Cite

INTRODUCTION

Cigarette smoking is a leading avoidable contributor to morbidity and mortality, playing a causal role in lung cancer, chronic obstructive pulmonary disease, and cardiovascular disease, among other diseases (1). Smoking cigarettes is addictive, primarily due to nicotine – a naturally occurring alkaloid in tobacco leaves (2) that has effects on mood and relaxation through its activity at neuronal nicotinic receptors in the brain (3). Nicotine in cigarette smoke is rapidly absorbed through the lungs and transferred to the bloodstream (4), which distributes it around the body. As a result, the pharmacokinetic profile of nicotine during cigarette smoking is distinguished by a swift rise in blood nicotine levels (5), and the consequent desirable effects are rapid (3). Prominent regulatory agencies and health-care bodies, such as the Royal College of Physicians and Public Health England, consider nicotine to be relatively harmless at the levels of exposure attained during cigarette smoking, as compared to the significant harm caused by other components of cigarette smoke (68). Instead, the harmful effects of smoking are due to the long-term inhalation of 8,700 or more identified chemicals (9), including many with a known link to the development of specific diseases (10). The individual health risk associated with cigarette smoking is correlated with the length of smoking history and the number of cigarettes smoked each day, such that smoking cigarettes for longer and with greater frequency leads to increased risk of disease (11, 12).

Quitting smoking substantially reduces the risk of an individual developing a smoking-related disease. However, while large proportions (> 50%) of smokers report a desire to stop smoking, and many make cessation attempts each year (13), fewer than 1 in 10 smokers successfully quit smoking each year (13, 14). As a result, an alternative approach based on tobacco harm reduction (THR) has been put forward (15). The principle of THR is to encourage smokers, who would not otherwise quit smoking, to switch from smoking combustible cigarettes to using alternative non-combustible/smokeless nicotine and tobacco products with decreased levels of toxicant emissions, such as e-cigarettes (6). While not entirely risk free, such switching could significantly reduce smokers’ exposure to harmful toxicants (6, 7, 16, 17) and potentially an individual’s risk of developing a smoking-related disease (18, 19).

Oral nicotine pouches (NPs) are alternative tobacco-free nicotine products that have the potential for THR. They are similar in form and usage to Swedish snus, a smokeless tobacco product, representative brands of which have been recognised by the U.S. Food and Drug Administration as a modified-risk tobacco product that has been epidemio-logically proven to offer significantly reduced risks of disease compared to cigarettes. Introduction to the market is expected to benefit the health of the population as a whole (20). Both snus and NPs are placed under the upper lip, allowing the nicotine to be absorbed through the oral mucosa. Whereas snus contains tobacco, NPs contain a cellulose matrix with pharmaceutical grade nicotine (2123). Therefore, when compared to snus, NPs do not contain tobacco and consequently have lower levels in key harmful and potentially harmful tobacco product and tobacco smoke constituents (23).

Recent in vitro toxicology studies have reported that NP extracts have significantly less biological activity than an equivalent reference snus product across multiple flavour variants and nicotine content (24, 25). As well as having lower toxicity, alternative non-combustible/smokeless nicotine and tobacco products must be able to deliver nicotine to smokers efficiently to be successfully adopted as part of a THR strategy (6, 26). Recent studies indicate that NPs may deliver nicotine sufficiently to smokers seeking satisfactory alternative products (27, 28), but to date there is little information of the effects of nicotine content and flavours on nicotine pharmacokinetics for these products.

In this study, we have determined and compared the pharmacokinetics of nicotine absorption among current cigarette smokers using nine different Velo NPs including a single product with a nicotine content of 4 mg and eight NPs with different flavours (e.g., fruit and mint/menthol), at a nicotine content of 7 mg. We have also assessed product-liking for the NPs. Based on our findings, we discuss the THR potential of NPs in delivering nicotine efficiently and providing a choice of flavours to smokers seeking an alternative to smoking.

METHODS
Study design

The present randomised, controlled, crossover clinical study was conducted at a single site in Kansas City, KS, USA. The study was registered on the U.S. Clinical-Trials.gov registry (NCT04846088). Approval was given by an Institutional Review Board (IRB; Ethics (WIRB Copernicus Group, Puyallup, WA, USA; study reference number 1305801) before study commencement. The study was conducted in accordance with the U.S. Code of Federal Regulations (CFR) governing Protection of Human Subjects (21 CFR Part 50), Financial Disclosure by Clinical Investigators (21 CFR Part 54), and IRBs (21 CFR Part 56). It was also carried out in accordance with the protocol and under the principles of the International Council for Harmonisation (ICH) of Technical Requirements for Pharmaceuticals for Human Use Guideline for Good Clinical Practice (GCP) E6 (R2). All participants provided written informed consent before enrollment and before undergoing any study procedures, including screening. At any time, they were free to quit smoking, withdraw their consent, or withdraw from the study.

Participants

Forty-two healthy male or female participants who met the inclusion criteria were enrolled in the study within 45 days of their screening visit to ensure that a minimum of 36 participants completed the study. An attempt was made to recruit a balance of sexes with no less than 40% of either sex represented and to recruit at least 15–20% black/African American participants to reflect the reported percentage of U.S. smokers in this group. Participants were aged 22–65 years inclusive and generally healthy as determined by clinical laboratory evaluations (including haematology, clinical chemistry, urinalysis, serology, urine drug and urine/breath alcohol screen), medical history, physical examination including oral examination, vital signs assessment and 12 lead electrocardiograms. All participants were current smokers with a minimum 1-year smoking history and who were smoking at least 10 per day of filtered non-menthol or menthol cigarettes between 83 and 100 mm in length as their primary source of tobacco use. Exhaled carbon monoxide and urine cotinine were assessed to confirm cigarette smoking status. Participants also self-reported use of smokeless tobacco products (e.g., moist snuff or snus) at least once or twice in their lifetime before screening. Female participants underwent a serum pregnancy test at screening and a urine pregnancy test at checkin to the clinical site. Women of childbearing potential were required to use an accepted form of contraception for 30 days before and after the study.

The main exclusion criteria were:

pregnancy or breastfeeding (women only);

self-reported non-inhalation of cigarette smoke;

self-reported previous or current use of any Velo or Dryft NPs;

presence of gum bleeding and/or abscess, open mouth sores or oral ulcers at screening or check-in;

history of significant allergic reaction to any substance including mint, wintergreen or spearmint flavouring;

whole-blood donation within 56 days of screening or blood plasma donation within 7 days of screening or between screening and check-in;

delaying a decision to quit using tobacco or nicotine products in order to participate in the study or selfreporting a quit attempt within 30 days of screening;

or current use of any smoking cessation aid.

Investigational products

The study products were nine Velo portioned oral NPs including a 4-mg/pouch of nicotine with one flavour (Velo Wintergreen) and 7-mg/pouch of nicotine with eight flavours (Velo Max Wintergreen, Spearmint, Peppermint, Citrus Burst, Black Cherry, Coffee, Dragon Fruit, and Cinnamon). The contents of each Velo or Velo Max pouch include a powdered cellulosic substrate, pharmaceutical grade tobacco-derived nicotine, pH adjusters, sweeteners, and other ingredients specific to each flavour. The powdered mixture is pouched in a porous wrapping material referred to as “fleece”.

Study procedures

All participants were screened against the inclusion and exclusion criteria and completed a questionnaire on their tobacco product use. Within 45 days of screening, they were admitted to the clinic on Day 1 of the study and were confined to the site for approximately 10 days until the last assessments had been made. On admission, participants’ eligibility was reconfirmed and they underwent vital signs assessments, physical and oral examinations, and a urine pregnancy test (females only). At the end of the study, they underwent clinical laboratory testing, physical and oral examinations, vital signs assessments, and a urine pregnancy test (females only).

On Day 1, all participants took part in a product familiarisation session lasting at least 45 min in which they tried the Velo Max 7-mg nicotine and peppermint flavour study NP. Throughout the study, participants were required to abstain from using any nicotine product for at least 12 h before the product use session the following morning. In each product use session, participants used a single study product as specified by predetermined randomisation sequences. Within 30 min of the start of the session, participants were asked to rinse their mouth with approximately 50 mL water and were then instructed to place a single Velo pouch in their mouth anywhere between their gum and their upper lip and allow the pouch to absorb saliva and moisten. Participants moved the pouch from one side of the mouth to the other approximately every 10 min, and the pouch remained in the mouth for a period of 45 min.

After product use and blood sampling was completed, participants were allowed to smoke their own brand of combustible cigarettes ad libitum until the 12 h abstinence period prior to the next product use session.

Blood sampling for nicotine pharmacokinetics

Venous blood samples were collected by direct venepuncture or through an indwelling cannula. Blood samples (4 mL) were taken at –5, 2, 5, 10, 15, 30, 45, 60, 90 and 180 min relative to product administration into K2-EDTA vacutainer tubes. To ensure anticoagulation, the tubes were inverted 10 times and centrifuged within 60 min (1,500 g, 4 °C, 10 min). The plasma was stored in two aliquots at –20 °C. The time from blood sample collection to plasma storage did not exceed 90 min.

Nicotine analysis was done by Altasciences (Laval, Quebec, Canada) using liquid chromatography with tandem mass spectrometry detection as previously described (28). In brief, nicotine was extracted from 0.15 mL plasma by protein precipitation and analysed using a Waters XBridge C18 column on an AB Sciex API 5000 quadrupole mass spectrometer in positive ion mode for the detection of nicotine.

Analyst® software version 1.6.3, was used to acquire and review chromatograms. The internal standard was nicotine-D4, and nicotine was quantified over a theoretical concentration range of 0.2–100.0 ng/mL. For the analysis only non-smokers prepared the spiking solutions, calibrant and quality control samples.

Furthermore, blank samples were injected before the pre-test to check for the presence of nicotine in the system. In addition to blank and zero standards, all runs had a set of 11 non-zero standards and 4 levels of Quality Control samples prepared with analyte-free human plasma. Incurred Sample Reanalysis evaluation was assessed concurrently to the sample analysis with at least 10% of the first 1000 analysable study samples and 5% of the remaining samples reassayed and compared to their original values. The sample analysis was conducted in accordance with U.S. Food and Drug Administration Guidance for Industry, (29) and European Medicines Agency Guideline on Bioanalytical Method Validation (30).

Subjective effects assessments

At the end of the pharmacokinetic session (180 min relative to the start of product use), participants completed a single product-liking questionnaire to evaluate the subjective effect of study product use. Answers were given as a numeric rating score from 0 to 10, with 0 corresponding to “strong disliking”, 5 corresponding to “neither like nor dislike” and 10 corresponding to “strong liking”.

Safety assessments

Adverse events (AEs) were defined as any untoward medical occurrence or condition experienced by a participant after signing the informed consent form until completion of the study, irrespective of whether it was considered to be related to the use of study products. An AE could be any unfavourable and unintended sign (e.g., abnormal laboratory finding), symptom or disease, without any judgment about causality.

All AEs, whether volunteered, elicited or noted on the physical examination/oral examination at the end of the study, were recorded throughout the study. The start and stop date and time of all AEs was captured. Participants who presented with unresolved or new AEs at study conclusion or early termination were followed up until the AE had resolved or stabilised. A product-emergent adverse event (PEAE) was defined as an AE that was not present prior to study product use or an AE that was present but worsened in intensity or frequency after study product use.

A serious adverse event (SAE) was defined as any medical occurrence that resulted in death or was life-threatening, required inpatient hospitalisation or prolongation of an existing hospitalisation, resulted in persistent or significant incapacity or substantial disruption of a person’s ability to conduct normal life functions, was a congenital anomaly or birth defect or was a severe medical event that required medical or surgical intervention to prevent one of the above outcomes.

Sample size and statistical methods

Using data from prior studies, it was estimated that 36 participants would be needed to have at least an 80% chance of obtaining a 95% confidence interval with a half-width of up to 20% of the means for the primary endpoints. The target number of participants to be recruited into this study was 42 participants, which allowed for approximately a 14% dropout rate with a goal of 36 participants completing the study.

Raw nicotine concentrations and derived baseline-adjusted concentrations were determined by compartmental methods using Phoenix® WinNonlin® version 8.0 (Certara, Princeton, NJ, USA). The primary endpoint nicotine pharmacokinetic parameters maximum plasma nicotine concentration (Cmax) and area under the nicotine concentration-versus-time curve from time zero to 180 min after the start of study product use (AUCnic 0–180) were adjusted for baseline plasma nicotine concentration under the assumption that nicotine elimination follows first-order kinetics. Negative concentrations resulting from baseline adjustment were forced to zero. The secondary pharmacokinetic endpoint time to maximum plasma nicotine concentration (Tmax) was also calculated based on the baseline-adjusted plasma nicotine concentrations. Observed plasma nicotine concentration values below the lower limit of quantification were set at half the lower limit of quantification, and missing data were considered as missing at random and not imputed. Demographic and pharmacokinetic parameters presented here are for the pharmacokinetic population, which included 41 participants who used at least one study product and it had sufficient data to derive at least one pharmacokinetic parameter.

The pharmacokinetic parameters Cmax and AUCnic 0–180 were summarised using descriptive statistics for each study product. These statistics included the number of nonmissing data points, mean, standard deviation (SD), coefficient of variation, minimum, median, and maximum. Additionally, geometric means, geometric SD, and geometric coefficient of variation calculated for Cmax and AUCnic 0–180 are reported. For Tmax only non-missing data points, minimum, median, and maximum values are reported.

Statistical comparisons of Cmax and AUCnic 0–180 between study products were performed using paired t-tests on a log-transformed scale. For Tmax, comparisons were made using a sign test on the original data scale. For all pharmacokinetic parameter comparisons, Bonferroni adjustment was used during the statistical analyses. With a total of 24 comparisons (8 product comparisons × 3 parameters), a p-value less than 0.002 (0.05/24) would indicate statistically significant difference.

For the product-liking subjective effects measure, the overall product-liking numeric rating score was summarised by product using descriptive statistics (non-missing data points, mean, SD, minimum, quartile 1, quartile 3, and maximum).

RESULTS
Participant demographics

A total of 42 participants met eligibility requirements and were enrolled into the study. The majority of randomised participants (95.2%) completed the study according to the protocol. All 42 participants (100%) completed Day 1, 41 participants (97.6%) completed Days 2 and 3, and 40 participants (95.2%) completed Days 4 through 9. Two participants voluntarily withdrew from the study early, for a family emergency on Day 4 (1 participant) and due to the number of blood draws on Day 1 (1 participant).

Basic participant demographic data are summarised in Table 1. The male:female ratio was 68:32; 80.5% of the participants were white and 19.5% were black/African American.

Demographic data of study participants collected at screening.

Variable Number of participants % Mean (SD)
Age (years) 42 40.4 – 11.38
Sex
Male 28 66.7
Female 14 33.3
Weight (kg) 42 81.17 – 17.02
Height (cm) 42 173.5 – 9.2
BMI (kg/m2) 42 26.8 – 4.45
Race
White 33 78.6
Black / African American 9 21.4
Ethnicity
Hispanic or Latino 6 14.3
Not Hispanic or Latino 36 85.7

Abbreviations: BMI: body-mass index; SD: standard deviation.

Nicotine pharmacokinetics

Mean plasma nicotine concentration-time curves for the Velo NPs are shown in Figure 1, while descriptive data and statistical comparisons of the pharmacokinetic parameters are summarised in Table 2 and Table 3, respectively. For each study product, the mean plasma nicotine concentration increased throughout use, reaching a peak at the end of the 45-min product use period. Mean coefficient of variation (CV) maximum nicotine concentration (Cmax) with baseline-adjustment was lowest for the Velo Wintergreen 4-mg pouch (11.224 ng/mL (28.7%)) and was similar for the Velo Max 7-mg pouches regardless of flavour, ranging from 16.220 ng/mL (33.3%) for Velo Max Citrus Burst 7-mg to 17.888 ng/mL (26.2%) for Velo Max Cinnamon 7-mg (Table 2).

Figure 1.

Baseline-adjusted plasma nicotine concentrations over time.

Figure shows the baseline adjusted mean nicotine concentrations with bars showing standard deviation for each timepoint. (A) shows the plasma nicotine profiles of the 7-mg nicotine-containing pouches. (B) shows the plasma nicotine profiles of the wintergreen-flavoured nicotine pouches.

Summary of baseline-adjusted nicotine pharmacokinetic parameters.

Data are presented as mean (coefficient of variation) for Cmax and AUCnic 0–180, and median (minimum and maximum) for Tmax.

Product Nicotine per pouch (mg) Cmax (ng/mL) AUCnic 0–180 (ng × min/mL) Tmax(min)
Velo Wintergreen 4 11.224 (28.7%) 1212.535 (27.4%) 46.0 (30.00–73.00)
Velo Max Wintergreen 7 16.807 (23.6%) 1773.454 (25.1%) 46.0 (30.00–90.00)
Velo Max Spearmint 7 16.991 (27.9%) 1 1862.770 (25.2%) 1 46.0 (30.00–90.00) 1
Velo Max Peppermint 7 17.476 (32.5%) 1 1894.044 (30.6%) 1 46.0 (30.00–90.00) 1
Velo Max Citrus Burst 7 16.220 (33.3%) 1 1752.167 (31.2%) 1 46.0 (15.00–90.00) 1
Velo Max Black Cherry 7 17.322 (28.0%) 1 1872.012 (25.9%) 1 46.0 (30.00–90.00) 1
Velo Max Coffee 7 16.354 (28.9%) 1 1829.397 (23.7%) 1 46.0 (15.00–90.00) 1
Velo Max Dragon Fruit 7 16.648 (29.8%) 1 1775.659 (28.8%) 1 46.0 (30.00–90.00) 1
Velo Max Cinnamon 7 17.888 (26.2%) 1965.491 (22.1%) 46.0 (30.00–90.00)

Abbreviations: Cmax: maximum plasma nicotine concentration; AUCnic 0–180: area under the plasma nicotine concentration-time curve between 0 and 180 min from the start of product use; Tmax: time of maximum plasma nicotine concentration and is the median of the actual recorded blood collection time. Non-missing data points = 41 in each case.

1 N = 40: Two participants voluntarily withdrew from the study, one participant on Day 1 and the second on Day 3.

Similarly, mean (CV) AUCnic 0–180 values were lowest for the Velo Wintergreen 4-mg pouch (1212.535 ng × min/mL (27.4%)) and both highest for all the Velo Max 7-mg pouches and similar between flavours, ranging from 1752.167 ng × min/mL (31.2%) for Velo Max Citrus Burst 7-mg and 1965.491 ng × min/mL (22.1%) for Velo Max Cinnamon 7-mg (Table 2).

Statistical comparison of Cmax and AUCnic 0–180 showed that both parameters were significantly different between the Velo Wintergreen 4-mg and Velo Max Wintergreen 7-mg NPs, but not between the Velo Max Wintergreen 7-mg NP and any of the other 7-mg study products (Table 3). Median Tmax values were identical at 46 min for all Velo NPs assessed, regardless of nicotine content or flavour (Table 2) and were not statistically significantly different between any of the study products (Table 3).

Statistical comparisons of nicotine pharmacokinetic parameters.

Comparison p value
Reference product Comparator product Cmax AUCnic 0–180 Tmax
Velo Max Wintergreen 7-mg Velo Wintergreen 4-mg < 0.0001 1 < 0.0001 1 0.63
Velo Max Wintergreen 7-mg Velo Max Spearmint 7-mg 0.19 0.13 0.99
Velo Max Wintergreen 7-mg Velo Max Peppermint 7-mg 0.19 0.12 0.26
Velo Max Wintergreen 7-mg Velo Max Citrus Burst 7-mg 0.67 0.45 0.65
Velo Max Wintergreen 7-mg Velo Max Black Cherry 7-mg 0.10 0.10 0.36
Velo Max Wintergreen 7-mg Velo Max Coffee 7-mg 0.83 0.35 0.50
Velo Max Wintergreen 7-mg Velo Max Dragon Fruit 7-mg 0.63 0.85 0.42
Velo Max Wintergreen 7-mg Velo Max Cinnamon 7-mg 0.015 0.003 0.81

1 Value indicates statistical significance.

Comparisons of Cmax and AUCnic 0–180 were performed using paired f-tests on a log-transformed scale. Tmax comparisons were made using a sign test on the original data scale. Bonferroni adjustment for multiple comparisons was performed such that a p value of < 0.002 indicated statistical significance. Abbreviations: Cmax: maximum plasma nicotine concentration; AUCnic 0–180: area under the plasma nicotine concentration-time curve between 0 and 180 min from the start of product use; Tmax: time of maximum plasma nicotine concentration.

Subjective effects

Data for the product-liking subjective effect assessment are presented in Table 4. Broadly speaking, mean scores for product-liking were similar for all Velo NPs assessed, regardless of flavour and nicotine content. Mean (SD) product-liking scores ranged from 5.1 (2.28) for the Velo Max Black Cherry 7-mg pouch to 6.1 (2.88) and 6.1 (2.43) for the Velo Max Coffee and Velo Max Dragon Fruit 7-mg pouches, respectively.

Summary of overall product-liking questionnaire scores.

Product Nicotine per pouch (mg) Number of participants Mean (SD) Median Min, max Q1, Q3
Velo Wintergreen 4 41 5.8 (2.22) 6 0, 10 4, 7
Velo Max Wintergreen 7 41 5.9 (2.25) 6 1, 10 4, 8
Velo Max Spearmint 7 40 5.6 (2.30) 6 0, 10 5, 7
Velo Max Peppermint 7 40 5.3 (2.13) 6 0, 10 4, 7
Velo Max Citrus Burst 7 40 6.0 (2.52) 6 0, 10 4, 8
Velo Max Black Cherry 7 40 5.1 (2.28) 5 0, 10 4, 7
Velo Max Coffee 7 40 6.1 (2.88) 6.5 0, 10 5, 8
Velo Max Dragon Fruit 7 40 6.1 (2.43) 6 0, 10 5, 8
Velo Max Cinnamon 7 41 5.5 (2.67) 6 0, 10 4, 7

The numerical score scale was from 0 to 10, with 0 corresponding to “strong disliking”, 5 corresponding to “neither like nor dislike” and 10 corresponding to “strong liking”.

Abbreviations: SD: standard deviation; Min: minimum; Max: maximum; Q1: first quartile; Q3: third quartile.

Adverse events

There were no serious or severe product-emergent adverse events (PEAEs) and no participants were withdrawn from the study for safety reasons. A total of 90 PEAEs were experienced by 28 (66.7%) of the 42 participants. Of these PEAEs, seven in total were reported by six participants (14.3%) following the use of Velo Max Peppermint during the product familiarisation session, this being the only product used during this session.

During the test sessions, the incidence of participants with PEAEs ranged from 25.0% for Velo Max Black Cherry 7-mg to 7.3% for Velo Max Wintergreen 7-mg. PEAE occurrences during the test sessions ranged from 15 follow ing use of Velo Max Peppermint 7-mg to 5 following the use of Velo Max Wintergreen 7-mg and Velo Max Spearmint 7-mg. Overall, the PEAE experienced most commonly was hiccups, reported by 5 participants (12.2%) after use of Velo Max Cinnamon 7-mg, 3 participants (7.5%) after use of Velo Max Black Cherry 7-mg, 3 participants (7.5%) after use of Velo Max Peppermint 7-mg, and 1 participant each after use of Velo Max Dragon Fruit 7-mg, Velo Max Wintergreen 7-mg, Velo Wintergreen 4-mg (2.4% each), and Velo Max Coffee 7-mg (2.5%). Of the participants who experienced at least one PEAE, most experienced PEAEs which were mild in severity (52.4%) while the remaining experienced PEAEs were moderate in severity (14.3%). A total of 21 participants (50.0%) experienced 57 product use-related PEAEs (categorised as either related or possibly related).

DISCUSSION

The present randomised crossover clinical study examined nicotine pharmacokinetics and product-liking among cigarette smokers who used 9 different Velo NPs during 9 separate 45 min use sessions. We found that the increase in plasma nicotine concentration, both in terms of Cmax and AUCnic 0–180, was greater when participants used Velo Max NPs containing a higher nicotine content (7-mg) compared to a Velo NP containing lower nicotine content (4-mg). However, median Tmax values did not differ between the NPs with different nicotine content. Importantly, we also found that when participants used Velo Max NPs with the same 7-mg nicotine content but with different flavours, Cmax, AUCnic 0–180 and Tmax were not significantly impacted. Lastly, product-liking was similar across all Velo NPs assessed regardless of nicotine content and flavour.

In the past years, several studies have been published on NP pharmacokinetics. Rensch et al. (31) described data from a study similar to the one reported here and examined nicotine pharmacokinetics in smokers when they either smoked a combustible cigarette or used a single NP containing approximately 3–4 mg nicotine for a 30-min period. Cmax values ranged from approximately 9–12 ng/mL, and these values were similar to those seen in our study for the Velo Wintergreen 4-mg pouch (Cmax was approximately 11 ng/mL). In addition, McEwan et al. (27) also described data from a study similar to the present one where the nicotine pharmacokinetics were studied in smokers using several commercially available NPs with nicotine content ranging from 6–10 mg nicotine for a 60-min period. In this case, Cmax values ranged from 11.9–18.4 ng/mL with three of these NPs containing between 6–10 mg nicotine having a similar Cmax to the Velo Max NPs containing 7 mg nicotine. However, this study also suggested that the physical design characteristics of these different NPs produced by different companies may have an effect on the nicotine pharmacokinetics and subjective effects. This might also explain a further pharmacokinetic study in smokers that used a 4-mg nicotine-containing NP with 60-min use time which reported a Cmax of 8.5 ng/mL and thus is lower than the level reported for the 4-mg nicotine NP reported in this study, even with a longer usage time (28). Finally, a study by Lunell et al. (21) assessed NP nicotine pharmacokinetics in snus users when they used an NP containing either 3, 6 or 8 mg nicotine or used snus products containing 8 or 18 mg nicotine, for a 60-min period. Mean Cmax for a 3-mg NP (7.7 ng/mL) was slightly lower than that seen for the 4-mg Velo NP assessed in our study (approximately 11 ng/mL) while the mean Cmax for the 6-mg (14.7 ng/mL) and the 8-mg (18.5 ng/mL) NPs (21) were similar to those in our study for the 7-mg NPs assessed (range approximately 16–18 ng/mL). Tmax values for all of the studies discussed including the present study were closely associated with the end of the product use time period (21, 23, 27, 31). Due to methodological differences in how the AUC-values were estimated, these cannot be compared.

A similar product to NPs is snus, a smokeless tobacco product which is accepted to present a lower risk to users than smoking combustible cigarettes (20, 32, 33). Examination of the literature concerning snus gives insight into the potential of NPs to be a reduced-risk alternative to tobacco products as NPs are similar in physical form to snus but do not contain tobacco.

In a study among snus users, Lunell et al. (21) compared nicotine pharmacokinetics between NPs and snus, reporting that the AUC was greater for an NP with 6 mg nicotine than for snus with 8 mg nicotine, indicating that NPs may be able to provide nicotine more efficiently than snus. In addition, extraction of nicotine during use seems to be more effective from NPs (58% on average over 60 min) (21, 23, 28) compared with 33% on average from snus (23). Therefore, NPs deliver slightly higher amounts of nicotine compared to snus, a product recognised as a reduced risk alternative to smoking that reduces exposure to harmful toxicants (33, 34) and produces beneficial changes in biomarkers of potential harm in smokers who switch to exclusive use of snus (35).

It should be noted that the pharmacokinetic studies discussed here controlled participants’ NP product usage time and that consumers will adjust usage times to suit their preferences.

An interesting facet of our data is the lack of an impact of flavour on nicotine pharmacokinetics of NPs. This was also noted in the study by Rensch et al. (31) where no significant impact of NP flavour on nicotine pharmacokinetics was reported. It has been suggested that menthol may increase the absorption of nicotine and toxicants associated with pouched tobacco products such as Swedish snus. This is described as increased rate of permeation and was studied in a porcine oral mucosa model by Squier et al. (36), which showed a significant increase in nicotine permeation in the presence of 0.08% menthol. However, the data from this study show that, contrary to this, there is no notable difference in nicotine absorption during use of NPs containing menthol, which is found in the tested Wintergreen, Spearmint, and Peppermint flavoured NPs, as compared to the other flavours tested.

The present study has some limitations. First, it was conducted among cigarette smokers in the U.S.; thus, the findings might not be generalisable to other groups (e.g., users of other nicotine products such as snus), or to other countries where patterns of tobacco and nicotine product use might vary. Second, the data were gathered from use of a single NP product for a fixed amount of time on a single day after overnight abstinence from nicotine. For normal everyday use plasma nicotine concentrations will be influenced by product use duration, as well as by factors such as product nicotine content, proportion of nicotine extracted from the product during use, average daily consumption, and the way in which the product is used by the consumer.

Average daily consumption is a particularly important factor in assessing daily nicotine exposure and blood plasma concentrations. Regarding NPs, an average daily consumption of 8.6 pouches per day among solus NP users has been reported, compared with 14 cigarettes per day reported among solus smokers in the same study (23). However, further studies are needed to establish daily nicotine exposure and the resulting plasma nicotine concentrations associated with NP use, as well as consumer behaviour when using NPs, in order to better inform of their tobacco harm reduction potential (37).

CONCLUSION

In conclusion, the present data provide important insight into nicotine delivery and pharmacokinetics in current smokers during use of NPs with varying nicotine content and various flavours. We demonstrate that Velo NPs deliver nicotine to a degree comparable with cigarette smoking but with slower uptake and that flavours do not have an impact on NP nicotine uptake.

Further, we also demonstrate that the use of Velo NPs was associated with a strong degree of product-liking. Overall, data from our study are broadly similar to those reported by others and support the idea that tobacco-free NPs deliver similar levels of nicotine to those achieved during cigarette smoking and may therefore provide a suitable alternative form of nicotine delivery for current smokers. Further studies are required to investigate the potential role of NPs in THR.

eISSN:
2719-9509
Language:
English
Publication timeframe:
4 times per year
Journal Subjects:
General Interest, Life Sciences, other, Physics