Tranylcypromine enhancement of nicotine self-administration. Neuropharmacology. 2007;52(6):1415–25. doi: 10.1016/j.neuropharm.2007.02.001. [DOI] [PubMed] [Google Scholar]
  • 147.Villegier AS, Blanc G, Glowinski J, et al. Transient behavioral sensitization to nicotine becomes long-lasting with monoamine oxidases inhibitors. Pharmacol Biochem Behav. 2003;76(2):267–74. doi: 10.1016/s0091-3057(03)00223-5. [DOI] [PubMed] [Google Scholar]
  • 148.Wooters TE, Bardo MT. The monoamine oxidase inhibitor phenelzine enhances the discriminative stimulus effect of nicotine in rats. Behav Pharmacol. 2007;18(7):601–8. doi: 10.1097/FBP.0b013e3282eff0d5. [DOI] [PubMed] [Google Scholar]
  • 149.Guillem K, Vouillac C, Azar MR, et al. Monoamine oxidase A rather than monoamine oxidase B inhibition increases nicotine reinforcement in rats. Eur J Neurosci. 2006;24(12):3532–40. doi: 10.1111/j.1460-9568.2006.05217.x. [DOI] [PubMed] [Google Scholar]
  • 150.Wayne GF, Connolly GN, Henningfield JE. Assessing internal tobacco industry knowledge of the neurobiology of tobacco dependence. Nicotine Tob Res. 2004;6(6):927–40. doi: 10.1080/14622200412331324839. [DOI] [PubMed] [Google Scholar]
  • 151.World Health Organization . The Scientific Basis of Tobacco Product Regulation: Report of a WHO Study Group. WHO Technical Report Series 945. WHO; Geneva, Switzerland: 2007. [PubMed] [Google Scholar]
  • 152.Royal College of Physicians . A report by the Tobacco Advisory Group of the Royal College of Physicians. Royal College of Physicians; London: 2007. Harm reduction in nicotine addiction. Helping people who can't quit. [Google Scholar]
  • 153.Henningfield JE, Benowitz NL, Ahijevych K, et al. Does menthol enhance the addictiveness of cigarettes? An agenda for research. Nicotine Tob Res. 2003;5(1):9–11. doi: 10.1080/1462220031000070543. [DOI] [PubMed] [Google Scholar]
  • 154.Gray N, Henningfield JE, Benowitz NL, et al. Toward a comprehensive long term nicotine policy. Tob Control. 2005;14(3):161–5. doi: 10.1136/tc.2004.010272. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Connolly GN. The marketing of nicotine addiction by one oral snuff manufacturer. Tobacco Control. 1995;4:73–79. [Google Scholar]
  • 156.Shatenstein S. Eliminating nicotine in cigarettes. Tobacco Control. 1999;8(1):106. author reply 07-09. [PMC free article] [PubMed] [Google Scholar]

RESOURCES