View Article

Abstract

Nicotine dependence remains a major global public health challenge, driven by the addictive potential of nicotine delivered through combustible and smokeless tobacco products. Nicotine exerts its effects primarily through activation of nicotinic acetylcholine receptors in the central nervous system, leading to dopamine release in reward pathways and reinforcing tobacco use. Despite widespread awareness of tobacco-related health risks, cessation is difficult due to withdrawal symptoms, craving, and neuroadaptations associated with chronic nicotine exposure. Nicotine Replacement Therapy (NRT) has emerged as a cornerstone pharmacological strategy for smoking cessation, providing controlled doses of nicotine without exposure to the toxic constituents of tobacco smoke.This review provides comprehensive pharmacological insights into the rational use of nicotine and NRT, focusing on their pharmacokinetic and pharmacodynamic properties. Key aspects such as absorption, distribution, metabolism, and elimination of nicotine are discussed, highlighting differences between smoking and therapeutic nicotine delivery systems. The neuropharmacological mechanisms underlying nicotine dependence, tolerance, and withdrawal are examined to explain the clinical effectiveness of NRT. Various NRT formulations, including transdermal patches, gums, lozenges, inhalers, and nasal sprays, are evaluated with respect to their onset of action, efficacy, and safety profiles. Clinical relevance is emphasized through discussion of NRT use in special populations, including individuals with cardiovascular disease, diabetes, mental health disorders, adolescents, and breastfeeding women. Evidence consistently indicates that NRT is significantly safer than continued tobacco use, with generally mild and localized adverse effects. Finally, future directions in nicotine harm reduction are explored, including combination therapies, personalized treatment approaches, and emerging non-combustible nicotine delivery systems. Overall, rational and evidence-based use of NRT represents an effective and safe approach to reducing nicotine dependence and tobacco-related morbidity and mortality.

Keywords

Nicotine, Nicotine Replacement Therapy, Pharmacokinetics, Neuropharmacology, Smoking Cessation

Introduction

Tobacco use remains one of the leading preventable causes of morbidity and mortality worldwide, contributing significantly to cardiovascular diseases, respiratory disorders, malignancies, and metabolic complications. According to global estimates, tobacco consumption accounts for more than eight million deaths annually, with a substantial proportion occurring in low- and middle-income countries. India represents one of the largest consumers of tobacco, with both smoked and smokeless forms widely prevalent. The high burden of tobacco use in India poses serious public health challenges due to early initiation, widespread cultural acceptance, and limited access to cessation services in certain regions [1,2]. Nicotine is the principal psychoactive component responsible for tobacco dependence. It produces addiction by rapidly activating nicotinic acetylcholine receptors in the central nervous system, leading to dopamine release in the mesolimbic reward pathway. Chronic exposure to nicotine results in neuroadaptations that reinforce compulsive tobacco use and precipitate withdrawal symptoms upon cessation. These withdrawal manifestations, including irritability, anxiety, impaired concentration, and intense craving, represent major barriers to successful quitting [3,4]. Behavioral interventions alone often prove insufficient for sustained abstinence, particularly in individuals with moderate to severe nicotine dependence. This has created a strong need for pharmacological approaches that target the neurobiological mechanisms of addiction. Nicotine Replacement Therapy provides a controlled and safer source of nicotine without exposure to toxic combustion products, thereby reducing withdrawal symptoms and cravings while allowing gradual dissociation from smoking behavior [5,6]. The present review aims to provide a comprehensive pharmacological overview of nicotine and Nicotine Replacement Therapy. It focuses on the pharmacokinetics of nicotine, its absorption, distribution, metabolism, and elimination, and highlights how these properties inform the rational clinical use of NRT in smoking cessation and tobacco harm reduction.

Figure 1. Pharmacological insights into nicotine dependence and the rationale for Nicotine Replacement Therapy (NRT).

The figure 1. illustrates the global and Indian burden of tobacco use and the central role of nicotine as the primary psychoactive component driving addiction. Nicotine activates nicotinic acetylcholine receptors in the central nervous system, leading to dopamine release in the mesolimbic reward pathway and resulting in neuroadaptation and dependence. Chronic exposure produces withdrawal symptoms such as irritability, anxiety, impaired concentration, and intense craving, which represent major barriers to smoking cessation. Nicotine Replacement Therapy provides controlled nicotine delivery without exposure to toxic combustion products, reducing withdrawal symptoms and cravings. Pharmacokinetic insights into nicotine absorption, distribution, metabolism, and elimination support the rational clinical use of NRT for effective smoking cessation and tobacco harm reduction.

2. Pharmacokinetics of Nicotine

2.1 Absorption

Nicotine absorption varies considerably depending on the route of administration and the formulation used. During cigarette smoking, nicotine is rapidly absorbed through the alveolar surface of the lungs due to the large surface area and extensive pulmonary blood flow. This results in swift entry of nicotine into systemic circulation and rapid delivery to the brain within seconds, producing strong reinforcing effects [7]. In contrast, smokeless tobacco products such as chewing tobacco and snuff allow nicotine absorption through the oral mucosa. The alkaline pH of these products enhances the non-ionized fraction of nicotine, facilitating efficient transmucosal absorption. Nicotine Replacement Therapy products such as gum, lozenges, sublingual tablets, inhalers, and nasal sprays are also absorbed through the oral or nasal mucosa, while transdermal patches deliver nicotine through the skin at a slow and sustained rate [8]. The degree of nicotine absorption is strongly influenced by pH, as nicotine is a weak base. Alkaline environments promote non-ionized nicotine, which readily crosses biological membranes, whereas acidic conditions reduce absorption. Formulation characteristics and user behavior further contribute to variability in nicotine uptake, particularly with oral NRT products [9].

2.2 Distribution

Following absorption, nicotine is rapidly distributed throughout the body, with a particular affinity for the brain. Due to its lipophilic nature, nicotine readily crosses the blood brain barrier, resulting in rapid central nervous system exposure. Brain nicotine concentrations rise quickly after smoking, reaching peak levels within minutes and closely correlating with the reinforcing effects of tobacco use [10]. Nicotine exhibits extensive tissue distribution, with a volume of distribution of approximately two to three liters per kilogram. Studies have demonstrated significant arterial venous concentration differences shortly after nicotine administration, particularly following inhalation. Arterial concentrations supplying the brain are substantially higher than venous concentrations, contributing to the intense pharmacological effects associated with smoking compared to slower delivery systems such as transdermal patches [11].

2.3 Metabolism

Nicotine is primarily metabolized in the liver, with cytochrome P450 enzyme CYP2A6 playing a dominant role. The major metabolite formed is cotinine, which accounts for approximately seventy percent of nicotine metabolism. Cotinine has a longer half-life than nicotine and is commonly used as a biomarker for nicotine exposure. Additional metabolites include nicotine N oxide and nor nicotine, which contribute minimally to pharmacological activity [12,13]. Genetic polymorphisms in CYP2A6 lead to considerable interindividual variability in nicotine metabolism. Individuals who metabolize nicotine rapidly tend to smoke more intensely and may experience greater difficulty quitting, whereas slow metabolizers often achieve better cessation outcomes with standard NRT doses [14].

2.4 Elimination

Nicotine and its metabolites are primarily eliminated through renal excretion. The rate of nicotine clearance is influenced by urinary pH, with acidic urine enhancing ion trapping and increasing excretion. On average, nicotine has a plasma half-life of approximately two hours, while cotinine exhibits a longer half-life of sixteen to twenty hours, allowing more stable measurement of exposure [15]. Significant interindividual variability exists in nicotine elimination due to differences in metabolic enzyme activity, renal function, age, sex, and concurrent drug use. These variations have important clinical implications, as they influence nicotine dependence severity, withdrawal intensity, and the optimal dosing of Nicotine Replacement Therapy [16].

3. Mechanisms of Action Of Nicotine

Nicotine exerts its pharmacological effects primarily through interaction with neuronal nicotinic acetylcholine receptors, leading to complex neurochemical and neuroadaptive changes that underlie reinforcement, dependence, and withdrawal. Understanding these mechanisms is essential for the rational use of nicotine and Nicotine Replacement Therapy in smoking cessation strategies.

3.1 Neuropharmacology

Nicotine is a tertiary amine alkaloid that readily crosses the blood brain barrier and binds selectively to nicotinic acetylcholine receptors located throughout the central and peripheral nervous systems. These receptors are ligand gated ion channels composed of five subunits arranged around a central pore and are activated by endogenous acetylcholine as well as exogenous nicotine. Binding of nicotine to nicotinic acetylcholine receptors results in rapid channel opening and influx of sodium and calcium ions, leading to neuronal depolarization and activation of downstream signaling pathways. The diversity of receptor subunit composition contributes to regional specificity and functional variability of nicotine responses in the brain [17]. Among the various receptor subtypes, the alpha four beta two receptor is the most abundant and widely distributed in the human brain and is considered the principal mediator of nicotine dependence. Activation of this receptor subtype is strongly associated with reinforcing effects and dopamine release in reward related brain regions. Genetic and experimental studies have demonstrated that deletion or mutation of the beta two subunit markedly reduces nicotine self-administration and reward related behaviors, highlighting its critical role in addiction [18,19]. The alpha seven homomeric receptor subtype is characterized by high calcium permeability and rapid desensitization and is implicated in cognitive processes such as learning, memory, and sensory gating. This receptor is thought to contribute to the cognitive enhancing effects reported by smokers [20]. The alpha three beta four receptor subtype is predominantly expressed in autonomic ganglia and brainstem regions and has been associated with cardiovascular and autonomic effects of nicotine, including increases in heart rate and blood pressure [21].

3.2 Neurotransmitter Modulation

Nicotine mediated activation of nicotinic acetylcholine receptors leads to the release of multiple neurotransmitters that collectively shape its psychopharmacological profile. The most prominent effect is stimulation of dopamine release within the mesolimbic reward pathway, particularly from neurons projecting from the ventral tegmental area to the nucleus accumbens. Dopamine release in this circuit is widely recognized as a critical neurobiological substrate for reinforcement and drug seeking behavior. Acute nicotine exposure increases dopamine transmission, producing pleasurable and rewarding effects that reinforce continued use [22,23]. In addition to dopamine, nicotine modulates several other neurotransmitter systems. Nicotine enhances glutamate release, which further stimulates dopaminergic neurons and amplifies reward signaling. At the same time, chronic nicotine exposure reduces inhibitory gamma aminobutyric acid signaling, thereby removing inhibitory control over dopamine neurons and sustaining dopaminergic activation [24]. Nicotine also increases the release of norepinephrine, contributing to heightened arousal and alertness, and serotonin, which influences mood regulation and emotional state. Endorphin release induced by nicotine may further contribute to stress reduction and perceived relaxation associated with smoking [25]. The combined modulation of these neurotransmitter systems explains both the reinforcing properties of nicotine and its effects on mood, cognition, and stress responsiveness.

3.3 Neuroadaptation and Dependence

Chronic exposure to nicotine results in neuroadaptive changes that underlie tolerance, dependence, and withdrawal. One of the most distinctive features of nicotine pharmacology is receptor upregulation, in which prolonged nicotine exposure leads to an increase in the number of nicotinic acetylcholine receptor binding sites, particularly the alpha four beta two subtype. This paradoxical increase is thought to result from receptor desensitization followed by compensatory overexpression, contributing to tolerance and the need for continued nicotine intake to maintain receptor occupancy and prevent withdrawal symptoms [26,27]. Nicotine withdrawal is characterized by negative affective states including irritability, anxiety, dysphoria, and increased stress sensitivity. These symptoms are associated with reduced dopamine release in reward circuits and activation of stress related neuropeptide systems. Increasing evidence implicates corticotropin releasing factor signaling within the amygdala as a key mediator of the emotional and stress related components of nicotine withdrawal. Activation of corticotropin releasing factor pathways enhances anxiety like behavior and promotes relapse by increasing nicotine seeking during abstinence [28]. The interplay between dopaminergic hypofunction and stress system hyperactivity represents a major challenge in smoking cessation and provides a strong rationale for the use of Nicotine Replacement Therapy. By maintaining partial receptor stimulation and stabilizing neurotransmitter release, NRT helps to alleviate withdrawal symptoms and reduce relapse risk in dependent individuals [29,30].

4. Nicotine Replacement Therapy (NRT)

4.1 Rationale and Therapeutic Principles

Nicotine replacement therapy is based on the principle of tobacco harm reduction and aims to support smoking cessation by delivering nicotine in controlled and safer forms without exposure to the harmful toxicants present in tobacco smoke. Combustible tobacco products contain thousands of chemicals including carcinogens carbon monoxide and oxidant gases that are primarily responsible for smoking related morbidity and mortality rather than nicotine itself. By substituting cigarettes with pharmaceutical nicotine products NRT reduces withdrawal symptoms and craving while allowing individuals to gradually dissociate nicotine intake from the behavioral and sensory cues of smoking [31]. The therapeutic rationale of NRT lies in its ability to stabilize plasma nicotine concentrations at levels sufficient to alleviate withdrawal symptoms but lower and slower than those achieved through cigarette smoking. This controlled delivery minimizes the reinforcing dopamine spikes associated with smoking and reduces the risk of dependence reinforcement [32]. Compared with continued tobacco use NRT significantly lowers cardiovascular respiratory and oncogenic risks and has been demonstrated to double quit rates when combined with behavioral counseling [33]. The predictable pharmacokinetic profile and well-established safety of NRT make it a first line pharmacotherapy for smoking cessation across diverse populations including those with stable cardiovascular disease [34].

4.2 Common Forms of NRT

Transdermal nicotine patches provide a sustained and steady release of nicotine through the skin over a period of sixteen to twenty-four hours resulting in stable plasma nicotine concentrations. This mode of delivery is particularly effective for reducing baseline withdrawal symptoms and preventing early morning cravings. Because patches avoid rapid nicotine peaks they are associated with lower abuse potential and minimal reinforcement effects making them suitable for highly dependent smokers and long-term cessation strategies [35]. Oral nicotine formulations such as gum lozenges and sublingual tablets deliver nicotine through the buccal mucosa allowing flexible dosing in response to acute cravings. These products produce faster nicotine absorption compared to patches but slower than cigarettes thereby providing symptomatic relief while maintaining safety. Correct usage technique including intermittent chewing for gum and passive dissolution for lozenges is essential to optimize absorption and reduce gastrointestinal side effects [36]. Nicotine inhalers and nasal sprays offer the most rapid nicotine delivery among NRT formulations closely mimicking the pharmacokinetic profile of smoking. Nasal sprays in particular achieve rapid increases in plasma nicotine levels and are effective for highly dependent smokers experiencing intense cravings. However due to local irritation and higher dependence potential these formulations are generally reserved for individuals who do not respond adequately to slower acting NRT options [37].

4.3 Combination Therapy

Combination nicotine replacement therapy involves the concurrent use of a long acting nicotine patch with a rapid acting formulation such as gum lozenge inhaler or nasal spray. This approach addresses both steady state nicotine requirements and breakthrough cravings providing superior withdrawal control compared with monotherapy. Clinical evidence consistently demonstrates higher abstinence rates with combination therapy particularly in heavy smokers and individuals with previous failed quit attempts [38]. Combination therapy allows individualized titration of nicotine doses based on dependence severity and craving patterns thereby improving adherence and treatment satisfaction. International smoking cessation guidelines recommend combination NRT for patients who continue to experience cravings despite adequate patch dosing or those with high baseline nicotine dependence. Importantly studies have shown that combination therapy does not increase adverse cardiovascular events and remains significantly safer than continued smoking reinforcing its role as an effective and rational cessation strategy [39,40].

5. Emerging Nicotine Delivery Systems

5.1 Electronic Cigarettes

Electronic cigarettes are battery powered devices designed to deliver nicotine through inhalation without the combustion of tobacco. Unlike conventional cigarettes, electronic cigarettes function by heating a liquid solution containing nicotine, solvents such as propylene glycol or glycerol, and flavoring agents to generate an aerosol that is inhaled by the user. This mechanism avoids the production of carbon monoxide and many carcinogenic byproducts associated with tobacco combustion, thereby reducing exposure to toxic substances [41]. When compared with conventional nicotine replacement therapy, electronic cigarettes demonstrate a faster rate of nicotine delivery, particularly with newer generation devices that are capable of producing higher aerosol volumes and more efficient nicotine absorption [42]. Although traditional NRT products such as patches and gums provide steady or intermittent nicotine delivery, electronic cigarettes more closely mimic the behavioral and sensory aspects of smoking, which may enhance user satisfaction and adherence [43]. Clinical studies indicate that experienced users of electronic cigarettes can achieve plasma nicotine concentrations comparable to combustible cigarettes, whereas NRT typically results in lower and slower nicotine absorption [44]. Potential benefits of electronic cigarettes include improved craving suppression and higher acceptability among smokers unwilling to quit nicotine use entirely. However, concerns remain regarding long term safety, variability in nicotine content, and exposure to aerosol constituents whose health effects are not fully understood. Evidence suggests that while electronic cigarettes are likely less harmful than smoking, they are not risk free and should be approached cautiously within tobacco harm reduction strategies [45].

5.2 Tobacco Free Nicotine Products

Tobacco free nicotine products represent an emerging category of non-combustible nicotine delivery systems designed to provide nicotine without tobacco derived toxins. Oral nicotine pouches are among the most widely studied products in this group. These pouches are placed between the gum and lip, allowing nicotine absorption through the oral mucosa without combustion or aerosol generation [46]. Pharmacokinetic studies demonstrate that nicotine pouches provide a slower onset of nicotine delivery compared to smoking but faster than transdermal patches. Peak plasma nicotine concentrations are dose dependent and influenced by pouch composition, moisture content, and duration of use [47]. Although nicotine exposure is generally lower than that achieved with cigarettes, user reported satisfaction and craving reduction are comparable, particularly with higher strength formulations [48]. From a user experience perspective, nicotine pouches offer advantages such as discretion, ease of use, and absence of smoke or vapor. Importantly, toxicological analyses reveal substantially lower levels of harmful and potentially harmful constituents compared to traditional tobacco products, supporting their potential role in tobacco harm reduction for adult smokers who are unable or unwilling to quit nicotine completely [49].

6. NRT Use in Special Populations

6.1 Pregnancy and Breastfeeding

Nicotine readily crosses the placenta and is transferred to infants through breast milk, raising concerns about fetal and neonatal exposure. The concentration of nicotine in breast milk is closely related to maternal plasma nicotine levels and the timing of nicotine intake relative to feeding [50]. While complete nicotine abstinence remains the ideal goal, NRT may be considered when the alternative is continued smoking, which exposes the infant to multiple toxic substances beyond nicotine [51]. Risk minimization strategies during breastfeeding include the use of short acting NRT formulations such as gum or lozenges and timing their use immediately after breastfeeding to allow nicotine levels to decline before the next feeding. Evidence indicates that controlled nicotine exposure through NRT poses significantly lower risks than tobacco smoking during lactation [52].

6.2 Adolescents

Adolescents exhibit unique patterns of nicotine dependence characterized by heightened neurobiological vulnerability and fluctuating motivation to quit. Safety considerations are critical, as children are more susceptible to nicotine toxicity at lower doses. Clinical studies suggest that NRT is generally safe in adolescents when used under medical supervision, with no serious adverse effects reported [53]. The effectiveness of NRT in this population is strongly linked to concurrent behavioral counseling. Counseling addresses psychosocial factors, improves adherence, and enhances cessation outcomes. Without structured support, pharmacological treatment alone is often insufficient for sustained abstinence in adolescents [54].

6.3 Cardiovascular Disease

Nicotine exerts acute hemodynamic effects by increasing heart rate and blood pressure through sympathetic nervous system activation. These effects raise concerns regarding NRT use in patients with cardiovascular disease. However, evidence indicates that the cardiovascular impact of therapeutic nicotine is substantially lower than that of smoking, which also involves carbon monoxide exposure and prothrombotic effects [55]. Clinical trials in patients with stable cardiovascular disease demonstrate that NRT does not significantly increase the risk of adverse cardiac events and can be safely used to support smoking cessation. Transdermal nicotine patches are particularly well tolerated due to their steady nicotine delivery profile [56].

6.4 Diabetes Mellitus

Nicotine influences glucose metabolism by increasing catecholamine release, which may impair insulin sensitivity and glycemic control. When initiating NRT, individuals with diabetes mellitus may experience transient changes in blood glucose levels, necessitating closer monitoring [57]. Despite these effects, smoking cessation supported by NRT leads to long term metabolic benefits that outweigh short term glycemic fluctuations. Regular blood glucose monitoring and appropriate adjustment of antidiabetic therapy are recommended during the cessation period [58].

6.5 Mental Health Disorders

Smoking prevalence is disproportionately high among individuals with mental health disorders, including depression, anxiety disorders, and schizophrenia. Nicotine withdrawal symptoms may be more pronounced in this population, contributing to lower cessation success rates [59]. NRT plays a critical role in facilitating smoking cessation among psychiatric patients by reducing withdrawal related anxiety, irritability, and dysphoria. Importantly, smoking cessation alters the metabolism of certain psychotropic medications, as smoking induces hepatic enzymes that increase drug clearance. When smoking is discontinued, careful monitoring and dose adjustment of medications such as antipsychotics and antidepressants are essential [60]. Combination NRT is often recommended to achieve adequate symptom control and improve cessation outcomes in this high-risk group.

7. Safety Profile and Adverse Effects Of Nicotine Replacement Therapy

Nicotine Replacement Therapy has been extensively evaluated for safety across diverse populations and clinical settings. Evidence from large scale clinical trials and meta analyses indicates that adverse effects associated with NRT are generally mild, transient and localized to the route of administration rather than systemic or life threatening. Common local adverse effects include skin irritation and pruritus with transdermal patches, oral and throat irritation with gums lozenges and inhalers, and nasal discomfort or rhinitis with nasal spray formulations. Systemic effects such as nausea dizziness headache palpitations and sleep disturbances have also been reported but occur infrequently and are typically dose dependent and self-limiting. Serious adverse events attributable directly to NRT are rare and occur at a substantially lower frequency than those associated with continued tobacco smoking [61]. When comparing risks of NRT with smoking, it is critical to recognize that nicotine itself is not the primary cause of smoking related morbidity and mortality. Combustible tobacco smoke contains thousands of toxic and carcinogenic compounds including carbon monoxide polycyclic aromatic hydrocarbons and tobacco specific nitrosamines which are absent in medicinal nicotine products. Multiple studies have demonstrated that NRT does not significantly increase cardiovascular risk even in patients with stable cardiovascular disease, whereas continued smoking markedly elevates the risk of myocardial infarction stroke and sudden cardiac death [62,63]. Thus from a pharmacological and clinical perspective NRT represents a substantially safer alternative to smoking. Despite strong evidence supporting its safety, patient misconceptions regarding NRT remain a significant barrier to its effective use. Many smokers mistakenly believe that NRT is as harmful as cigarettes or that prolonged use may cause cancer or severe addiction. These misconceptions are often reinforced by inadequate counseling and misunderstanding of nicotine pharmacology. Addressing these concerns through patient education is essential, as exaggerated fear of adverse effects frequently leads to premature discontinuation or refusal of NRT, thereby reducing cessation success rates [64,65].

8. Limitations and Causes of Nicotine Replacement Therapy Failure

Although NRT is an evidence based and effective intervention, its real-world effectiveness is often limited by several pharmacological behavioral and psychological factors. One major limitation is inadequate nicotine delivery. Most NRT formulations deliver nicotine more slowly and at lower peak plasma concentrations than cigarettes. As a result some individuals, particularly heavy or highly dependent smokers, may experience insufficient relief of cravings and withdrawal symptoms, leading to relapse [66]. Underdosing and poor adherence are additional contributors to NRT failure. Many users employ lower doses than recommended or discontinue therapy prematurely due to fear of side effects or misunderstanding of proper use. Incorrect chewing technique with nicotine gum or insufficient duration of patch therapy can significantly reduce nicotine absorption and therapeutic efficacy. Studies have shown that adherence to recommended dosing regimens is strongly correlated with cessation success, highlighting the importance of healthcare provider guidance and follow up [67,68]. Behavioral and psychological factors also play a crucial role in treatment outcomes. Smoking is not solely a pharmacological dependence but a complex behavior reinforced by habitual cues emotional regulation and social contexts. NRT addresses the neurochemical component of dependence but does not directly modify learned behaviors or coping strategies. Without concurrent behavioral counseling stress management and motivational support many individuals continue to associate smoking related rituals with reward and relief, increasing the likelihood of relapse. This underscores the need for combined pharmacological and behavioral interventions to optimize cessation outcomes [69].

9. Tobacco Harm Reduction Perspective

From a tobacco harm reduction perspective NRT plays a central role in reducing the health burden associated with nicotine addiction. Harm reduction strategies acknowledge that while complete nicotine abstinence is ideal, substituting high risk combustible tobacco products with substantially lower risk nicotine delivery systems can yield significant public health benefits. NRT provides nicotine in controlled pharmaceutical doses without exposure to combustion related toxins, thereby reducing the risk of cancer cardiovascular disease and respiratory illness [70]. The emergence of alternative nicotine products such as electronic cigarettes and tobacco free oral nicotine pouches has expanded the harm reduction landscape. While these products differ in delivery kinetics and user experience, medicinal NRT remains the most extensively studied and regulated option with a well-established safety profile. At a population level increased access to NRT has been associated with higher quit rates reduced smoking prevalence and decreased tobacco related mortality. Public health strategies that integrate NRT into cessation programs and primary healthcare settings have demonstrated measurable benefits [71,72]. Regulatory and ethical considerations are central to the harm reduction debate. Medicinal nicotine products are subject to rigorous quality safety and efficacy standards, whereas newer nicotine delivery systems often exist within evolving regulatory frameworks. Ethical concerns arise regarding youth initiation marketing practices and long term dependence. However restricting access to safer nicotine alternatives may unintentionally perpetuate smoking related harm. Balanced regulation that prioritizes adult smokers while preventing youth uptake is therefore essential for ethical and effective harm reduction policy [73].

10. FUTURE DIRECTIONS

Future advances in nicotine replacement therapy are expected to focus on personalized nicotine treatment strategies. Individual variability in nicotine metabolism dependence severity and behavioral patterns suggests that a one size fits all approach may be suboptimal. Tailoring NRT type dose and duration to individual patient characteristics may improve efficacy and adherence [74]. Pharmacogenetics represents a promising area of research in this context. Genetic polymorphisms affecting nicotine metabolism particularly in enzymes such as CYP2A6 influence nicotine clearance withdrawal severity and treatment response. Identifying genetic markers associated with NRT responsiveness could allow clinicians to optimize dosing strategies and select appropriate formulations for individual patients [75,76]. The development of faster and safer nicotine delivery systems also represents an important future direction. Novel formulations aim to more closely mimic the rapid nicotine delivery of cigarettes while maintaining a favorable safety profile. Such innovations may enhance craving relief reduce relapse and improve patient satisfaction. Continued research integrating pharmacology clinical outcomes and real-world effectiveness will be essential to refine NRT and strengthen its role in global tobacco control efforts [77].

CONCLUSION

Nicotine dependence is a complex neuropharmacological condition driven by rapid nicotine delivery, activation of nicotinic acetylcholine receptors, and dopaminergic reinforcement pathways. Understanding the pharmacokinetic and pharmacodynamic properties of nicotine provides critical insight into the rationale behind Nicotine Replacement Therapy. By delivering nicotine in controlled doses without exposure to toxic combustion products, NRT effectively alleviates withdrawal symptoms and cravings while substantially reducing health risks associated with tobacco use. The clinical significance of rational NRT use lies in appropriate product selection, adequate dosing, and sufficient duration of therapy tailored to the level of nicotine dependence. Evidence consistently demonstrates that NRT is safe, well tolerated, and markedly safer than continued smoking, even in individuals with comorbid conditions such as cardiovascular disease and mental health disorders. Misconceptions regarding NRT safety and improper use remain major barriers to treatment success and must be addressed through patient education and clinical guidance. Importantly, nicotine addiction extends beyond pharmacological dependence to include behavioral and psychological components. Therefore, optimal smoking cessation outcomes are achieved when pharmacotherapy with NRT is integrated with behavioral counseling, motivational support, and relapse prevention strategies. A comprehensive and individualized approach that combines pharmacological insight with behavioral intervention remains essential for effective tobacco cessation and long-term harm reduction.                                          

REFERENCE

  1. World Health Organization. WHO global report on trends in prevalence of tobacco use 2000 to 2025. 4th ed. Geneva WHO; 2021.
  2. Ministry of Health and Family Welfare Government of India. Global Adult Tobacco Survey India 2016 to 2017. New Delhi MoHFW; 2018.
  3. Prochaska JJ, Benowitz NL. Current advances in research in treatment and recovery: Nicotine addiction. Science advances. 2019 Oct 16;5(10): eaay9763.
  4. Dani JA, De Biasi M. Cellular mechanisms of nicotine addiction. Pharmacology Biochemistry and Behavior. 2001 Dec 1;70(4):439-46.
  5. Henningfield JE Fant RV Buchhalter AR Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005; 55:281 299.
  6. Stead LF Perera R Bullen C et al. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012;11:CD000146.
  7. Benowitz NL Porchet H Jacob P. Nicotine dependence and tolerance in man pharmacokinetic and pharmacodynamic investigations. Prog Brain Res. 1989; 79:279 287.
  8. Le Houezec J Benowitz NL. Basic and clinical psychopharmacology of nicotine. Clin Chest Med. 1991; 12:681 699.
  9. Benowitz NL Jacob P. Determinants of nicotine intake while chewing nicotine gum. Clin Pharmacol Ther. 1987; 41:467 473.
  10. Henningfield JE London ED Benowitz NL. Arterial venous differences in plasma nicotine concentrations after cigarette smoking. JAMA. 1990; 263:2049 2050.
  11. Porchet HC Benowitz NL Sheiner LB. Apparent tolerance to the acute effect of nicotine results in part from distribution kinetics. J Clin Invest. 1987; 80:1466 1471.
  12. Benowitz NL. Clinical pharmacology of nicotine. Annu Rev Med. 1986; 37:21 32.
  13. Hukkanen J Jacob P Benowitz NL. Metabolism and disposition kinetics of nicotine. Pharmacol Rev. 2005; 57:79 115.
  14. Malaiyandi V Sellers EM Tyndale RF. Implications of CYP2A6 genetic variation for smoking behaviors and nicotine dependence. Clin Pharmacol Ther. 2005; 77:145 158.
  15. Benowitz NL Jacob P Jones RT Rosenberg J. Interindividual variability in the metabolism and cardiovascular effects of nicotine. J Pharmacol Exp Ther. 1982; 221:368 372.
  16. Dempsey D Benowitz NL. Risks and benefits of nicotine to aid smoking cessation in pregnancy. Drug Saf. 2001; 24:277 322. •  Gotti C, Zoli M, Clementi F. Brain nicotinic acetylcholine receptors native subtypes and their relevance. Trends Pharmacol Sci. 2006; 27:482–491.
  17. Picciotto MR, Zoli M, Rimondini R, Lena C, Marubio M, et al. Acetylcholine receptors containing the beta two subunit are involved in the reinforcing properties of nicotine. Nature. 1998; 391:173–177.
  18. Maskos U, Molles BE, Pons S, Besson M, Guiard BP, et al. Nicotine reinforcement and cognition restored by targeted expression of nicotinic receptors. Nature. 2005; 436:103–107.
  19. Levin ED, Bettegowda C, Blosser J, Gordon J. AR R17779 an alpha seven nicotinic agonist improves learning and memory in rats. Behav Pharmacol. 1999; 10:675–680.
  20. Hajós M, Hurst RS, Hoffmann WE, Krause M, Wall TM, et al. The selective alpha seven nicotinic acetylcholine receptor agonist PNU 282987 enhances GABAergic synaptic activity and restores auditory gating deficits. J Pharmacol Exp Ther. 2005; 312:1213–1222.
  21. Aberger K, Chitravanshi VC, Sapru HN. Cardiovascular responses to nicotine microinjection in the ventrolateral medulla. Brain Res. 2001; 892:138–146.
  22. Dani JA, De Biasi M. Cellular mechanisms of nicotine addiction. Pharmacol Biochem Behav. 2001; 70:439–446.
  23. Nestler EJ. Is there a common molecular pathway for addiction? Nat Neurosci. 2005; 8:1445–1449.
  24. Mansvelder HD, McGehee DS. Cellular and synaptic mechanisms of nicotine addiction. J Neurobiol. 2002; 53:606–617.
  25. Wonnacott S. Presynaptic nicotinic acetylcholine receptors. Trends Neurosci. 1997; 20:92–98.
  26. Wang H, Sun X. Desensitized nicotinic receptors in brain. Brain Res Rev. 2005; 48:420–437.
  27. Brody AL, Mandelkern MA, London ED, Olmstead RE, Farahi J, et al. Cigarette smoking saturates brain alpha four beta two nicotinic acetylcholine receptors. Arch Gen Psychiatry. 2006; 63:907–915.
  28. George O, Ghozland S, Azar MR, Cottone P, Zorrilla EP, et al. CRF CRF one system activation mediates withdrawal induced increases in nicotine self-administration. Proc Natl Acad Sci USA. 2007; 104:17198–17203.
  29. Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005; 55:281–299.
  30. Russell MA. The future of nicotine replacement therapy. Br J Addict. 1991;86(5):653–658.
  31. Benowitz NL. Neurobiology of nicotine addiction implications for smoking cessation treatment. Am J Med. 2008;121(4 Suppl 1): S3–S10.
  32. Stead LF, Perera R, Bullen C, Mant D, Lancaster T. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012;11:CD000146.
  33. Joseph AM, Norman SM, Ferry LH, Prochazka AV, Westman EC, Steele BG, et al. The safety of transdermal nicotine as an aid to smoking cessation in patients with cardiac disease. N Engl J Med. 1996;335(24):1792–1798.
  34. Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005;55(5):281–299.
  35. Benowitz NL, Jacob P. Nicotine absorption and cardiovascular effects with smokeless tobacco use comparison with cigarettes and nicotine gum. Clin Pharmacol Ther. 1988;44(1):23–28.
  36. Sutherland G, Russell MAH, Stapleton J, Feyerabend C, Ferno O. Nasal nicotine spray a rapid nicotine delivery system. Psychopharmacology. 1992;108(4):512–518.
  37.  Blondal T, Gudmundsson LJ, Olafsdottir I, Gustavsson G, Westin A. Nicotine nasal spray with nicotine patch for smoking cessation. BMJ. 1999;318(7179):285–289.
  38. Bader P, McDonald P, Selby P. An algorithm for tailoring pharmacotherapy for smoking cessation. Tob Control. 2009;18(1):34–42.
  39. Mills EJ, Wu P, Lockhart I, Wilson K, Ebbert JO. Adverse events associated with nicotine replacement therapy systematic review and meta-analysis. Tob Induc Dis. 2010;8(1):8.
  40. Knight West O, Bullen C. Electronic cigarettes for the management of nicotine addiction. Subst Abuse Rehabil. 2016; 7:111 118.
  41. Vansickel AR, Cobb CO, Weaver MF, Eissenberg T. A clinical laboratory model for evaluating the acute effects of electronic cigarettes. Cancer Epidemiol Biomarkers Prev. 2010; 19:1945 1953.
  42. Bullen C, Howe C, Laugesen M, McRobbie H, Parag V, Williman J, et al. Electronic cigarettes for smoking cessation. Lancet. 2013; 382:1629 1637.
  43. Farsalinos KE, Spyrou A, Tsimopoulou K, Stefopoulos C, Romagna G, Voudris V. Nicotine absorption from electronic cigarette use. Sci Rep. 2014; 4:4133.
  44. Talih S, Balhas Z, Eissenberg T, Salman R, Karaoghlanian N, El Hellani A, et al. Effects of device characteristics on nicotine yield. Nicotine Tob Res. 2015; 17:150 157.
  45. Lunell E, Fagerstrom K, Hughes J, Pendrill R. Pharmacokinetic comparison of a novel nicotine pouch. Nicotine Tob Res. 2020; 22:1757 1763.
  46. Liu J, Rensch J, McEwan M, Gale N. Nicotine pharmacokinetics of oral nicotine pouches. Psychopharmacology. 2022; 239:2863 2873.
  47. Chapman F, McDermott S, Rudd K, McEwan M. Pharmacokinetic and subjective effects of nicotine pouches. Psychopharmacology. 2022; 239:2931 2943.
  48. McEwan M, Gale N, Hardie G, Proctor C. Toxicological assessment of tobacco free nicotine products. Regul Toxicol Pharmacol. 2021; 123:104937.
  49. Dempsey DA, Benowitz NL. Risks and benefits of nicotine to aid smoking cessation in pregnancy. Drug Saf. 2001; 24:277 322.
  50. Woolf A, Burkhart K, Caraccio T, Litovitz T. Childhood poisoning involving nicotine patches. Pediatrics. 1997;99: e4.
  51. Hale TW. Medications and mothers’ milk. 14th ed. Amarillo: Hale Publishing; 2010.
  52. Smith TA, House RF, Croghan IT, Gauvin TR, Offord KP, Hurt RD. Nicotine patch therapy in adolescent smokers. Pediatrics. 1996; 98:659 667.
  53. Moolchan ET, Robinson ML, Ernst M, Cadet JL, Pickworth WB, Heishman SJ, et al. Safety and efficacy of nicotine patch and gum in adolescents. Pediatrics. 2005; 115:407 414.
  54. Benowitz NL, Gourlay SG. Cardiovascular toxicity of nicotine. J Am Coll Cardiol. 1997; 29:1422 1431.
  55. Joseph AM, Norman SM, Ferry LH, Prochazka AV, Westman EC, Steele BG, et al. Safety of transdermal nicotine in cardiac patients. N Engl J Med. 1996; 335:1792 1798.
  56. Persson LG, Hjalmarson A. Smoking cessation in patients with diabetes mellitus. Scand J Prim Health Care. 2006; 24:75 80.
  57. Williams JM, Hughes JR. Pharmacotherapy for tobacco dependence in mental illness. Psychiatr Ann. 2003; 33:457 466.
  58. Fagerstrom KO, Aubin HJ. Management of smoking cessation in psychiatric disorders. Curr Med Res Opin. 2009; 25:511 518.
  59. Mills EJ, Wu P, Lockhart I, Wilson K, Ebbert JO. Adverse events associated with nicotine replacement therapy. Tob Induc Dis. 2010; 8:8.
  60. Mills EJ, Wu P, Lockhart I, Wilson K, Ebbert JO. Adverse events associated with nicotine replacement therapy for smoking cessation. Tob Induc Dis. 2010;8(1):8.
  61. Hughes JR. Dependence on and abuse of nicotine replacement medications. Nicotine Tob Res. 2011;13(6):465–470.
  62. Benowitz NL, Gourlay SG. Cardiovascular toxicity of nicotine implications for nicotine replacement therapy. J Am Coll Cardiol. 1997;29(7):1422–1431.
  63. Joseph AM, Norman SM, Ferry LH, Prochazka AV, Westman EC, Steele BG, et al. The safety of transdermal nicotine as an aid to smoking cessation in patients with cardiac disease. N Engl J Med. 1996;335(24):1792–1798.
  64. Foulds J, Hughes JR, Hyland A, Le Houezec J, McNeill A, Melvin C, et al. Barriers to use of FDA approved smoking cessation medications. Nicotine Tob Res. 2009;11(2):161–168.
  65. McNeill A, Foulds J, Bates C. Regulation of nicotine replacement therapies. Addiction. 2001;96(12):1757–1768.
  66. Russell MAH. The future of nicotine replacement. Br J Addict. 1991;86(5):653–658.
  67. Stead LF, Perera R, Bullen C, Mant D, Lancaster T. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012;11:CD000146.
  68. Benowitz NL, Jacob P III. Suppression of nicotine intake during ad libitum cigarette smoking by high dose transdermal nicotine. J Pharmacol Exp Ther. 1998;287(3):958–962.
  69. Balfour DJK. The neurobiology of tobacco dependence. Nicotine Tob Res. 2004;6(6):899–912.
  70. Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005;55(5):281–299.
  71. Stapleton JA. Progress on nicotine replacement therapy for smokers. BMJ. 1999;318(7188):289–290.
  72. Stead LF, Lancaster T. Interventions to reduce harm from continued tobacco use. Cochrane Database Syst Rev. 2007;3:CD005231.
  73. World Health Organization. Tobacco harm reduction approaches. WHO Technical Report Series. Geneva; 2020.
  74. Bader P, McDonald P, Selby P. An algorithm for tailoring pharmacotherapy for smoking cessation. Tob Control. 2009;18(1):34–42.
  75. Lerman C, Tyndale R, Patterson F, Wileyto EP, Shields PG, Pinto A, et al. Nicotine metabolite ratio predicts efficacy of nicotine patch therapy. Clin Pharmacol Ther. 2006;80(6):600–608.
  76. Benowitz NL. Pharmacogenetics of nicotine addiction and treatment. Annu Rev Pharmacol Toxicol. 2008; 48:57–81.
  77. Hatsukami DK, Benowitz NL, Rennard SI, Oncken C, Hecht SS. Biomarkers to assess the utility of potential reduced exposure tobacco products. Nicotine Tob Res. 2006;8(2):169–191.

Reference

  1. World Health Organization. WHO global report on trends in prevalence of tobacco use 2000 to 2025. 4th ed. Geneva WHO; 2021.
  2. Ministry of Health and Family Welfare Government of India. Global Adult Tobacco Survey India 2016 to 2017. New Delhi MoHFW; 2018.
  3. Prochaska JJ, Benowitz NL. Current advances in research in treatment and recovery: Nicotine addiction. Science advances. 2019 Oct 16;5(10): eaay9763.
  4. Dani JA, De Biasi M. Cellular mechanisms of nicotine addiction. Pharmacology Biochemistry and Behavior. 2001 Dec 1;70(4):439-46.
  5. Henningfield JE Fant RV Buchhalter AR Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005; 55:281 299.
  6. Stead LF Perera R Bullen C et al. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012;11:CD000146.
  7. Benowitz NL Porchet H Jacob P. Nicotine dependence and tolerance in man pharmacokinetic and pharmacodynamic investigations. Prog Brain Res. 1989; 79:279 287.
  8. Le Houezec J Benowitz NL. Basic and clinical psychopharmacology of nicotine. Clin Chest Med. 1991; 12:681 699.
  9. Benowitz NL Jacob P. Determinants of nicotine intake while chewing nicotine gum. Clin Pharmacol Ther. 1987; 41:467 473.
  10. Henningfield JE London ED Benowitz NL. Arterial venous differences in plasma nicotine concentrations after cigarette smoking. JAMA. 1990; 263:2049 2050.
  11. Porchet HC Benowitz NL Sheiner LB. Apparent tolerance to the acute effect of nicotine results in part from distribution kinetics. J Clin Invest. 1987; 80:1466 1471.
  12. Benowitz NL. Clinical pharmacology of nicotine. Annu Rev Med. 1986; 37:21 32.
  13. Hukkanen J Jacob P Benowitz NL. Metabolism and disposition kinetics of nicotine. Pharmacol Rev. 2005; 57:79 115.
  14. Malaiyandi V Sellers EM Tyndale RF. Implications of CYP2A6 genetic variation for smoking behaviors and nicotine dependence. Clin Pharmacol Ther. 2005; 77:145 158.
  15. Benowitz NL Jacob P Jones RT Rosenberg J. Interindividual variability in the metabolism and cardiovascular effects of nicotine. J Pharmacol Exp Ther. 1982; 221:368 372.
  16. Dempsey D Benowitz NL. Risks and benefits of nicotine to aid smoking cessation in pregnancy. Drug Saf. 2001; 24:277 322. •  Gotti C, Zoli M, Clementi F. Brain nicotinic acetylcholine receptors native subtypes and their relevance. Trends Pharmacol Sci. 2006; 27:482–491.
  17. Picciotto MR, Zoli M, Rimondini R, Lena C, Marubio M, et al. Acetylcholine receptors containing the beta two subunit are involved in the reinforcing properties of nicotine. Nature. 1998; 391:173–177.
  18. Maskos U, Molles BE, Pons S, Besson M, Guiard BP, et al. Nicotine reinforcement and cognition restored by targeted expression of nicotinic receptors. Nature. 2005; 436:103–107.
  19. Levin ED, Bettegowda C, Blosser J, Gordon J. AR R17779 an alpha seven nicotinic agonist improves learning and memory in rats. Behav Pharmacol. 1999; 10:675–680.
  20. Hajós M, Hurst RS, Hoffmann WE, Krause M, Wall TM, et al. The selective alpha seven nicotinic acetylcholine receptor agonist PNU 282987 enhances GABAergic synaptic activity and restores auditory gating deficits. J Pharmacol Exp Ther. 2005; 312:1213–1222.
  21. Aberger K, Chitravanshi VC, Sapru HN. Cardiovascular responses to nicotine microinjection in the ventrolateral medulla. Brain Res. 2001; 892:138–146.
  22. Dani JA, De Biasi M. Cellular mechanisms of nicotine addiction. Pharmacol Biochem Behav. 2001; 70:439–446.
  23. Nestler EJ. Is there a common molecular pathway for addiction? Nat Neurosci. 2005; 8:1445–1449.
  24. Mansvelder HD, McGehee DS. Cellular and synaptic mechanisms of nicotine addiction. J Neurobiol. 2002; 53:606–617.
  25. Wonnacott S. Presynaptic nicotinic acetylcholine receptors. Trends Neurosci. 1997; 20:92–98.
  26. Wang H, Sun X. Desensitized nicotinic receptors in brain. Brain Res Rev. 2005; 48:420–437.
  27. Brody AL, Mandelkern MA, London ED, Olmstead RE, Farahi J, et al. Cigarette smoking saturates brain alpha four beta two nicotinic acetylcholine receptors. Arch Gen Psychiatry. 2006; 63:907–915.
  28. George O, Ghozland S, Azar MR, Cottone P, Zorrilla EP, et al. CRF CRF one system activation mediates withdrawal induced increases in nicotine self-administration. Proc Natl Acad Sci USA. 2007; 104:17198–17203.
  29. Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005; 55:281–299.
  30. Russell MA. The future of nicotine replacement therapy. Br J Addict. 1991;86(5):653–658.
  31. Benowitz NL. Neurobiology of nicotine addiction implications for smoking cessation treatment. Am J Med. 2008;121(4 Suppl 1): S3–S10.
  32. Stead LF, Perera R, Bullen C, Mant D, Lancaster T. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012;11:CD000146.
  33. Joseph AM, Norman SM, Ferry LH, Prochazka AV, Westman EC, Steele BG, et al. The safety of transdermal nicotine as an aid to smoking cessation in patients with cardiac disease. N Engl J Med. 1996;335(24):1792–1798.
  34. Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005;55(5):281–299.
  35. Benowitz NL, Jacob P. Nicotine absorption and cardiovascular effects with smokeless tobacco use comparison with cigarettes and nicotine gum. Clin Pharmacol Ther. 1988;44(1):23–28.
  36. Sutherland G, Russell MAH, Stapleton J, Feyerabend C, Ferno O. Nasal nicotine spray a rapid nicotine delivery system. Psychopharmacology. 1992;108(4):512–518.
  37.  Blondal T, Gudmundsson LJ, Olafsdottir I, Gustavsson G, Westin A. Nicotine nasal spray with nicotine patch for smoking cessation. BMJ. 1999;318(7179):285–289.
  38. Bader P, McDonald P, Selby P. An algorithm for tailoring pharmacotherapy for smoking cessation. Tob Control. 2009;18(1):34–42.
  39. Mills EJ, Wu P, Lockhart I, Wilson K, Ebbert JO. Adverse events associated with nicotine replacement therapy systematic review and meta-analysis. Tob Induc Dis. 2010;8(1):8.
  40. Knight West O, Bullen C. Electronic cigarettes for the management of nicotine addiction. Subst Abuse Rehabil. 2016; 7:111 118.
  41. Vansickel AR, Cobb CO, Weaver MF, Eissenberg T. A clinical laboratory model for evaluating the acute effects of electronic cigarettes. Cancer Epidemiol Biomarkers Prev. 2010; 19:1945 1953.
  42. Bullen C, Howe C, Laugesen M, McRobbie H, Parag V, Williman J, et al. Electronic cigarettes for smoking cessation. Lancet. 2013; 382:1629 1637.
  43. Farsalinos KE, Spyrou A, Tsimopoulou K, Stefopoulos C, Romagna G, Voudris V. Nicotine absorption from electronic cigarette use. Sci Rep. 2014; 4:4133.
  44. Talih S, Balhas Z, Eissenberg T, Salman R, Karaoghlanian N, El Hellani A, et al. Effects of device characteristics on nicotine yield. Nicotine Tob Res. 2015; 17:150 157.
  45. Lunell E, Fagerstrom K, Hughes J, Pendrill R. Pharmacokinetic comparison of a novel nicotine pouch. Nicotine Tob Res. 2020; 22:1757 1763.
  46. Liu J, Rensch J, McEwan M, Gale N. Nicotine pharmacokinetics of oral nicotine pouches. Psychopharmacology. 2022; 239:2863 2873.
  47. Chapman F, McDermott S, Rudd K, McEwan M. Pharmacokinetic and subjective effects of nicotine pouches. Psychopharmacology. 2022; 239:2931 2943.
  48. McEwan M, Gale N, Hardie G, Proctor C. Toxicological assessment of tobacco free nicotine products. Regul Toxicol Pharmacol. 2021; 123:104937.
  49. Dempsey DA, Benowitz NL. Risks and benefits of nicotine to aid smoking cessation in pregnancy. Drug Saf. 2001; 24:277 322.
  50. Woolf A, Burkhart K, Caraccio T, Litovitz T. Childhood poisoning involving nicotine patches. Pediatrics. 1997;99: e4.
  51. Hale TW. Medications and mothers’ milk. 14th ed. Amarillo: Hale Publishing; 2010.
  52. Smith TA, House RF, Croghan IT, Gauvin TR, Offord KP, Hurt RD. Nicotine patch therapy in adolescent smokers. Pediatrics. 1996; 98:659 667.
  53. Moolchan ET, Robinson ML, Ernst M, Cadet JL, Pickworth WB, Heishman SJ, et al. Safety and efficacy of nicotine patch and gum in adolescents. Pediatrics. 2005; 115:407 414.
  54. Benowitz NL, Gourlay SG. Cardiovascular toxicity of nicotine. J Am Coll Cardiol. 1997; 29:1422 1431.
  55. Joseph AM, Norman SM, Ferry LH, Prochazka AV, Westman EC, Steele BG, et al. Safety of transdermal nicotine in cardiac patients. N Engl J Med. 1996; 335:1792 1798.
  56. Persson LG, Hjalmarson A. Smoking cessation in patients with diabetes mellitus. Scand J Prim Health Care. 2006; 24:75 80.
  57. Williams JM, Hughes JR. Pharmacotherapy for tobacco dependence in mental illness. Psychiatr Ann. 2003; 33:457 466.
  58. Fagerstrom KO, Aubin HJ. Management of smoking cessation in psychiatric disorders. Curr Med Res Opin. 2009; 25:511 518.
  59. Mills EJ, Wu P, Lockhart I, Wilson K, Ebbert JO. Adverse events associated with nicotine replacement therapy. Tob Induc Dis. 2010; 8:8.
  60. Mills EJ, Wu P, Lockhart I, Wilson K, Ebbert JO. Adverse events associated with nicotine replacement therapy for smoking cessation. Tob Induc Dis. 2010;8(1):8.
  61. Hughes JR. Dependence on and abuse of nicotine replacement medications. Nicotine Tob Res. 2011;13(6):465–470.
  62. Benowitz NL, Gourlay SG. Cardiovascular toxicity of nicotine implications for nicotine replacement therapy. J Am Coll Cardiol. 1997;29(7):1422–1431.
  63. Joseph AM, Norman SM, Ferry LH, Prochazka AV, Westman EC, Steele BG, et al. The safety of transdermal nicotine as an aid to smoking cessation in patients with cardiac disease. N Engl J Med. 1996;335(24):1792–1798.
  64. Foulds J, Hughes JR, Hyland A, Le Houezec J, McNeill A, Melvin C, et al. Barriers to use of FDA approved smoking cessation medications. Nicotine Tob Res. 2009;11(2):161–168.
  65. McNeill A, Foulds J, Bates C. Regulation of nicotine replacement therapies. Addiction. 2001;96(12):1757–1768.
  66. Russell MAH. The future of nicotine replacement. Br J Addict. 1991;86(5):653–658.
  67. Stead LF, Perera R, Bullen C, Mant D, Lancaster T. Nicotine replacement therapy for smoking cessation. Cochrane Database Syst Rev. 2012;11:CD000146.
  68. Benowitz NL, Jacob P III. Suppression of nicotine intake during ad libitum cigarette smoking by high dose transdermal nicotine. J Pharmacol Exp Ther. 1998;287(3):958–962.
  69. Balfour DJK. The neurobiology of tobacco dependence. Nicotine Tob Res. 2004;6(6):899–912.
  70. Henningfield JE, Fant RV, Buchhalter AR, Stitzer ML. Pharmacotherapy for nicotine dependence. CA Cancer J Clin. 2005;55(5):281–299.
  71. Stapleton JA. Progress on nicotine replacement therapy for smokers. BMJ. 1999;318(7188):289–290.
  72. Stead LF, Lancaster T. Interventions to reduce harm from continued tobacco use. Cochrane Database Syst Rev. 2007;3:CD005231.
  73. World Health Organization. Tobacco harm reduction approaches. WHO Technical Report Series. Geneva; 2020.
  74. Bader P, McDonald P, Selby P. An algorithm for tailoring pharmacotherapy for smoking cessation. Tob Control. 2009;18(1):34–42.
  75. Lerman C, Tyndale R, Patterson F, Wileyto EP, Shields PG, Pinto A, et al. Nicotine metabolite ratio predicts efficacy of nicotine patch therapy. Clin Pharmacol Ther. 2006;80(6):600–608.
  76. Benowitz NL. Pharmacogenetics of nicotine addiction and treatment. Annu Rev Pharmacol Toxicol. 2008; 48:57–81.
  77. Hatsukami DK, Benowitz NL, Rennard SI, Oncken C, Hecht SS. Biomarkers to assess the utility of potential reduced exposure tobacco products. Nicotine Tob Res. 2006;8(2):169–191.

Photo
Prathamesh Chandankhede
Corresponding author

Vidyaniketan College of pharmacy, Anjangaon surji Amravati, Maharashtra, India

Photo
Bhakti Chipada
Co-author

Vidyaniketan College of pharmacy, Anjangaon surji Amravati, Maharashtra, India

Photo
Dr. Anand Khode
Co-author

Vidyaniketan College of pharmacy, Anjangaon surji Amravati, Maharashtra, India

Prathamesh Chandankhede*, Bhakti Chipada, Dr. Anand Khode, Pharmacological Insights into the Rational Use of Nicotine and Nicotine Replacement Therapy, Int. J. Sci. R. Tech., 2025, 2 (12), 425-436. https://doi.org/10.5281/zenodo.18056447

More related articles
Review on: - Formulation & Evaluation of Dental Ge...
Sana Sultana Nadaf , Mayuri Auti , ...
Misuse and Abuse of Over the Counter Medication: A...
Kalyani Patil, Ankita Taskar, Sakshi Benke, Komal Ahire, Bhavisha...
Branding A Legend, The Journey of Bajaj Chetak from Classic to Electric...
Aasifa Hussain, Sahayaraj Antony Michael, Dr. Sivasaktibalan D. P. , ...
A Comprehensive Review on High Performance Liquid Chromatography (HPLC): Introdu...
Deep Jyoti Shah, Supriya Hazra, Sourav Bhowmick, Amisha Kumari, Bhumika Kumari, Dibya Kumari, Neha K...
Related Articles
Implementation of Hybrid Soft-Hard Exosuit for Adaptive Ankle Movement Therapy D...
Ranjithkumar S., Krishnaragavan V., Pranitha V. K. J., Harivansh B. R., Dharshini D., ...
Knowledge and Attitude Regarding Human Milk Banking Among Antenatal Mothers in S...
Manphun Wangsu, Nongmeikapam Monika, Iarihunlang Lyngdoh, ...
Reinventing Medicines: Drug Repurposing as A New Frontier in Cancer Therapy...
Tejswini Gaikwad, Rashee Shahu, Sunita Kode, Shivcharan Kamble, Pooja Rasal, ...
Immuno-Oncology in Cancer Therapy: Mechanistic Insights, Clinical Applications, ...
Tejaswini Bhavar, Sonal Bhatkar , Rutuja Rathod, Sumit Kolse , Prajkta Mahajan , ...
More related articles
Misuse and Abuse of Over the Counter Medication: A Growing Public Health Concern...
Kalyani Patil, Ankita Taskar, Sakshi Benke, Komal Ahire, Bhavisha Chaudhari, Vinod Bairagi, ...