4,039
Views
1
CrossRef citations to date
0
Altmetric
Review Article

Review of biomarkers to assess the effects of switching from cigarettes to modified risk tobacco products

, , , &
Pages 213-244 | Received 06 Sep 2017, Accepted 13 Dec 2017, Published online: 09 Jan 2018

Abstract

Context: One approach to reducing the harm caused by cigarette smoking, at both individual and population level, is to develop, assess and commercialize modified risk alternatives that adult smokers can switch to. Studies to demonstrate the exposure and risk reduction potential of such products generally involve the measuring of biomarkers, of both exposure and effect, sampled in various biological matrices.

Objective: In this review, we detail the pros and cons for using several biomarkers as indicators of effects of changing from conventional cigarettes to modified risk products.

Materials and methods: English language publications between 2008 and 2017 were retrieved from PubMed using the same search criteria for each of the 25 assessed biomarkers. Nine exclusion criteria were applied to exclude non-relevant publications.

Results: A total of 8876 articles were retrieved (of which 7476 were excluded according to the exclusion criteria). The literature indicates that not all assessed biomarkers return to baseline levels following smoking cessation during the study periods but that nine had potential for use in medium to long-term studies.

Discussion and conclusion: In clinical studies, it is important to choose biomarkers that show the biological effect of cessation within the duration of the study.

Introduction

The Biomarkers Definition Working Group in 2001 defined a biomarker as “a characteristic that can be objectively measured and evaluated as an indicator of normal conditions and disease processes or of pharmacological responses” (Biomarkers Definitions Working Group Citation2001). Clinical biomarkers can be grouped by function.

Diagnostic – for the identification of disease or abnormal pathology, differential diagnosis and monitoring the response to therapy (Hijazi et al. Citation2013, Holdenrieder Citation2016).

Prognostic – as an indicator of disease prognosis. In oncology it is generally accepted that prognostic markers are patient/tumour factors that affect patient outcome independently of treatment administered (Tsao et al. Citation2015).

Predictive – a biomarker is predictive if the treatment effect is different for biomarker-positive patients compared with biomarker-negative patients (Ballman Citation2015, Tsao et al. Citation2015).

Most of the biomarkers in this review can be considered biomarkers of effect. They consist of measurable biochemical, physiologic, behavioural or other alteration in an organism that, depending on the magnitude, can be associated with an established or possible health impairment or disease. Some may also be considered biomarkers of exposure: a chemical, its metabolite or the product of an interaction between a chemical and some target molecule or cell that is measured in the human body (Committee on Human Biomonitoring for Environmental Toxicants Citation2006).

In summary, disease-related biomarkers give an indication of the probable effect of treatment on a patient (predictive biomarker), if a disease already exists (diagnostic biomarker) or how such a disease may develop in an individual case regardless of the type of treatment (prognostic biomarker) (Ballman Citation2015).

Biomarkers have demonstrated their value in early efficacy and safety evaluation and met scientific acceptance in basic and clinical research as primary endpoints for evaluation. The relevance of specific biomarker use is strengthened through evidence showing repeatedly to predict clinical outcomes (Strimbu and Tavel Citation2010). There is consensus that such surrogates for clinical outcomes require rigorous qualification and assessment. But even when this has been done, in line with best available knowledge, failures in clinical studies can occur. For example, suppression of arrhythmias as a surrogate endpoint for decreased morbidity due to cardiovascular disease (CVD) resulted in the approval of two drugs that in later studies were shown to increase morbidity (Fleming and DeMets Citation1996). In many instances, not all treatment effects are fully accounted for by a single biomarker (Biomarkers Definitions Working Group Citation2001). Therefore, for complex diseases, such as Chronic Obstructive Lung Disease (COPD), it has been suggested that a panel of biomarkers might prove more accurate (Leung and Sin Citation2013, Sun et al. Citation2016) and that a selection of biomarkers might increase the probability of success in clinical trials (Carroll Citation2016).

Expert opinion is unanimously in support of the fact that smoking cessation is the best way to reduce the risks of cigarette smoke-related diseases including COPD, CardioVascular Disease (CVD) and lung cancer. Smokers who stop smoking between the ages of 25–34 years do not differ in their life expectancy to those who have never smoked (Jha et al. Citation2013). For many smokers looking for better alternatives to cigarettes, the use of non-burning or smokeless tobacco products offers the possibility to consume nicotine with reduced risk to their health (Nutt et al. Citation2014, Hilton et al. Citation2016). What is already known from the plethora of existing data about toxicant exposure and subsequent health consequences of combustible tobacco consumption renders the use of clean nicotine in comparison to tobacco smoke a means to reduce harm (Cahill et al. Citation2013). However, the increased use by consumers of alternative, reduced risk nicotine products warrants confirmation and substantiation of the risk profile of this new product class. This substantiation requires the study of health outcomes in relation to product use in comparison to complete abstinence or continued smoking of conventional products. A number of biomarkers of biological effect and risk have been proposed to provide information about the risk profile of alternative tobacco products in the absence of, or not yet available, data of long-term use of the new nicotine-containing products. The objective of this publication is to report the results based on the analysis of data pertaining to a large number of biomarkers that have previously been used in studies investigating the effects of cigarette smoke and in studies where subjects have stopped smoking or switched to a reduced risk product (RRPs – products that present, are likely to present or have the potential to present less risk of harm to smokers who switch to these products versus continued smoking). A biomarker fit for purpose to substantiate reduced risk should be associated with the smoking of combustible cigarettes, decline following smoking cessation and/or smoking reduction and should be associated with at least one smoking-related disease (Biomarkers Definitions Working Group Citation2001, Strimbu and Tavel Citation2010).

Clinical significance

This review provides information related to a number of biomarkers that have been used to assess the effects of cigarette smoking. Many of them are used to monitor changes that occur following cessation (or switching to a modified risk product). To have meaningful studies with reliable outcomes, it is important to carefully select the biomarkers to be measured and the duration of the study. Here we provide evidence of biomarkers that do change following cessation and the time within which such a change occurs.

Methods

The PubMed database was searched for publications related to the biomarkers listed in . Three searches of the PubMed literature data base were conducted for English language articles within the time frame of 2008 to May 2017 for each of the biomarkers of interest.

Table 1. List of biomarkers that were searched in the PubMed database and published between 2008 and 2017.

Search 1: To identify literature that linked smoking, nicotine replacement or cessation to the biomarker using the search string: (biomarker) AND (smokers OR smoking OR nicotine replacement OR cessation) AND (“2008”[Date - Publication]: “2017”[Date - Publication]) AND English[Language]).

Search 2: To identify literature reporting an association between the biomarker and cardiovascular disease using the search string: (biomarker) AND ((cardiovascular disease) AND (association)) AND (“2008”[Date - Publication]: “2017”[Date - Publication]) AND English[Language]).

Search 3: To identify literature reporting an association between the biomarker and respiratory disease using the search string: (biomarker) AND ((respiratory disease) AND (association)) AND (“2008”[Date - Publication]: “2017”[Date - Publication]) AND English[Language]).

A set of specific criteria was applied to the identified studies to determine which papers should be selected for inclusion:

  • Studies investigating the association of cigarette smoking, smoking cessation or nicotine replacement therapy (NRT) with the biomarker;

  • Studies investigating the association of the biomarker with CVD or respiratory disease.

The following exclusion criteria were applied:

  •  E1. Studies that used the measurement of the biomarker only for control purposes (e.g. results were adjusted for differences in the biomarker between populations, but the biomarker itself was not an endpoint);

  •  E2. Studies involving dietary, pharmacological or exercise interventions in which the biomarker was an endpoint, but was lacking a clear link between the biomarker and either a disease of relevance (CVD, COPD, lung cancer) or smoking;

  •  E3. Studies of passive or parental smoking;

  •  E4. Disease not covered by the article;

  •  E5. Studies focussing on a narrow population (e.g. pregnant women, individuals with a particular haplotype, individuals with a specific disease) or studies limited to human-derived tissue (e.g. retina, gums);

  •  E6. Studies reporting results from in vivo animal experiments;

  •  E7. Studies in which the biomarker was measured for toxicological or forensic purposes;

  •  E8. Studies with no comparison between smoking and non-smoking/switching/cessation;

  •  E9. The biomarker was not measured or was presented in a fashion that made the data unusable.

The number of articles retrieved for each biomarker in each of the three searches and the numbers excluded according to the criteria (E1–E9 listed above) are presented in .

Table 2. Numbers of publications (2008–2017) and the reasons for their exclusion.

The information retrieved for each biomarker is presented in the Findings section as a summary of the biology, pathological role and an indication of the validity of the biomarker based on the information available in the literature, although it is important to note that for many of the biomarkers discussed below literature published prior to 2008 has been cited. This was done when it was not possible to reach a meaningful conclusion based on more recent data.

For some of the biomarkers, data were retrieved by searching monographs and reviews and extracting relevant literature from them. The number of articles found, and the numbers excluded for these biomarkers is presented in .

Table 3. Literature retrieved from monographs and review articles.

Findings

Respiratory biomarker

Forced expiratory volume in 1 s (FEV1)

The respiratory disease that is most commonly associated with cigarette smoking is chronic obstructive pulmonary disease (Ford et al. Citation2013) (COPD) which is defined by a reduction in airflow that is not entirely reversible. Current diagnostic practice recommends that the obstruction be confirmed by the use of a post-bronchodilator forced expiratory volume in 1 s (FEV1) to forced vital capacity (FVC) ratio of less than 0.7. (Vestbo et al. Citation2016) although it is accepted that this fixed ratio may lead to an underestimation of airflow obstruction in younger subjects and an over-diagnosis in the more elderly (partly attributable to normal aging) (Cerveri et al. Citation2008, Runarsdottir et al. Citation2013, van Dijk et al. Citation2015, Woodruff et al. Citation2016).

The published data suggest that the link between cigarette smoking and annual decline in FEV1 is moderate with one recent study showing a peak in the rate of decline of 53.8 ml per year in subjects with COPD GOLD grade 1 (Bhatt et al. Citation2016), and another showing that at age 45 years the estimated rate of FEV1 decline was larger in current smokers than former and never smokers (Mirabelli et al. Citation2016) and a reduction in FEV1 is generally observed with increasing smoking history (Tommola et al. Citation2016). For each smoking history, however, there is considerable heterogeneity in FEV1 response. Also among non-smokers, there is a subset with severe lung function impairment that will be diagnosed as COPD due to exposure to wood smoke, traffic pollutants, environmental tobacco smoke and occupational exposures etc. (Hu et al. Citation2010, Moreira et al. Citation2013, Diaz-Guzman and Mannino Citation2014, Pallasaho et al. Citation2014, Ramirez-Venegas et al. Citation2014).

Through years of regulatory practice, the FDA accepts the use of FEV1 as a surrogate endpoint of disease status. All the articles in which smokers were compared to non-smokers showed that smoking decreased FEV1 over time to a greater extent than the decrease attributable to aging.

The landmark lung health study between 1986 and 1994 (U.S./Canadian Lung Health study) (Anthonisen et al. Citation1994), involving almost 6000 subjects, showed that subjects who stopped smoking for 1 year had improved lung function.

The current literature search identified some studies reporting a short-term improvement in FEV1 after smoking cessation. However, this was not consistently the case, probably due to the relatively large variability (Becklake Citation1986, Crapo and Jensen Citation2003) of this marker.

Several studies have shown that subclinical inflammatory changes in small airways exist years before the advanced stages of COPD (Cosio et al. Citation1978, Guy et al. Citation1994, Takizawa et al. Citation2000, Verbanck et al. Citation2004). Although relatively few longitudinal studies additional to the lung health study have assessed the effects of smoking cessation on inflammation in smokers without chronic respiratory symptoms, these studies demonstrated that the inflammatory response decreased rapidly within the first few months after smoking cessation. It seems likely that the favourable changes in lung function as assessed by FEV1 in studies of less than 1-year duration may be best explained by a decrease in the inflammatory state of the lung following quitting smoking.

FEV1, which is the accepted marker for COPD progression, will have its role in the assessment of novel risk reduced tobacco products although studies will need to be of fairly large size to account for the inherent variability of the measure. They will also need to be of longer duration. It may be that after two decades the time has come to reproduce the results of the lung health study since there have been many changes in the pattern of tobacco consumption and products on the market in the intervening years. Although lung function remains stable during the years between the age of 20–35, FEV1 then declines due to the natural aging processes (with an accelerated decline after the age of 70 years) (Sharma and Goodwin Citation2006, Skloot Citation2017). Ethnicity and gender also affect spirometric measures of pulmonary function (Quanjer et al. Citation2012a, Coates et al. Citation2016, Mirabelli et al. Citation2016, LaVange et al. Citation2017)

The small airways have been proposed to be the major site of increased airway resistance in COPD (Hogg et al. Citation1968, Flenley Citation1988, Hogg et al. Citation2004, Burgel Citation2011, Hogg et al. Citation2013) inducing a slowing in the terminal portion of the spirogram interpreted as a reduction in the mean expiratory flow between 25 and 75% of the forced expiratory flow (FEF25–75%). Although it is thought not to contribute to clinical decision making (Quanjer et al. Citation2012b, Citation2014) several reports have shown that FEF25–75% is reduced in smokers compared to non-smokers (Casale et al. Citation1991, Urrutia et al. Citation2005, Bird and Staines-Orozco Citation2016) with one study showing a dose–response between smoking and lower FEF25–75% (Gold et al. Citation1996). In a study in which smokers were invited to switch to an e-cigarette and abstain from smoking, the FEF25–75% increased significantly (p = 0.034) from 85.7% to 100.8% predicted over 52 weeks (Cibella et al. Citation2016). There are, however, several reasons why the FEF25–75% may not be useful: it is dependent upon the FVC, it lacks the repeatability of FEV1, it has a wide normal range (Quanjer et al. Citation2012b, Al Ghobain et al. Citation2014, Balasubramaniam et al. Citation2014, Gutierrez et al. Citation2014, Rufino et al. Citation2017) and it is reduced in the presence of proximal airway narrowing (Johns et al. Citation2014). These drawbacks severely limit the use of FEF25–75% for diagnostic purposes, but might not preclude its use in aetiological studies where differences between groups may be valuable. However, without clear validation FEF25–75% cannot be recommended as a biomarker for use in short-term smoking/cessation/switching studies.

Biomarkers of oxidative stress

In eukaryotic organisms, more than 90% of reactive oxygen species (ROS) are produced by the mitochondria, via the mitochondrial electron transport chain (Skulachev Citation2012, Sies Citation2014) although smaller amounts are also produced by electron transport chains in endoplasmic reticular (Brignac-Huber et al. Citation2011), nuclear (Vartanian and Gurevich Citation1989) and plasmatic (Luthje et al. Citation2013) membranes. When the levels of these free radicals become sufficient to overwhelm the antioxidant defence mechanisms then “oxidative stress” occurs. Oxidative stress and the effects it causes, such as lipid peroxidation, are involved in a number of smoking-related diseases including atherosclerosis, cancer, inflammation and COPD (Barrera Citation2012, McGuinness and Sapey Citation2017) and in diseases such as accelerated aging and depressive disorders (Maurya et al. Citation2016). Furthermore, the chronic inflammation associated with these diseases also induces oxidative stress (Thanan et al. Citation2014).

Cigarette smoke exposure elicits a series of adaptive cellular stress responses (Bialas et al. Citation2016, Jiang et al. Citation2017) and a number of biomarkers have been evaluated as surrogate measures for these responses. These are discussed below although it is unlikely that a single biomarker would be capable of reflecting the complexity of cellular stress responses to exogenous stressors such as cigarette smoke in man.

8-Iso-15(S)-prostaglandin F (8-iso-PGF)

The isoprostanes are prostaglandin (PG)-like compounds that are formed from the peroxidation of the ubiquitous polyunsaturated fatty acid, arachidonic acid. Unlike PGs, however, which are formed via the action of the cyclooxygenase enzymes, isoprostanes are generated as a result of the free radical-mediated peroxidation of arachidonic acid (Morrow et al. Citation1990) although there is increasing evidence that they may be formed via the actions of PG endoperoxide synthase enzymes which are induced during inflammation (van't Erve et al. Citation2015, Citation2016)

The 2001 Institute of Medicine report (Stratton et al. Citation2001) suggests the measurement of one of the four positional/regioisomers of 8-iso-PGF (also termed 8-epi-PGF), for the assessment of oxidative damage. For many years, 8-iso-PGF has been regarded as the gold standard for the detection of oxidative stress (Morrow Citation2005, Ho et al. Citation2013a)

This literature search provided no evidence of a strong link between 8-iso-PGF and smoking (e.g. urinary 8-iso-PGF levels were 0.73 µg/g creatinine (95% CI, 0.61–0.87) in non-smokers and 0.86 µg/g creatinine (95% CI, 0.70–1.05) in smokers (Calapai et al. Citation2009) contrary to some earlier publications (Reilly et al. Citation1996) where smokers had a pre-cessation mean of 145.5 ± 24.9 compared to non-smokers 63.7 ± 5 pmol/mmol creatinine 8-iso-PGF in urine. In heavy smokers’, urinary 8-iso-PGF levels fell to 114.6 ± 27.1 after 2 weeks of cessation. A 90-day study in Japanese subjects found that switching from menthol cigarettes (mCC) to a menthol tobacco heating system (mTHS) product resulted in a 12.7% lower urinary 8-iso-PGF concentration compared to continued mCC users (Lüdicke et al. Citation2017). A recent meta-analysis of over 200 publications found that tobacco smoking caused a relatively small increase in free 8-iso-PGF (van't Erve et al. Citation2017) which the authors note as interesting for a condition that has long been thought to have a high level of oxidative damage. Given that “for a biomarker of exposure to be accepted as a biomarker of risk or a surrogate endpoint of disease, there should be a strong biological rational as well as compelling data from clinical and epidemiologic studies”(Committee on Scientific Standards for Studies on Modified Risk Tobacco Products Citation2012) it would appear that although 8-iso-PGF, as a marker of oxidative stress (and a specific marker for lipid peroxidation), is valuable in establishing a diagnosis or assessing disease progression due to oxidative stress (Janssen Citation2001), it may not be suitable for the assessment of cigarette smoking cessation in the short term, i.e. less than 3 months’ duration, since the current literature search provided no substantial evidence of a strong direct link between 8-iso-PGF and smoking cessation except after long periods of time (Harman et al. Citation2003, Lowe et al. Citation2009).

Contrary to the current literature search, one earlier publication has shown that urinary 8-iso-PGF levels can decrease rapidly following smoking cessation (Morita et al. Citation2005). A small (27 healthy, smoking medical students) study that had no non-smoking controls; another study that compared circulating levels of 8-iso-PGF in smokers and non-smokers (242 ± 147 and 103 ± 19 pmol/L, respectively) with levels falling to 156 ± 67 pmol/L after 2 weeks of smoking cessation (Morrow et al. Citation1995) and a very small study (heavy smokers n = 5; moderate smokers n = 5; non-smokers n = 14) that showed 8-iso-PGF levels falling within 3 weeks of cessation but failing to reach levels seen in non-smokers (Reilly et al. Citation1996). Normal plasma levels of 8-iso-PGF are affected by ethnic background with one study showing that West African immigrants had lower 8-iso-PGF levels than US-born African Americans who themselves had lower levels than non-Hispanic white Americans (Annor et al. Citation2017). Similar differences have been reported for urinary levels of 8-iso-PGF (Il'yasova et al. Citation2012)

Thromboxane B2 (TXB2)

A potentially important platelet-derived marker is thromboxane A2 (TXA2). TXA2 is generated in platelets from its precursor prostaglandin H2 by the action of the enzyme thromboxane synthase, which in turn is formed by the action of cyclooxygenase-1 on arachidonic acid (Moore Citation1982). Its importance is attributed to the fact that, amongst other actions, it is a vasoconstrictor, a potent hypertensive agent and it can initiate the release reaction which is followed by platelet aggregation (Gryglewski et al. Citation1978). The literature indicates that increased levels of TXA2 are associated with increased risk of atherogenesis (Dogne et al. Citation2005, Nakahata Citation2008). TXA2 is highly unstable; thus, its levels are measured using two metabolites related to thromboxane B2 (TXB2) which are 11-dehydro-TXB2 and 2,3-dinor-TXB2 (Catella et al. Citation1986). The literature search indicated that TXB2 may be a valid biomarker; therefore, a second literature search was conducted covering years prior to 2008. In this search, 14 cross-sectional studies were identified that compared levels of one or both of the TXB2 metabolites in smokers and non-smokers, and these are listed in .

Table 4. Association of TXB2 metabolites with smoking status.

As can be seen from , all 14 studies showed that one or both TXB2 metabolites were elevated in smokers compared to non-smokers. For 12 of these studies, the results were statistically significant at the 5% level. Study 10 (McAdam et al. Citation2005) did not provide a p value, while the difference reported by study 14 (Prasad et al. Citation2016) was found not to be statistically significant. Of particular interest are the results of study 13 (Frost-Pineda et al. Citation2011), the only large study, where the difference was statistically significant with p < 0.0001. Therefore, published evidence would appear to support the view that one or both TXB2 metabolites are elevated in smokers compared to non-smokers.

To assess the potential utility of TXB2 metabolites for the assessment of RRPs, it is also essential that their levels have been found to decline following smoking cessation. Only three studies were found that determined the levels of at least one of the TXB2 metabolites following smoking cessation and the results from these can be seen in .

Table 5. Changes in levels of TXB2 metabolites following smoking cessation.

Although only three studies were found, they all showed a statistically significant decrease in the two TXB2 metabolites following smoking cessation. These data support that either 2,3-dinor-TXB2 or 11-dehydro-TXB2 is a potentially useful biomarker for the evaluation of reduced risk products. This conclusion is substantially reinforced by two studies where 11-dehydro-TXB2 was used to evaluate such a product. One study (Roethig et al. Citation2008) evaluated the change in 11-dehydroxy-TXB2 levels in a 12-month randomized controlled trial comparing smokers who switched to a second-generation electrically heated cigarette smoking system (N = 50) with those who continued smoking conventional cigarettes (N = 32). There was a 20% decrease for those subjects who switched compared to a slight increase for those subjects who continued to smoke conventional cigarettes (p = 0.003). Martin Leroy et al. (Citation2012) reported the results of a similar randomized controlled trial where 237 smokers were switched to an electrically heated cigarette smoking system and 79 subjects continued to smoke their own brand. In this 1-month study, there was a decrease of 10.2% in the median level of 11-dehydro-TXB2 in subjects who switched compared to a 2.9% decrease in those subjects who continued to smoke (p < 0.001).

The last requirement to qualify TXB2 metabolites as useful biomarkers in studies examining reduced risk products is that they are associated with at least one smoking-related disease. There is clear evidence that this is the case for CVD, since TXB2 metabolites are markers of platelet activation and aggregation (Sharma and Berger Citation2011). On the other hand, there is no evidence that TXB2 metabolites would be a useful marker for smoking-related respiratory diseases.

Biomarkers of inflammation

The 2014 U.S. Surgeon General’s Report “The Health Consequences of Smoking: 50 Years of Progress” (US Department of Health and Human Services Citation2014) identifies inflammation (and oxidative stress) among others, as key mechanisms underlying all major smoking-related diseases, and there is ample evidence showing that both mechanisms are intricately linked to each other and to disease development in smokers. Cigarette smoking is causally linked to the development of CVD contributing to endothelial injury and dysfunction, a pro-atherogenic lipid profile, chronic inflammation and an abnormally increased tendency toward coagulation. COPD is characterized by an abnormal inflammatory response and is linked to a number of sustained cellular stress responses including endoplasmic reticulum, hypoxic stress and autophagic stress (US Department of Health and Human Services Citation2014). Furthermore, most of the inflammatory processes identified in smokers with COPD can also be observed in smokers with lung cancer (Yanbaeva et al. Citation2007, Faux et al. Citation2009, Mons et al. Citation2016), suggesting a shared aetiology. Given the importance of inflammation in the development of smoking-related diseases (Hamilton and Rath Citation2015, Bozinovski et al. Citation2016, Zaynagetdinov et al. Citation2016) several inflammation-related biomarkers have been proposed.

C-reactive protein (CRP)

C-reactive protein (CRP) increases in response to a wide range of acute and chronic inflammatory conditions such as bacterial, viral or fungal infections as well as rheumatic and other inflammatory diseases, malignancies and tissue injury and necrosis. The literature searches demonstrated that CRP is a well-established biomarker of inflammation, suitable for both clinical purposes and epidemiological studies. The association between CRP and the risk for CVD has been documented in many large, prospective epidemiological studies, although some meta-analyses appear to de-emphasize the significance of CRP as a prognostic marker (Adukauskiene et al. Citation2016). Furthermore, CRP has also been reported to be predictive for respiratory diseases (Danesh et al. Citation2004, Pepys Citation2005, Lowe and Pepys Citation2006) and various types of cancer, particularly of lung cancer (Agarwal et al. Citation2013, Firouzjahi et al. Citation2013, Deng et al. Citation2014, Kawada Citation2015). Numerous studies have produced a large variation in what normal levels of CRP should be (Zhang et al. Citation2016, Zhu et al. Citation2016). CRP levels in smokers have been shown in one study to be 2.53 mg/L compared to 1.35 mg/L in non-smokers (p < 0.0001) (Tonstad and Cowan Citation2009) with levels falling within 5 years of smoking cessation but taking over 20 years to revert to levels of never smokers. Similar results were found in another study with never smokers having CRP levels of 1.13 mg/L while even light smokers had 1.87 mg/L (p < 0.001) (Wannamethee et al. Citation2005). At present, there is consensus that CRP is an indicator for acute and chronic inflammation processes rather than being directly involved in the pathophysiological processes of CVD, COPD or cancer. A major limitation of the suitability of CRP as a biomarker of effect for the evaluation of reduced risk tobacco products would be the very long period of time required to achieve non-smoker levels after switching (Lowe et al. Citation2001). CRP levels increase with increasing age (Wyczalkowska-Tomasik et al. Citation2016) and levels differ between male and female subjects (Woloshin and Schwartz Citation2005, Siemons et al. Citation2014). Ethnic background also affects this biomarker with percentage of subjects with levels >3 mg/L being 31, 40, 51 and 58% in white men, black men, white women and black women, respectively (Khera et al. Citation2005).

White blood cell count (WBC)

White blood cell count (WBC) is elevated in cigarette smokers (5–30% higher than non-smokers) (Asthana et al. Citation2010, Shiels et al. Citation2014, Kryfti et al. Citation2015) and has been proposed as a biomarker of cigarette smoke-related cardiovascular risk (Fernandez et al. Citation2012). Elevated peripheral WBC levels are also an important determinant of the level of pulmonary function (Korhonen et al. Citation2011). The literature provides confirmation that a fall in WBC count occurs rapidly following smoking cessation with one study indicating that a significant decrease in WBC count occurred after only 3 days of smoking cessation [from 8.73 × 109/L to 8.15 × 109/L (p = 0.0001) (Sparrow et al. Citation1984)]. In one cross-sectional study, the WBC count fell from 6.03 × 109/L to 5.64 × 109/L (p = 0.00009) within 1 year of smoking cessation whereas continuing smokers had WBC counts of 6.09 × 109/L (Roethig et al. Citation2010). The literature searches confirm that WBC count can be used as a fully validated biomarker of inflammation and thus diseases associated with it. There is a very large data set [e.g. (Higuchi et al. Citation2016) and a meta-analysis accessible on the authors web site (Holt Citation1987, Yanbaeva et al. Citation2007)] that establishes that cigarette smoking significantly increases WBC counts, and there is also a very large data set (Lee et al. Citation2014) showing that there is a strong association between the level of WBC count and various CVD endpoints, particularly coronary heart disease (CHD). There is also a reasonable amount of literature confirming an association of WBC count with the other two major smoking-related diseases – lung cancer (Danesh et al. Citation1998, Pearson et al. Citation2003, Madjid et al. Citation2004) and COPD (Allin et al. Citation2016). WBC numbers are generally higher in males than females (Ruggiero et al. Citation2007, Nilsson et al. Citation2009) and there are slight decreases in the elderly (>65 years). The NHANES study found that there were differences in the mean WBC count between racial groups, between sexes and among age groups within race-sex groups in adults between the ages of 25–74 years (McGrath et al. Citation1982). Although some recent studies find no effect of age on WBC (Kverneland et al. Citation2016) many support the differences seen in different races (Zezulka et al. Citation1987, Nalls et al. Citation2008, Lim et al. Citation2010).

Soluble intercellular adhesion molecule (sICAM-1)

Intercellular Adhesion Molecule (ICAM)-1 is a glycoprotein constitutively expressed on the surface of endothelial cells and leukocytes. ICAM-1 expression can be induced by pro-inflammatory cytokines, which are also thought to signal its shedding from the membrane so that it can be detected as soluble ICAM-1 (sICAM-1) in serum or plasma. Smokers have significantly higher plasma sICAM-1 levels (145.1 ng/mL) than non-smokers (110.3 ng/mL) (p < 0.0001), which was associated with endothelial dysfunction and clinical CVD (Nordskog et al. Citation2015). Studies have shown that ICAM-1 is overexpressed in the airway epithelium of subjects with COPD (Gross et al. Citation2012) and that release of sICAM-1 by bronchial epithelial cells following smoke exposure is significantly elevated in COPD (Chan et al. Citation2008, Rusznak et al. Citation2000). sICAM-1 levels have been shown to decline within 4–6 weeks following cessation of cigarette smoking (Aaron et al. Citation2015). Although care needs to be taken when comparing results, due to the fact that the most frequently employed immunoassays depend on antibodies incapable of recognizing all population variants of the adhesion molecule (Halvorsen et al. Citation2007, Tsai et al. Citation2012), sICAM-1 is considered to be a useful and reliable clinical risk endpoint in the assessment of vascular inflammation (Abdi et al. Citation2014). As a consequence, it should also be valid for evaluating a decrease in CVD risk associated with the use of RRPs. There are differences in sICAM levels in subjects of different ethnic backgrounds with levels being lower in individuals of African origin compared to whites (Hwang et al. Citation1997, Lutsey et al. Citation2006, Miller and Cappuccio Citation2007). Although plasma ICAM levels fall in children between the ages of 9 and 16 years (Nash et al. Citation1996), recent work suggests that there are no age-related differences between 23 and 74 years of age (Deneva-Koycheva et al. Citation2011, Bottino et al. Citation2015) which contradicts earlier evidence in which a linear relationship between the plasma concentration of sICAM and age were found in a comparison between subjects <40 (n = 52) and subjects >55 (n = 38) years of age (Miles et al. Citation2001). There is evidence that higher levels of sICAM in older subjects is significantly associated with frailty (Lee et al. Citation2016) and whilst there is no difference between healthy males and females (Deneva-Koycheva et al. Citation2011), sICAM levels are lower in pre-menopausal women than in post-menopausal women (Nyberg et al. Citation2014).

Fibrinogen (FBG)

Fibrinogen (FBG) is an acute phase protein produced in the liver that increases quickly and markedly in the circulation in response to vascular injury, infection and inflammation. FBG plays an essential role in coagulation, mediating fibrin assembly and platelet recruitment to form a platelet plug (thus limiting bleeding and promoting wound healing). Serum FBG levels show a positive association with age (Drenos et al. Citation2007, Wang et al. Citation2017) but there are no differences between genders (Wang et al. Citation2017). Ethnic differences have been reported with South Asians having similar levels to Caucasians and Chinese which are higher than those seen in Japanese subjects (Ishikawa et al. Citation1997, Gijsberts et al. Citation2015). South African black people were found to have higher levels of FBG than white South Africans (Lammertyn et al. Citation2015). FBG has been shown to be involved in major smoking-related diseases, such as CVD (Witkowska Citation2005), respiratory diseases (Koenig Citation2003, Marano et al. Citation2015) and cancer (Shibata et al. Citation2013, Lock-Johansson et al. Citation2014). A limitation of FBG as a biomarker of effect for smoking-related diseases is, however, that not all studies have found significant differences in FBG levels between smokers and non-smokers. One study that did find a significant relationship was published in 2003 and reported an odds ratio of 2.29 (95% CI, 1.69–3.09) for clinically elevated FBG in smokers of ≥24 cigarettes/day compared to non-smokers (Everett et al. Citation2007). On the other hand, a more recent, albeit smaller study, found no significant difference (p = 0.09) (Bazzano et al. Citation2003). In another study, there was no reduction in FBG levels in subjects who refrained from smoking for 1 year (King et al. Citation2017) One study found no significant difference between smokers who continued to smoke conventional cigarettes compared to those who switched to an RRP (p = 0.88) (Nordskog et al. Citation2015). It is important to note that many studies have shown that the functionality of FBG as an acute phase protein in response to inflammatory conditions is better reflected in changes in CRP than in corresponding changes of FBG levels (Al Rifai et al. Citation2017). Although some studies have shown an association between increased levels of FBG, CRP and a variety of other inflammatory cytokines in COPD (Martin Leroy et al. Citation2012), the usefulness of these measurements as biomarkers in clinical and investigational settings will rely on further refinement to understand whether they are associated with specific aspects of disease activity (Tran and Kalhan Citation2008).

Tumour necrosis factor-α (TNF-α)

Tumour necrosis factor-α (TNF-α) is an important cytokine involved in inflammatory processes, cell survival, growth and differentiation as well as apoptosis and anti-apoptosis (Rosenberg and Kalhan, Citation2012). Since inflammation plays a crucial role in CVD, particularly coronary artery disease (CAD), TNF-α has been intensively investigated as a biomarker. It is persistently elevated in post-myocardial infarction (MI) patients and is an independent risk factor for secondary coronary events in these patients, independent of other known risk factors (Bellisarii et al. Citation2001, Barnes Citation2009, Zelova and Hosek Citation2013). Plasma TNF-α concentration is associated with the degree of early atherosclerosis and correlates with metabolic and cellular perturbations that are considered to be important for the atherosclerotic process (Ridker et al. Citation2000). TNF-α also plays an important role in orchestrating inflammatory airway diseases, and there is an association between COPD and TNF-α (Skoog et al. Citation2002). In one study, serum TNF-α concentrations were found to be significantly elevated in COPD patients (6.97 pg/mL) compared to controls (5.3 pg/mL) (Wu et al. Citation2015) and another reported that smokers who smoked more than one pack of cigarettes/day had higher levels of serum TNF-α than those smoking less than one pack/day (Petrescu et al. Citation2010). However, in one study, serum TNF-α was lower in smokers with COPD (Higashimoto et al. Citation2008) while another reported lower levels of TNF-α in EBC from healthy smokers compared to levels in subjects with COPD (Maskey-Warzechowska et al. Citation2017). TNF-α has been measured in lung-derived matrices such as sputum (Ji et al. Citation2014), bronchoalveolar lavage fluid (BALF) and exhaled breath condensate (EBC) (Keatings et al. Citation1996). A recent study measured increased levels of TNF-α in the sputum of smokers although there were no differences in serum levels of TNF-α between the two groups (Kleniewska et al. Citation2016). TNF-α has also been studied in EBC (Garey et al. Citation2004, Gessner et al. Citation2005, Kuban and Foret Citation2013) and serum (Carpagnano et al. Citation2007, Dalaveris et al. Citation2009) from patients with lung cancer. Because smoking has inflammatory effects and is an established risk factor for inflammation-related diseases such as COPD, CVD and certain types of cancer, it is reasonable to assume that smoking should be correlated with increased systemic and also respiratory tract-related TNF-α levels. The information retrieved from the literature searches appear to confirm this assumption in general. The available evidence is limited, however, and the effect of smoking on increasing TNF-α levels is weak [although it has been shown that soluble TNF-α levels are significantly (p = 0.05) higher in acute exacerbations of COPD than in stable COPD (Tan et al. Citation2017)]. Particularly, there is only very weak and partly contradictory evidence for a dose-response relationship between TNF-α and smoking. Thus, it may be concluded that TNF-α, although mechanistically involved in the pathogenesis of inflammation-related diseases such as COPD, CVD and cancer, demonstrates no convincing evidence that it will be a suitable cytokine for measuring effects of smoking cessation or switching to a potentially less harmful product. Serum concentrations of TNF-α do not appear to change with increasing age (Kim et al. Citation2011, Kleiner et al. Citation2013, Wyczalkowska-Tomasik et al. Citation2016) except in subjects with type 2 diabetes where increases are seen over time, especially in females above the age of 41 years (Fatima et al. Citation2017). TNF-α levels are also not affected by gender (Lee et al. Citation1993, Arican et al. Citation2005) except in Chinese subjects in which levels are higher in females than males in the age range 20–45 years (Yuan et al. Citation2015). No differences were found between blacks and whites in a study of 508 males and females over 35 years of age (Paalani et al. Citation2011).

Myeloperoxidase (MPO)

Myeloperoxidase (MPO) is a pro-oxidant haeme-enzyme that is stored in and released from neutrophilic granulocytes which are an essential part of the innate (non-specific) immune system in humans (Derin et al. Citation2008). It also metabolizes tobacco smoke procarcinogens such as benzo(a)pyrene and aromatic amines into highly reactive intermediates (Chevrier et al. Citation2003). In a number of studies, a significant association between MPO activity and CVD was observed (Nussbaum et al. Citation2013), and circulating MPO may play a role as a diagnostic and a prognostic tool for CVD patients (Rudolph et al. Citation2008, Ikitimur and Karadag Citation2010, Anatoliotakis et al. Citation2013). It can be involved in the development of CVD, in particular atherosclerosis, by several mechanisms, including oxidation of low-density lipoprotein (LDL) to ox-LDL (Nicholls and Hazen Citation2005, Schindhelm et al. Citation2009, Koeth et al. Citation2013), plaque destabilization (Delporte et al. Citation2013), overproduction of MPO at the site of arterial inflammation and impairment of nitric oxide (NO)-induced vascular relaxation (Niccoli et al. Citation2010). Overall, the literature search revealed that the role of MPO as a risk and clinical marker for CVD (Meuwese et al. Citation2007, Baldus et al. Citation2003, Brennan et al. Citation2003, Thukkani et al. Citation2003), respiratory diseases (COPD and emphysema (Sapey et al. Citation2008, Stone et al. Citation2012) and various types of cancer (Hoy et al. Citation2002) is well established. However, from the 156 publications retrieved in the three searches, there was limited evidence that smoking leads to a dose-dependent increase in MPO levels (measured as amount or activity of the enzyme) or a decrease upon long-term smoking cessation (Andelid et al. Citation2007, Rudolph et al. Citation2012). One study found no increases in MPO in smokers compared to never smokers (Levitzky et al. Citation2008). Overall, the information available does not provide strong evidence for the suitability of the measurement of the amount or activity of MPO as a biomarker of effect in smoking studies. There is little evidence to suggest that there are age or gender-related differences in plasma MPO levels in healthy subjects (Meuwese et al. Citation2007, Sbarouni et al. Citation2011, Guven et al. Citation2013) and there is also no difference between American Africans and Caucasians (Kubala et al. Citation2008). Serum MPO levels do not change with increasing age except in subjects with type 2 diabetes (Shankar et al. Citation2012).

Exhaled nitric oxide (FeNO)

The fraction of exhaled nitric oxide (FeNO) is a promising biomarker for the diagnosis, follow-up and guide to therapy in patients with asthma (Malinovschi et al. Citation2012, Godinho Netto et al. Citation2016) and other respiratory diseases. It can also be used to measure initial physiological changes in the upper and lower airways. The measurement of FeNO is of limited relevance for CVD, although smoking decreases NO generation in endothelial cells (Messner and Bernhard Citation2014) which leads to a reduced endothelial-dependent vasodilatation (EDV) (Barua et al. Citation2001). Moreover, NO insufficiency leads to thrombosis, because endothelial NO limits platelet activation, adhesion and aggregation (Barua et al. Citation2001). FeNO is decreased in heart diseases such as congestive heart failure (Sumino et al. Citation1998, Hare et al. Citation2002). The literature search revealed that there is considerable variation in the findings when FeNO is measured in subjects with COPD with one early publication stating that smokers had lower FeNO than non-smokers [an observation also reported recently (Xu et al. Citation2016)] and that after 8 weeks of smoking cessation, levels in the former smokers rose to those seen in non-smokers although the number of subjects was very small (Loscalzo Citation2001). Generally, FeNO levels are near normal in COPD except during exacerbations (Robbins et al. Citation1997) and have so far not been shown to be predictive of important health outcomes in COPD (Verleden et al. Citation1999, Kharitonov and Barnes Citation2002, Kharitonov and Barnes Citation2006) with the exception of a recent study that showed that completely abstaining from smoking for 52 weeks led to near normalization of FeNO (and a concomitant drop in exhaled carbon monoxide) (Campagna et al. Citation2016). While some interest has been focussed on measuring alveolar FeNO (Yoon and Sin Citation2011), the levels are not associated with disease severity or smoking status (Brindicci et al. Citation2005). A recent study has concluded that reference values for FeNO among respiratory healthy non-smokers should be outlined stratified for gender using individual reference values and a separate cut-off limit for current smokers (Toren et al. Citation2017). More work will be required before the value of FeNO measurement in COPD is fully understood. Overall, therefore, it can be concluded that currently FeNO appears not to be a suitable biomarker for evaluating acute (Lappas et al. Citation2016) effects of smoking and cessation/switching.

Sputum neutrophils

Although the literature searches found no published papers on the associations between CVD/heart disease or cancer and sputum neutrophil numbers, several studies are available that show that numbers of sputum neutrophils are associated with respiratory diseases such as COPD (Brindicci et al. Citation2005), asthma (Tsoumakidou et al. Citation2003, Singh et al. Citation2010, Zuiker et al. Citation2015) and chronic fibrosis (Chaudhuri et al. Citation2014). Most studies report an increase in the number of sputum neutrophils, as would be expected as a consequence of airway exposure to tobacco smoke, in samples from smokers compared to non-smoking controls. This is not always the case, however, and other studies have reported no differences (Conese et al. Citation2014) or even a decrease (Swan et al. Citation1991, Zuiker et al. Citation2015). Likewise, some studies have found no change or an increase in the number of sputum neutrophils following smoking cessation [e.g. from 60.1% before smoking cessation to 80.1% 1 year later (Lensmar et al. Citation1998)]. In terms of determining inflammatory biomarkers, sputum is almost as suitable as BALF or lung biopsies (Willemse et al. Citation2005), and its collection is minimally invasive and relatively safe. Although the majority of the evidence suggests that sputum neutrophil numbers are elevated in smokers, this association has been investigated only in relatively small studies. The available evidence for a dose–response relationship is limited and inconsistent. Smoking cessation is not found to steadily decrease the sputum neutrophil levels. To the contrary, levels were found to remain elevated or even to increase after smoking cessation and to remain so even after several months or years (Keatings et al. Citation1996), suggesting that smoking-related airway inflammation persists for a considerable time period. Taken together, the information would suggest that even though neutrophils are mechanistically involved in the pathogenesis of inflammation-related airways diseases they are not a suitable biomarker of effect in smoking and smoking cessation studies. Age has been reported to have either no effect on sputum neutrophil numbers from healthy subjects (Belda et al. Citation2000, Spanevello et al. Citation2000, Busse et al. Citation2017) or to cause an increase as subjects get older (>50 years) (Thomas et al. Citation2004, Pignatti et al. Citation2011, Carpagnano et al. Citation2013, Simpson et al. Citation2013), with one showing a greater increase in females than males (Pignatti et al. Citation2011).

Lipoprotein-associated phospholipase A2 (Lp-PLA2)

Lipoprotein-associated phospholipase A2 (Lp-PLA2), also known as platelet-activating factor acetylhydrolase (PAF-AH), is a vascular-specific inflammatory enzyme, predominantly expressed by macrophages, lymphocytes and foam cells in atherosclerotic plaques. Over 80% of circulating Lp-PLA2 is associated with atherogenic LDL-cholesterol, while about 10% is associated with high-density lipoprotein-cholesterol (HDL-C) (Willemse et al. Citation2005). Circulating Lp-PLA2 is constitutively active and is upregulated by mediators of inflammation at the transcriptional level as a physiological response to inflammatory stimuli (Tsimihodimos et al. Citation2002, Caslake and Packard Citation2003, Racherla and Arora Citation2012, Vittos et al. Citation2012). A number of epidemiological studies have demonstrated that elevated levels of Lp-PLA2 are associated with increased risk of cardiovascular events independent of established risk factors (Stafforini Citation2009). Plasma Lp-PLA2 was shown to be independently associated with coronary heart disease (CHD) in a cross-section study on 531 Chinese subjects (Yang et al. Citation2017) and a meta-analysis of 15 studies with 30,857 subjects showed that Lp-PLA2 was independently associated with cardiovascular events, particularly in subjects with stable CHD not receiving Lp-PLA2 inhibitor therapy (Li et al. Citation2017). Plasma levels of Lp-PLA2 are also associated with the severity of CHD (Ge et al. Citation2016). Lp-PLA2 is not widely used as biomarker in clinical or epidemiological studies on respiratory diseases, although increased levels have been reported in BALF from patients with acute respiratory distress syndrome (Nakos et al. Citation2004, Citation2005). In one study, smoking cessation lowered the plasma Lp-PLA2 to levels similar to those seen in never smokers (Persson et al. Citation2007). Differences between smokers and non-smokers were investigated in a small number of studies, which in general showed significant differences between smokers and non-smokers (e.g. 256 and 191 ng/mL, respectively; p < 0.001; Persson et al. Citation2007). Moreover, one study has shown a significant dose–response relationship between smoking intensity and Lp-PLA2 blood levels (Tselepis et al. Citation2009). It is highly likely that the link between smoking and the elevation of Lp-PLA2 levels is linked to the inflammatory and oxidative stress processes caused by smoking (Tselepis et al. Citation2009). The searches revealed no systematic studies showing that smoking cessation leads to a decrease in Lp-PLA2 levels and no published data are available about the effect on circulating Lp-PLA2 levels following switching from conventional cigarettes to an RRP or other nicotine product. For this reason, Lp-PLA2 is not likely to be a suitable biomarker for use in smoking/cessation/switching studies until more data becomes available. It has been shown that Lp-PLA2 activity was lower in women compared with men and was lowest in black, intermediate in Hispanic and highest in white subjects (Brilakis et al. Citation2008).

CVD Biomarkers

Albumin (ALB)

Albuminuria is not only a predictor of declining renal function but also is independently associated with adverse cardiovascular outcomes (Imaizumi et al. Citation1990). How smoking might actually damage the kidney and lead to (micro)albuminuria is not yet fully elucidated, although a number of potential mechanisms for the involvement of smoking in the renal pathophysiological processes have been discussed, including smoking-related increases in blood pressure leading to alterations in kidney haemodynamics (Stephen et al. Citation2014); smoking-related atherogenic effects (Metcalf et al. Citation1993, Orth Citation2002, Hogan et al. Citation2007); effects of chronic smoking on endothelial dysfunction leading to a faster decline in renal function (Hogan et al. Citation2007); carboxyhaemoglobin-induced renal hypoxia (Orth Citation2002); elevated levels of nephrotoxic cadmium (Metcalf et al. Citation1993); smoke-induced formation of advanced glycation end products (AGE) that increase vascular permeability (Hogan et al. Citation2007); smoking-induced insulin resistance (Pinto-Sietsma et al. Citation2000) and smoke-induced disturbances in glycaemic control leading to increased glomerular permeability (Pinto-Sietsma et al. Citation2000). The published literature shows that there is some evidence that smoking leads to increased urinary albumin (ALB) levels [e.g. 4.6 mg/L in non-smokers and 5.4 mg/L in smokers (p < 0.001) (Metcalf et al. Citation1993, Al Rifai et al. Citation2017); 62 mg/24 h in non-smokers and 100.4 mg/24 h in smokers (p < 0.01) (Metcalf et al. Citation1993)]. Most of the studies available, however, were conducted with diabetic subjects. Effects of smoking on ALB excretion appears to be higher in patients with diabetes or renal damage or dysfunction than in healthy subjects. Nevertheless, in almost all studies that investigated the dose–response relationship between ALB levels and the smoking dose, a significant association was found (Corradi et al. Citation1993). Former smokers, who stopped smoking at least 1 year before, were reported to have ALB levels between current and never smokers (Metcalf et al. Citation1993, Ikeda et al. Citation1997, Pinto-Sietsma et al. Citation2000, Hogan et al. Citation2007). However, in no case have acute effects (observed within days or weeks) of smoking cessation on ALB excretion been reported in the literature.

As noted above, a major limitation in the evaluation of urinary ALB as a biomarker of effect for smoking-related diseases is the fact that most of the studies have been performed with diabetic subjects. More data for healthy subjects (whether smokers, non-smokers or ex-smokers) are required. Therefore, taken together, the information available does not strongly suggest the use of urinary ALB as a biomarker of effect for studies on smoking or smoking cessation, although this conclusion could change if and when more data become available for ALB excretion in healthy subjects as well as additional results after smoking cessation. Some studies have reported decreases in serum ALB concentrations with increasing age (Greenblatt Citation1979, Cooper and Gardner Citation1989, Veering et al. Citation1990, Salive et al. Citation1992) although the decreases could be attributed to poorer nutritional status, inflammation or external loss (Kaysen and Levin Citation2002). Others have reported that hypoalbuminaemia is not a consequence of normal aging (Campion et al. Citation1988) and that age does not change the rate of synthesis or concentrations of ALB (Fu and Nair Citation1998). Ethnicity affects ALB levels in healthy subjects with differences between males and females (Tembe et al. Citation2014, Lim et al. Citation2015).

Lipids

Lipids are essential for a number of cellular processes including synthesis of cellular membranes, formation of lipoprotein particles (complex particles composed of multiple proteins), signal transduction and energy storage among others and the tight regulation of their metabolism contributes to the maintenance of cellular homeostasis. LDL is one of the five major groups of lipoprotein which are, from least dense to most dense, chylomicrons, very low-density lipoprotein (VLDL), intermediate-density lipoprotein (IDL), LDL and HDL. All of these lipoproteins can carry cholesterol, and the vast majority of studies indirectly measure their levels by determining the level of bound cholesterol (e.g. LDL-C, HDL-C). Because of the association between LDL-C and CVD, in particular atherosclerotic CVD, it is often referred to as “bad cholesterol”, as it can transport lipid molecules into artery walls, attract macrophages, and thus drive atherosclerosis (Metcalf et al. Citation1993, Freedman et al. Citation2005, Voulgari et al. Citation2011). A possible mechanism by which smoking might increase LDL-C is by increasing the level of free fatty acids (FFA) in blood, which in turn stimulates the hepatic synthesis and excretion of cholesterol. It has also been suggested that smoking may alter the expression of genes influencing the LDL particle diameter and thus increase the atherogenic potency of LDL-C (Otokozawa et al. Citation2010, Ference and Mahajan Citation2013, Imes and Austin Citation2013). The literature searches revealed that in general but not always smoking increased LDL-C levels, but there is conflicting evidence with respect to the effects of smoking cessation with some studies reporting lowering of levels (Hozawa et al. Citation2006) and others reporting no change (Allen et al. Citation1994, Ludviksdottir et al. Citation1999). Overall, the information available does not provide strong evidence for the suitability of LDL-C as a biomarker of effect in cessation or switching studies.

HDL has been suggested to fulfil a number of biological functions, the most important of which is reverse cholesterol transfer (RCT) which involves removal of cholesterol from the plasma, macrophages and other cells for transfer (primarily) to the liver and eventual removed from the organism. The fact that cholesterol bound to HDL (HDL-C) is in the process of being removed from the organism is why HDL-C has been universally regarded as “good cholesterol.” Other functions hypothesized for HDL are: anti-oxidative activity, anti-inflammatory activity, anti-apoptotic activity, vasodilatory effects and inhibition of platelet aggregation (Freeman et al. Citation1993, Hatsukami et al. Citation2005, Hozawa et al. Citation2006). Whilst the association of HDL-C with various CVD endpoints has been well-known for many years its association with respiratory disease such as COPD is inconsistent with some reports showing association (Kontush and Chapman Citation2006) and others showing no association (Cirillo et al. Citation2002). A recent study found that adults with low HDL-C (and high triglyceride), particularly if they had diabetes, were at increased risk of incident CHD and stroke (Lee et al. Citation2017). Although a limited number of studies were retrieved when searching for HDL and cancer, all the studies identified showed a decreased level of HDL-C in lung cancer compared to some type of control or an association of low levels of HDL-C with the risk of lung cancer (Garcia-Larsen et al. Citation2014, Sato et al. Citation2014, Wei et al. Citation2014). There was overwhelming evidence that following smoking cessation, HDL increased and could revert to normal levels within 4–8 weeks (Kucharska-Newton et al. Citation2008, Ahn et al. Citation2009). In one study, 12 months of smoking cessation increased HDL-C from 42.4 to 44.7 mg/dL (p < 0.001) (Forey et al. Citation2013) and another reported a 9.3% increase in HDL-C one year after smoking cessation (Mandraffino et al. Citation2017). Two studies were found that determined the comparison of levels of HDL-C in subjects who switched to a second-generation electrically heated cigarette smoking system. Roethig et al. (Citation2008) reported an increase of 5 mg/dL for HDL-C after 12 months in smokers who switched, compared to an increase of only 1 mg/dL for those who continued to smoke conventional cigarettes (p = 0.008). In a similar study, Martin Leroy et al. (Citation2012) reported an increase in HDL-C after 1 month following switching from 59.0 to 63.9 mg/dL (p < 0.0001). Despite the widespread use of the decrease of HDL-C as a marker for the increase in CVD and vice versa, there is an issue with HDL-C as a biomarker. Recent studies have shown that levels of HDL-C are not causally related to CVD (Hovingh et al. Citation2015, Pownall and Gotto Citation2016). During the past 5 years, several inhibitors of cholesteryl ester transport protein (CETP) have been evaluated as a possible approach to lower the risk of CVD. CETP is known to transfer cholesterol from HDL to VLDL, thus inhibition of CETP would be expected to increase the amount of cholesterol bound to HDL and, in turn, the level of HDL-C. Three potential CETP inhibitors have now been evaluated in phase 3 trials. As anticipated, in all three trials levels of HDL-C were indeed significantly elevated. However, none of the three compounds showed any effect with respect to reducing the risk of CVD (Voight et al. Citation2012). The most likely explanation is that while the amount of cholesterol bound to HDL increased, the actual amount of HDL did not. In conclusion, therefore, the published data would qualify HDL-C as a valid biomarker for use in smoking/cessation/switching studies. A Finnish study reported that HDL-C levels increased with advancing age with a more pronounced effect in females but with no ethnic differences (Thelle et al. Citation1982) and more recent studies have shown differences in HDL-C in American Africans, Latinos and whites (Bauman et al. Citation1998) and between Hispanics, non-Hispanic whites and American Mexicans (Chang et al. Citation2011). It has also been suggested that aging alters HDL composition which may lead to functional impairment or the onset or progression of CVD (Holzer et al. Citation2013).

Apolipoprotein A1 (ApoA1)

ApoA1 is the major HDL apoprotein which belongs to the group of reverse cholesterol transport proteins having anti-atherogenic properties (Schwartz et al. Citation2012, Shinkai Citation2012, McLain et al. Citation2016) as well as exerting innate protective effects in systemic inflammation (Tall Citation1998) and having anti-oxidant properties through its thiol groups (Sharifov et al. Citation2013). Thus, as would be expected, ApoA1 is inversely associated with the risk of CVD. The role of ApoA1 in preventing CVD has been underlined by observations that autoimmunity to the protein is associated with an increased risk of CVD (Szadkowski and Myers Citation2008). Importantly, ApoA1-mimetics have seen therapeutic success for treating CVD (Vuilleumier et al. Citation2004, Teixeira et al. Citation2012, Vuilleumier et al. Citation2014). Smoking (amongst other factors) is correlated with an unfavourable apolipoprotein profile (Frondelius et al. Citation2017). There are a number of mechanisms by which smoking decreases the degree of functionally of intact ApoA1. Smoking promotes the MPO-mediated nitration and chlorination of tyrosine-192 in ApoA1 (Hovingh et al. Citation2010, Smith Citation2010, Imaizumi et al. Citation2011, Gordon and Davidson Citation2012, Leman et al. Citation2014) and also enhances the reaction of lipid peroxidation products with lysine-226 in ApoA1 (Hadfield et al. Citation2013). Acrolein and other oxidants and toxicants in smoke can cause oxidation of the thiol groups in ApoA1 (Zheng et al. Citation2004, Shao et al. Citation2005, Shao Citation2012). The literature search did not identify many studies comparing ApoA1 levels in smokers and non-smokers [one was identified in which quantification was by LC-MC and found reduced levels in smokers compared to non-smokers (Wang et al. Citation2015)], but a number of older studies have found significant differences (Szadkowski and Myers Citation2008). The literature search did find a small study that investigated the effect of smoking cessation on ApoA1 levels. Quitting smoking for 12 weeks significantly increased the ApoA1 levels in smokers from 151.7 to 158.6 mg/dL (p < 0.01) (Dedonder-Decoopman et al. Citation1980, Gnasso et al. Citation1984, Haffner et al. Citation1985). Overall, the data on differences between smokers and non-smokers, dose–response relationships and smoking cessation suggest that the inverse association of the serum or plasma ApoA1 levels with smoking is plausible. The number of relevant studies is limited, however, and to date no study has investigated the use of ApoA1 with respect to switching from a conventional cigarette to an RRP. Therefore, until further data are available, it is reasonable to continue to use HDL-C as the preferred lipid-associated biomarker of effect. Plasma levels of ApoA1 were not affected by age or gender in a Finnish study (Sirnio et al. Citation2017) but were found to decrease by 1.21 µg/mL per decade between the ages of 56–105 years in an Australian study (Muenchhoff et al. Citation2017). In a study in the Chinese Han population, ApoA1 levels were reported to increase between the ages of 18 and 50 years (Lu et al. Citation2012).

Oxysterols (OxS)

Oxidized cholesterol species (oxysterols, OxS) play important roles in many biological processes, including cholesterol homeostasis (Iwaoka et al. Citation2014). Compared to other biomarkers of effect linked to CVD and related diseases, the evidence for a predictive power of OxS is rather limited. In particular, oxysterols do not appear to have any advantages over other biomarkers for oxidative stress, inflammation or risk of atherogenic plaque formation. Nevertheless, there is growing evidence in the literature for the involvement of OxS in the pathogenesis of atherosclerosis (Bjorkhem Citation2002, Bjorkhem et al. Citation2002, Olsen et al. Citation2012). One report was retrieved from the literature that showed an association between respiratory disease and blood levels of 25-hydroxycholesterol in humans (Sugiura et al. Citation2012) but little published information is available for the association between smoking and OxS blood levels. The most probable mechanism by which smoking could elevate OxS levels is by oxidative stress and stimulation of inflammatory processes. Although mechanistically involved in the pathogenesis of many age-related diseases (Leoni et al. Citation2013), there is no convincing evidence that OxS would be suitable biomarkers of risk for these diseases in general or for use in smoking cessation or switching studies designed to provide information about the risk profile of alternative reduced risk tobacco products. Multiple sterols have been shown to vary significantly by sex, age and ethnicity (Diczfalusy et al. Citation2008, Stiles et al. Citation2014).

Homocysteine (HCY)

Homocysteine (HCY) is a sulfhydryl-containing non-protein amino acid which has an additional methylene bridge compared to cysteine. It is biosynthesized from the amino acid methionine to which it can also be recycled in a process that involves folate and various B-vitamins (B6 and B12). When the upstream or downstream metabolic pathway of HCY is blocked or delayed for various reasons, it leads to the accumulation of HCY, causing hyperhomocysteinaemia which, although causing no symptoms, affects the interior lining of blood vessels, increasing the risk of atherosclerosis (Sun et al. Citation2017); increases the risk of deep vein thrombosis (Kamat et al. Citation2010, Ekim et al. Citation2015) and pulmonary embolism (Cellai et al. Citation2014); may be related to Alzheimer’s disease (Sharma and Lipincott Citation2017) and other types of dementia (Xie et al. Citation2017) and may be associated with pre-eclampsia in pregnancy (Sayyah-Melli et al. Citation2016, Wadhwani et al. Citation2016). Thus, hyperhomocysteinaemia is associated with various diseases, in particular CVD (Olkkonen Citation2012, Lathe et al. Citation2014). A small number of older publications (Andersson et al. Citation2001, Seemungal et al. Citation2007, Fimognari et al. Citation2009), and one more recent (Khan et al. Citation2016) have investigated increased homocysteine levels in COPD. Associations between HCY and cancer were found to be inconsistent. Circulating HCY levels increase with age and are higher in men than women in all age groups (Marti-Carvajal et al. Citation2013), and levels may increase with chronic vitamin B12 deficiency, for example in vegetarians (Nygard et al. Citation1995). Smoking may increase plasma HCY levels by a number of mechanisms including: increase in reactive oxygen species levels) (Obersby et al. Citation2013); inactivation of enzymes involved in the metabolism of HCY (De Bree et al. Citation2002, Hu et al. Citation2005, Baccarelli et al. Citation2007); a reduced intake of dietary vitamins/folate by smokers (De Bree et al. Citation2002, Sobczak Citation2003, Baccarelli et al. Citation2007); and raised levels of the free (sulfhydryl) form of HCY in smokers induced by components of tobacco smoke (Subar et al. Citation1990, Sobczak Citation2003, Hu et al. Citation2005, McEligot et al. Citation2006, Baccarelli et al. Citation2007, Vardavas et al. Citation2008). Data from the US National Health and Nutrition Examination Survey (NHANES) study (Sobczak Citation2003) showed HCY levels of 11.04 µmol/L compared to 9.45 and 9.29 µmol/L in smokers, former smokers and never smokers, respectively (p < 0.001 for current smokers versus never smokers). A study in a Chinese aged population (with stable CVD) found circulating homocysteine values of 16.8, 14.6 and 14.1 µmol/L smokers, former smokers (at least 3 months smoking abstinence) and never smokers, respectively (p = 0.004) (Bazzano et al. Citation2003). Because present knowledge does not supply strong evidence for a relationship between HCY and CVD (or other diseases) (Chen et al. Citation2015), and since no acute effects of smoking cessation on HCY concentrations in blood have been reported in the literature it would appear that the information currently available does not suggest the use of HCY as a biomarker of effect for use in smoking cessation/switching studies. Plasma HCY levels are affected by age, sex and ethnicity (El-Sammak et al. Citation2004, Golbahar et al. Citation2004, Strassburg et al. Citation2004, Refsum et al. Citation2006).

P-selectin (P-sel)

P-Selectin (P-Sel), a component of the membrane of the α-granules of platelets and also of Weibel-Palade bodies, together with E- and L-selectin, is a member of the selectin protein family of adhesion molecules. The expression of membrane-bound P-Sel is a suitable indicator for platelet activation in vivo, a process which is involved in several pathophysiological processes such as atherosclerosis, thrombotic diseases and cancer (Chen et al. Citation2015). P-Sel participates in the very early inflammatory processes of leukocyte rolling, tethering, attachment and transmigration across the vascular endothelium, thus initiating events in atherothrombosis (Blann et al. Citation2003, Geng et al. Citation2004, Kappelmayer et al. Citation2004, Merten and Thiagarajan Citation2004, Chen and Geng Citation2006, Antonopoulos et al. Citation2014). There are few reports on a direct involvement or association of P-Sel in the development of respiratory diseases. The occurrence of pulmonary embolism as a complicating event in deep venous thrombosis might be regarded as an indirect involvement of P-Sel in pulmonary disease (Merten and Thiagarajan Citation2004, Chen and Geng Citation2006). In many, but not all, studies there were statistically significantly higher P-Sel levels and expression activities in smokers compared to non-smokers (Antonopoulos et al. Citation2014). The literature search identified only a single study that investigated levels of P-Sel following smoking cessation and which reported no significant decrease in P-Sel levels following three weeks of smoking cessation (Bermudez et al. Citation2002, Butkiewicz et al. Citation2006, Neubauer et al. Citation2009, Lupia et al. Citation2010). On the other hand, one older study was found which did report a statistically significant decrease in P-Sel following 6 weeks of smoking cessation (Lupia et al. Citation2010). In summary, data providing evidence validating the role of P-Sel in smoking cessation is sparse and would need to be expanded before it can be recommended for assessment of novel tobacco products. Circulating P-selectin levels have been shown to vary between sexes, ethnic groups and with age (Lee et al. Citation2008, Bielinski et al. Citation2015).

Thiocyanate (SCN)

Thiocyanate (SCN), also known as rhodanide, is a pseudohalogen anion and is the main metabolite and detoxification product of cyanide. SCN is also involved in the myeloperoxidase system, which catalyzes the formation of hypochlorite, hypobromite and hypothiocyanite (OSCN) by oxidizing chloride (Cl), bromide (Br) and SCN, respectively, in the presence of hydrogen peroxide (H2O2). The formation of OSCN- depends on the plasma thiocyanate levels and is the driving force in determining the extent of thiol oxidation with a direct influence on the redox balance in plasma. SCN is thus an important mediator of inflammation-induced damage to proteins, for example in smokers, who exhibit significantly elevated SCN blood levels (Blann et al. Citation1997b). Mean serum/plasma SCN concentrations for smokers and non-smokers have been reported to be predominantly in the range of 90–180 and 40–75 µmol/L, respectively, with corresponding mean concentration ranges in urine of 150–560 and 75–285 µmol/L. Levels in saliva were in the ranges 2500–3300 and 1200–1300 µmol/L (Morgan et al. Citation2011). Urinary SCN was reported to be associated with active smoking in a dose-related manner subjects over 20 years of age (Jain Citation2016). A recent study showed that salivary SCN was increased in smokers compared to non-smokers and the increase was related to the number of cigarettes smoked (and correlated with an increase in the number of extranuclear micronuclei) (Baldawa et al. Citation2016) and another reported salivary SCN concentrations of 0.8 ± 0.3 mM in control subjects compared to 2.3 ± 0.8 mM in smokers (Hegde et al. Citation2016). The much higher salivary SCN concentrations are the result of the active secretion of thiocyanate by the salivary glands (Scherer Citation2006). Another important biochemical property of SCN is its ability to catalyze the endogenous formation of carcinogenic nitrosamines from secondary amines and nitrite as precursors (Tsuda and Kurashima Citation1991), and smokers are at higher risk for this reaction due to their elevated SCN levels in body fluids, particularly in saliva (Bartsch et al. Citation1989, Tsuda and Kurashima Citation1991, Tricker Citation1997). The role of SCN as a substrate of MPO and the MPO-catalyzed oxidation of thiocyanate with subsequent lipoprotein carbamylation might implicate it in the pathogenesis of atherosclerosis, particularly in smokers (Bartsch et al. Citation1989). SCN levels in urine have been shown to correlate with emphysema, chronic bronchitis, cancer, wheezing and coughing (Wang et al. Citation2007), although this correlation may be a function of SCN acting as a biomarker of exposure for smoking. Following smoking cessation, SCN concentrations fall to those of non-smokers within 3–6 weeks. Although cyanide and SCN may come from other sources (mainly via dietary intake), making it potentially a non-specific biomarker of exposure to tobacco smoke, there are clear differences between smokers and non-smokers making SCN a suitable biomarker for use in smoking studies. New instrument-free analytical systems for measuring thiocyanate levels in saliva have been used to estimate non-smokers as having 0.28–0.87 mM and smokers to have 0.78–4.28 mM (Pena-Pereira et al. Citation2016). Measured plasma SCN levels are higher in females than in males (Foss and Lund-Larsen Citation1986, Zil and Rahman Citation2006) and urinary thiocyanate content was higher in non-Hispanic whites than non-Hispanic blacks who had higher levels than American Mexicans (Jain Citation2016).

Von Willebrand factor (vWF)

Von Willebrand factor (vWF) is a blood glycoprotein of importance for maintaining haemostasis. Deficiency in, or a defect of, vWF leads to diseases such as von Willebrand disease (the most common hereditary blood coagulation abnormality), thrombotic thrombocytopenic purpura (TTP, Moschcowitz syndrome), gastrointestinal bleeding (Heyde's syndrome) and possibly haemolytic-uraemic syndrome (HUS). More recently vWF has been found to be involved in several pathologic processes beyond haemostasis, such as angiogenesis, cell proliferation, inflammation, and tumour cell survival (Shiue Citation2015). Increased plasma levels of vWF (which are presumed to arise from adverse changes to the endothelium, the major site of vWF biosynthesis) are associated with thrombosis, cardiovascular, neoplastic and connective tissue diseases (Lenting et al. Citation2012). Several studies in initially healthy subjects showed rather weak associations between vWF levels and CVD risk that did not always reach statistical significance. This association becomes much stronger in patients with pre-existing vascular disease, particularly survivors of myocardial infarction (MI) and patients with type 1 and 2 diabetes (Sadler Citation1998, Franchini and Lippi Citation2007, Spiel et al. Citation2008), and the association between vWF levels and cardiovascular risk was found to be stronger in smokers compared to non-smokers (Lip and Blann Citation1995, Blann et al. Citation1997a, Whincup et al. Citation2002, Blann Citation2006, Vischer Citation2006, Franchini and Mannucci Citation2008, Frankel et al. Citation2008, Spiel et al. Citation2008, Paulinska et al. Citation2009, van Galen et al. Citation2012, Lenting et al. Citation2013, Sonneveld et al. Citation2015). Evidence for an association between elevated blood levels of vWF and respiratory diseases is rather limited, and no significant correlation between serum vWF levels and subsequent decline in FEV1 was found, indicating that vWF is not a suitable predictor for increased risk of accelerated decline in FEV1 (O'Callaghan et al. Citation2005). On the other hand, elevated levels of vWF have been reported to have some predictive and prognostic value for cancer in general (Chambers et al. Citation1999) and non-small cell lung cancer in particular (Blann et al. Citation1997a). Some studies provide evidence that smoking is associated with an increase of vWF blood levels [e.g. 102.4, 100.2 and 99.0 IU/dL in smokers, ex-smokers and never smokers, respectively (p < 0.05 comparing smokers to never smokers) (Martini et al. Citation2005), and 114 and 60 IU/dL in smokers and non-smokers, respectively (p = 0.03) (Kumari et al. Citation2000)]. One large prospective study found that vWF levels were not associated with smoking status based on a univariate analysis, but did report a small but statistically significant increase in vWF levels for current smokers versus never smokers (p < 0.05) using multiple logistic regression (Al-Awadhi et al. Citation2008). There is limited evidence for a dose-response relationship between smoking and vWF levels. In some studies, a rapid increase of serum vWF was found when samples were drawn immediately following the smoking of two cigarettes in smokers who had abstained from tobacco overnight (Conlan et al. Citation1993). Only limited information is available about the effect of smoking cessation on blood vWF concentrations but the sparse data available suggest that vWF levels may decline within weeks after smoking cessation [e.g. from 114.3 to 97.4 IU/dL after 6 weeks (p = 0.01) (Blann and McCollum Citation1993, Blann et al. Citation1998)]. There does appear to be some evidence for the association of vWF levels with smoking status and with smoking cessation. Data from the two switching studies that have been cited previously do not, however, support a utility for vWF as a biomarker. Martin Leroy et al. (Blann et al. Citation1997c) did indeed find a statistically significant decrease in vWF levels after one month for smokers who switched to an RRP, but they reported virtually the same difference for subjects who continued to smoke conventional cigarettes. Roethig et al. (Martin Leroy et al. Citation2012), however, did not find a significant difference in smokers who switched to an RRP (p = 0.37). Published data shows that vWF levels are higher in females than males (Payne et al. Citation2014, Zhou et al. Citation2014), with higher levels in African Americans than Caucasians (Payne et al. Citation2014). Several studies also show that levels of vWF increase with age (Ishikawa et al. Citation1997, Cohen et al. Citation2012, Cowman et al. Citation2015).

Miscellaneous biomarkers

Glycated haemoglobin (HbA1c)

Glycated haemoglobin (HbA1c) is a long-term marker of blood glucose levels, reflecting the average glucose plasma concentrations over approximately 2 months prior to the test (Roethig et al. Citation2008) and is the “gold standard” for the diagnosis, therapeutic monitoring and prognosis of patients with diabetes mellitus. In epidemiological studies (Miao Jonasson et al. Citation2012), significant associations between HbA1c and CVD, cardiovascular (CV) death or total mortality were observed, although it has to be borne in mind that by far the largest number of studies on the associations between HbA1c and diseases were conducted with diabetic patients of both type 1 and type 2 diabetes. There is almost no evidence for an association between HbA1c and respiratory disease. A meta-analysis of 14 observational studies with 98,978 participants (with diabetes) found that smokers had significantly higher HbA1c levels than non-smokers (Kar et al. Citation2016). Only sparse information is available on the dose-response relationship between HbA1c and the smoking dose, determined for example by daily cigarette consumption (CPD), pack-years or suitable biomarkers of exposure such as cotinine in body fluids. The data that are available (Boeing et al. Citation2000, Anan et al. Citation2006, Kotani et al. Citation2010), however, show moderate but statistically significant associations with smoking and in this respect would qualify HbA1c as a possible biomarker of effect [e.g. 5.11 and 4.99% in smokers and non-smokers respectively (p < 0.001) (Gulliford and Ukoumunne Citation2001, Schwab et al. Citation2008, Clair et al. Citation2011, Vlassopoulos et al. Citation2013, Ohkuma et al. Citation2015), 4.86 and 4.67% in smokers and non-smokers respectively (p < 0.05) (Simon et al. Citation1989) and 8.6 and 7.6% (p = 0.002) in smokers and non-smokers respectively (Nilsson et al. Citation1995)]. Since the half-life of HbA1c is determined by the lifespan of red blood cells (∼4 months), it is obvious that changes in HbA1c levels can only be expected in long-term observations (months–years). There is general agreement in the results retrieved that HbA1c levels in ex-smokers (after smoking cessation periods of 3 months or more) are close to those of never smokers (Baggio et al. Citation2002), which would generally qualify HbA1c as a suitable long-term (months–years) biomarker of effect in studies with large populations (e.g. post-market studies). In subjects with normal glucose tolerance, HbA1c levels rise by about 0.1% per decade (slightly less in those with impaired glucose tolerance or impaired fasting glucose) (Davidson and Schriger Citation2010). Ethnic background also affects HbA1c with non-Hispanic blacks having higher levels than Mexican Americans who have higher levels than non-Hispanic whites (age range 40–75 years) (Boltri et al. Citation2005, Davidson and Schriger Citation2010). Whilst HbA1c is the current “gold standard” for the diagnosis of diabetes it should not be forgotten that the value is dependent upon the life span of circulating red blood cells leading to the suggestion that in children a correction factor should be employed (An et al. Citation2016) and that in the elderly, HbA1c is actually unsuitable for diagnosing diabetes (Wu et al. Citation2017).

Carboxyhaemoglobin (COHb)

Carbon monoxide (CO) is produced by the incomplete combustion of organic material, including tobacco [in which it has been described as the most toxic component of burning cigarettes (Gulliford and Ukoumunne Citation2001, Hokanson et al. Citation2006, Clair et al. Citation2011, Voulgari et al. Citation2011, Choi et al. Citation2013, Gerber et al. Citation2013, Vlassopoulos et al. Citation2013, Ohkuma et al. Citation2015)]. CO binds to haemoglobin (about 200 times more strongly than oxygen) to form carboxyhaemoglobin (COHb), and the resulting lowered oxygen binding and transport means that less oxygen is available at peripheral tissue sites leading to hypoxia. COHb may be considered a biomarker of exposure and biological effect, the latter because the saturation of haemoglobin with CO is a direct measure for the biological effect of CO, namely, impairment of the oxygen supply of organs.

Cigarette smoking is known to elevate both respiratory and blood CO levels and, therefore, raising the COHb [e.g. 5.5% in smokers compared to 0.7% in non-smokers (Leone Citation2015)] levels which are associated with increased numbers of cardiac events, including fatal and non-fatal myocardial infarction. Lowering of COHb rapidly leads to an increase in the respiratory indices of vital capacity (VC), forced expiratory flow 25–75% (FEF25–75) and FEV1 (MacNee et al. Citation1989). The levels of COHb (in smokers and non-smokers) correlate with CVD risk (Hedblad et al. Citation2006, Pezzuto et al. Citation2013). Following smoking cessation, the levels of COHb decline quickly (Lind et al. Citation2004, Hedblad et al. Citation2005, Citation2006, Lüdicke et al. Citation2016) as its half-life is between one and four hours in blood with its elimination being mainly dependent on ventilation rate and level of physical activity (Roethig et al. Citation2007, Tricker et al. Citation2012, van Staden et al. Citation2013). Although the literature search provided no evidence of a direct link between COHb levels and disease, COHb should be considered for use in smoking/cessation/switching studies as it reflects exposure to carbon monoxide which is associated with respiratory, cardiovascular and inflammatory diseases. Carbon monoxide in blood, as assessed by the COHb%, is an accepted indicator of the cardiovascular risk in smokers. The biological, epidemiological and clinical trial information is in line with the induced effects observed upon smoking cessation, suggesting that a decrease in COHb mediates some of the reduced CVD risk observed after smoking cessation.

Wheeze, cough and sputum (W/C/S)

In general, wheeze, cough and sputum (W/C/S) are common symptoms among individuals with COPD (Scherer Citation2006), while cough is significantly associated with a number of other chronic respiratory diseases such as chronic bronchitis, bronchiectasis and asthma. Cough, in particular smoker’s cough, is an unequivocal sign of chronic bronchitis, which is itself a well-established risk factor for COPD and associated with poorer prognosis and higher mortality in patients with this disease (Putcha et al. Citation2014). It has been reported that chronic bronchitis, defined as cough and phlegm production on most days for 3 months or more in adults <50 years old (but not in subjects >50 years) represents an early marker of susceptibility to effects of cigarette smoking on COPD mortality (Vestbo Citation2014), while persistent wheeze, chronic cough and chronic phlegm together with dyspnoea were found to have strong predictive value for a decline in FEV1 and were, therefore, suggested to be suitable diagnostic tools for therapeutic intervention (Guerra et al. Citation2009). Cough (most frequently productive cough) as well as wheeze and breathlessness are typical symptoms occurring in all stages and types of lung cancer and have some predictive value for the mortality in this disease (Sherman et al. Citation1992). It has been suggested that smoker’s cough is a symptom for a disease (chronic bronchitis) rather than a mere biomarker of an upcoming respiratory disorder (Vollmer et al. Citation1989, Lange et al. Citation2003, Molassiotis et al. Citation2010a), and that it can be considered as a life threatening condition (Vestbo Citation2014). In general, there is a reduction in the respiratory symptoms of chronic cough, chronic phlegm production, wheezing and shortness of breath following smoking cessation (Gillissen Citation2011). Recent studies suggest that a single exposure to an e-cigarette (nicotine vapour) can enhance the cough reflex which is suppressed in otherwise healthy cigarette smokers (Dicpinigaitis et al. Citation2016).

Overall, while W/C/S symptoms are suitable clinical biomarkers for the management of respiratory diseases, their suitability as biomarkers of effect for use in smoking/cessation/switching studies is limited, mainly due to the fact that long-term studies (lasting at least for months–years) are required to obtain valid data.

4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL)

4-(Methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) is one of seven known tobacco-specific nitrosamines (TSNA). Although levels of NNAL are relatively low in cigarette smoke, they are relatively much higher in smokers since NNAL is the main metabolite of NNK (4-(methynitrosoamino)-1-(3-pyridyl)-1-butanone) (Viegi et al. Citation1988b, Kanner et al. Citation1999). NNK is present in smoke at much higher levels than is NNAL and is metabolized to NNAL in the liver and lungs. Of the seven known TSNAs, NNK is one of the most potent carcinogens in cigarette smoke (Hecht et al. Citation2013). Since NNAL is the principal NNK metabolite in human lung (Hecht Citation2008), NNAL could be considered an important biomarker (Richter et al. Citation2009) of both exposure and effect (particularly given that NNK can only be detected in very low levels in body fluids sampled from smokers).

The literature searches did not identify any studies that attempted to determine if there was a specific correlation of NNAL levels and risk of a CVD endpoint, although one study had investigated the association of total NNAL levels with various biomarkers for CVD (Wei et al. Citation2016). In addition, only one study was identified that concerned NNAL and non-malignant respiratory disease (asthma) (Liu et al. Citation2011). Most of the studies identified have investigated a link between NNAL levels and cancer [primarily lung cancer since NNK, the precursor of NNAL, has been reported to cause lung cancer in all animal species irrespective of the route of administration (Ho et al. Citation2013b)].

Almost all of the studies identified in the searches demonstrated a good correlation between NNAL and other markers of cigarette exposure, e.g. nicotine (Hecht Citation1998) and cigarette consumption (Xia et al. Citation2011, Benowitz et al. Citation2012, Agaku et al. Citation2014). Most studies have shown a relatively slow rate of decline of NNAL following smoking cessation with half-lives of 8–23 days (Hertsgaard et al. Citation2008, Calapai et al. Citation2009).

It is important to note that a recent study investigated levels of total NNAL (free NNAL and NNAL glucuronides) in smokers of a large number of different e-cigarette brands compared to smokers of conventional cigarettes (Carmella et al. Citation2009, Goniewicz et al. Citation2009, Stepanov et al. Citation2009). Although this was a relatively small study (28 e-cigarette users, 17 conventional cigarette smokers), the results were highly significant, with a geometric mean of total NNAL of 0.02 pmol/mL in e-cigarette users and a geometric mean of 1.48 pmol/mL in conventional cigarette users (p < 0.0001) suggesting that e-cigarette users are exposed to far less total NNAL, and thus NNK in smoke, than are conventional smokers.

The earliest study that investigated a link between NNAL and lung cancer in humans was published in 2009 (Hecht et al. Citation2015) This study used a nested case control design to assess the association between total NNAL and lung cancer in 100 smoking lung cancer cases and 100 smoking controls drawn from the Prostate, Lung, Colorectal and Ovarian Cancer Screening Trial (PLCO). NNAL was the only factor statistically significantly associated with the risk of lung cancer following adjustment for other factors associated with lung cancer including age at randomization, family history of lung cancer, cotinine levels and years of cigarette smoking. The OR for the association of NNAL with lung cancer for a unit standard deviation increase (40 fmol/mL of serum) was 1.57 (95% CI, 1.08–2.28).

In the same year, the first of two papers that investigated the association of total NNAL levels with lung cancer risk using two Far Eastern cohorts was published (Church et al. Citation2009). The first cohort was the Shanghai Cohort study, which consisted of 18,244 men enrolled from 1 January 1986 to 30 September 1989. The second was the Singapore Chinese Health study, which consisted of 63,247 men and women enrolled between 1 April 1993 and 31 December 1998. Once again, a nested case control design was used. A total of 259 cases of smoking-related lung cancer diagnosed by March 1997 were matched to an equal number of controls in the Shanghai study, while the Singapore study consisted of 99 cases and an equal number of controls. Adjusted [age, year of interview, year of sample collection, study location, number of cigarettes per day (cpd), number of years smoking and cotinine levels] odds ratios (ORs) were calculated for each tertile of total NNAL and compared to the first tertile as reference. For both studies, combined the OR for the second total NNAL tertile was 1.43 (95% CI, 0.86–2.37), while for the third tertile OR =2.11 (95% CI, 1.25–3.54).

The second paper, published in 2011, focussed solely on the results obtained for the Shanghai cohort study (Yuan et al. Citation2009). This paper presented results on a larger number of individuals than did the previous publication, in that additional cases of lung cancer had been diagnosed. As a consequence, data were available for 476 cases and controls. The results for total NNAL, adjusted for the same covariates used above, were OR (second tertile) = 1.67 (95% CI, 1.13–2.47) and OR (third tertile) = 1.98 (95% CI, 1.32–2.98), results that are clearly comparable to the results published in the earlier paper. One last result of this paper should be noted. Of five factors that were investigated in this study in a multiple factors model, including cigarettes/day, years smoking and total cotinine, NNAL had the weakest association with lung cancer risk (OR =1.28; 95% CI, 1.02–1.59). The authors hypothesize that this may be because of the low levels of TSNA in Chinese cigarettes.

It has been known for some time that changes in TSNA levels in smoke are widely different depending on the country in which the cigarettes are manufactured. This is particularly true for Chinese cigarettes and US cigarettes, with a difference of somewhat more than 10-fold (Yuan et al. Citation2011). However, no real comparison of risk associated with smoking as a function of TSNA smoke levels has yet been published although this point was recently addressed by Canadian investigators (Wu et al. Citation2005) who conducted a study in which total NNAL levels were measured in 507 Canadian smokers. The mean level of total NNAL in this sample was 71.2 pg/mL (95%CI, 63.1–80.4), considerably less than the level in a large American study by Xia et al. which reported total NNAL levels of 299 pg/mL (95% CI, 253–353) in 1373 smokers (Czoli and Hammond Citation2015). The authors question, however, “…whether the differences between US and Canadian cigarettes translate into meaningful differences in risk for these populations.”

There is a clear association between NNAL and the smoking of conventional cigarettes and an unequivocal decline in NNAL following smoking cessation as well as switching to electronic cigarettes, making it a valid biomarker of exposure suitable for use in smoking cessation/switching studies. The situation is less clear with respect to the utility of NNAL levels as a biomarker of effect. As can be seen from the above results, the association of NNAL levels with lung cancer risk appears to be weak. As has been pointed out, however, the levels of NNK, the source of NNAL, are very low in Chinese cigarettes, and much of the data presented were obtained from Chinese smokers. However, the relationship of a sizable difference in total NNAL levels in American and Canadian smokers has been questioned as a predictor of risk. In a meta-analysis based on 40 US and 22 Chinese studies it was found that among ever smokers (i.e. current and former smokers combined) lung cancer mortality (per 100,000), in the 70–74 year age range, was 286.7 in the US and 315.6 in China (Lee and Forey Citation2013). It could be expected that if TSNA was a causal factor, levels should be similar in Chinese and US cigarettes. In contrast, TSNA levels have been found to be substantially higher in US than in Chines cigarettes (Ashley et al. Citation2003). Nevertheless, until a definitive study has been conducted, such as a comparison of age adjusted lung cancer rates in Chinese and American smokers, it seems best to err on the side of safety and assume that NNAL is also a biomarker of risk.

Summary and conclusion

summarizes the interpretations of this literature review on these 25 potential biomarkers and outlines their relationship to smoking, smoking cessation and respiratory and cardiovascular disease (graded by the authors based on the literature findings).

Table 6. Summary of findings for each biomarker investigated with respect to its relationship to smoking, smoking cessation and two smoking-related disease groups.

An ideal biomarker for use in smoking/cessation/switching studies would be one that was associated with the effect of smoking cigarettes, was associated with at least one smoking-related disease and declined following cessation/smoking reduction or switching to a reduced risk product. It is clear from analysing the information retrieved from the literature that few of the biomarkers investigated fulfil all of these criteria. It is also clear that one single biomarker is unlikely to provide all the information needed. Useful biomarkers should consistently relate to a disease, represent biologically real pathways and change as the pathology changes. Many of the studies alluded to in this review might have failed to find high sensitivity and high specificity biomarkers due to the sometimes small number of subjects, various analysis techniques or data interpretation (Xia et al. Citation2011). It is unfortunate that highly standardized assays for biomarker identification and analysis are not frequently found, and although thousands of publications have been written, only a small number of successfully validated biomarkers have entered clinical use (Issaq et al. Citation2011, Poste Citation2011). A further complication is that many of the reviewed biomarkers reflect a complex network of multiple interacting molecular pathways coupled with their adaptive feedback and cross-talk loops which masks the chance of the biomarker to reflect responses of the system as a whole. For many reasons the relationship between markers and outcomes are neither unique nor simple (Drucker and Krapfenbauer Citation2013) and can be modified by many factors. Choosing a panel of biomarkers that will be able to reflect changes that might be expected within the duration of the study and associated with more than one smoking-related disease would seem an optimal approach. Epidemiological data being available for more than half a century, the chronic disease risks of cigarette smoking are well understood, as are the reductions in risk achieved over time by smoking cessation. In contrast, the chronic disease risk assessment of switching from cigarettes to novel modified risk tobacco products must rest on biomarker data obtained in pre-market studies. In spite of the uncertainty implied by this restriction to indirect evidence, not making such products available to consumers is not a realistic regulatory strategy. The reason is that this null option would not be precautionary but rather would entail doing harm, by restricting cigarette smokers who do not quit using tobacco to consume products of a category with well-known and considerable disease risks.

The risk estimation problem with analysing biomarker data is to quantify the degree of effective dose reduction that is achieved by cigarette smokers switching to a novel modified risk tobacco product, based on effects observed in measurements which are indicative of, but are not directly quantifying, risk reduction. Rather than attempting a direct estimation of the degree of risk reduction achieved by switching to modified risk tobacco products as compared to continued smoking, a re-parametrization of the estimation problem has recently been undertaken (Martin et al. Citation2017), making two reasonable assumptions. The first is on the informativeness of a comprehensive set of biomarkers measured in multiple clinical studies with regard to predicting chronic disease risks to be halfway between 0 and 100%. The second assumption is that, for each biomarker to contribute to a reduction in the effective dose, the minimal change upon switching from smoking to a novel product must be at least half of that observed upon smoking cessation.

Sensitivity analyses based on variations of these conservative parameter settings as well as on the omission of individual biomarkers from the estimation supported the robustness of the risk assessment. Future epidemiological health outcomes data can be used for assessing the model predictions and for quantifying the predictive accuracy of the observed biomarker effects.

Finally, limiting the period of the search (2008–2017) was intended to provide recent evidence supporting the use or not of the biomarkers of interest. However, for some biomarkers (e.g. TXB2), the recent literature provided little information and in these cases, we have resorted to using publications made prior to 2008.

Acknowledgements

We should like to dedicate this manuscript to our friend and colleague, Dr Edward (Ted) Sanders who sadly passed away on 4 July 2017. The authors would like to thank Dr Nathalie Debbas (Medi-Axe, Clamart, France) for contributions made to the sections on FEV1 and 8-epi-PGF2α.

Disclosure statement

All costs related to the preparation of this review have been covered by Philip Morris International and RW and FL are employees of the company.

References

  • Aaron, C.P., et al., 2015. Intercellular adhesion molecule 1 and progression of percent emphysema: the MESA lung study. Respiratory medicine, 109, 255–264.
  • Abdi, A.I., et al., 2014. Measuring soluble ICAM-1 in African populations. PLoS one, 9, e108956.
  • Adukauskiene, D., et al., 2016. Clinical relevance of high sensitivity C-reactive protein in cardiology. Medicina (Kaunas), 52, 1–10.
  • Agaku, I.T., Vardavas, C.I., and Connolly, G.N., 2014. Cigarette rod length and its impact on serum cotinine and urinary total NNAL levels, NHANES 2007-2010. Nicotine &Amp; tobacco research, 16, 100–107.
  • Agarwal, R., et al., 2013. The relationship between C-reactive protein and prognostic factors in chronic obstructive pulmonary disease. Multidisciplinary respiratory medicine, 8, 63.
  • Aggarwal, A.N., Gupta, D., and Jindal, S.K., 2006. The relationship between FEV1 and peak expiratory flow in patients with airways obstruction is poor. Chest, 130, 1454–1461.
  • Ahn, J., et al., 2009. Prediagnostic total and high-density lipoprotein cholesterol and risk of cancer. Cancer epidemiology, biomarkers & prevention, 18, 2814–2821.
  • Al-Awadhi, A.M., et al., 2008. Effects of cigarette smoking on hematological parameters and von Willebrand factor functional activity levels in asymptomatic male and female Arab smokers. Medical principles and practice, 17, 149–153.
  • Al Ghobain, M.O., et al., 2014. Spirometric reference values for healthy nonsmoking Saudi adults. The clinical respiratory journal, 8, 72–78.
  • Al Rifai, M., et al., 2017. The relationship between smoking intensity and subclinical cardiovascular injury: the Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis, 258, 119–130.
  • Allen, S.S., Hatsukami, D., and Gorsline, J., 1994. Cholesterol changes in smoking cessation using the transdermal nicotine system. Transdermal Nicotine Study Group. Preventive medicine, 23, 190–196.
  • Allin, K.H., Bojesen, S.E., and Nordestgaard, B.G., 2016. Inflammatory biomarkers and risk of cancer in 84,000 individuals from the general population. International journal of cancer, 139, 1493–1500.
  • An, G., et al., 2016. A novel physiology-based mathematical model to estimate red blood cell lifespan in different human age groups. The AAPS journal, 18, 1182–1191.
  • Anan, F., et al., 2006. Smoking is associated with insulin resistance and cardiovascular autonomic dysfunction in type 2 diabetic patients. European journal of clinical investigation, 36, 459–465.
  • Anatoliotakis, N., et al., 2013. Myeloperoxidase: expressing inflammation and oxidative stress in cardiovascular disease. Current topics in medicinal chemistry, 13, 115–138.
  • Andelid, K., et al., 2007. Myeloperoxidase as a marker of increasing systemic inflammation in smokers without severe airway symptoms. Respiratory medicine, 101, 888–895.
  • Andersson, A., et al., 2001. Hyperhomocysteinemia and changed plasma thiol redox status in chronic obstructive pulmonary disease. Clinical chemistry and laboratory medicine, 39, 229–233.
  • Annor, F., et al., 2017. African ancestry gradient is associated with lower systemic F2-isoprostane levels. Oxidative medicine and cellular longevity, 2017, 8319176.
  • Anthonisen, N.R., et al., 1994. Effects of smoking intervention and the use of an inhaled anticholinergic bronchodilator on the rate of decline of FEV1. The Lung Health Study. JAMA, 272, 1497–1505.
  • Antonopoulos, C.N., et al., 2014. The role of soluble P selectin in the diagnosis of venous thromboembolism. Thrombosis research, 133, 17–24.
  • Arican, O., et al., 2005. Serum levels of TNF-alpha, IFN-gamma, IL-6, IL-8, IL-12, IL-17, and IL-18 in patients with active psoriasis and correlation with disease severity. Mediators of inflammation, 2005, 273–279.
  • Ashley, D.L., et al., 2003. Tobacco-specific nitrosamines in tobacco from U.S. brand and non-U.S. brand cigarettes. Nicotine & tobacco research, 5, 323–331.
  • Asthana, A., et al., 2010. Effects of smoking intensity and cessation on inflammatory markers in a large cohort of active smokers. American heart journal, 160, 458–463.
  • Baccarelli, A., et al., 2007. Air pollution, smoking, and plasma homocysteine. Journal of thrombosis and haemostasis, 115, 176–181.
  • Baggio, B., et al., 2002. Effects of cigarette smoking on glomerular structure and function in type 2 diabetic patients. Journal of the American society of nephrology, 13, 2730–2736.
  • Balasubramaniam, M., Sivapalan, K., and Thuvarathipan, R., 2014. Lung function parameters of healthy Sri Lankan Tamil young adults. Ceylon medical journal, 59, 49–53.
  • Baldawa, P.S., et al., 2016. Levels of salivary thiocyanate and its relation with occurrence of micronuclei using exfoliative cytology in smokers and nonsmokers. Indian journal of dental research, 27, 568–573.
  • Baldus, S., et al., 2003. Myeloperoxidase serum levels predict risk in patients with acute coronary syndromes. Circulation, 108, 1440–1445.
  • Ballman, K.V., 2015. Biomarker: predictive or prognostic?. Journal of clinical oncology, 33, 3968–3971.
  • Barnes, P.J., 2009. The cytokine network in chronic obstructive pulmonary disease. American journal of respiratory cell and molecular biology, 41, 631–638.
  • Barrera, G., 2012. Oxidative stress and lipid peroxidation products in cancer progression and therapy. ISRN oncology, 2012, 137289.
  • Barrow, S.E., et al., 1989. Cigarette smoking: profiles of thromboxane- and prostacyclin-derived products in human urine. Biochimica et biophysica acta, 993 (1), 121–127.
  • Bartholo, T.P., Costa, C.H., and Rufino, R., 2014. Evaluation of von Willebrand factor in COPD patients. Jornal brasileiro de pneumologia, 40, 373–379.
  • Bartsch, H., et al., 1989. Human exposure to endogenous N-nitroso compounds: quantitative estimates in subjects at high risk for cancer of the oral cavity, oesophagus, stomach and urinary bladder. Cancer surveys, 8, 335–362.
  • Barua, R.S., et al., 2001. Dysfunctional endothelial nitric oxide biosynthesis in healthy smokers with impaired endothelium-dependent vasodilatation. Circulation, 104, 1905–1910.
  • Bauman, W.A., et al., 1998. Ethnicity effect on the serum lipid profile in persons with spinal cord injury. Archives of physical medicine and rehabilitation, 79, 176–180.
  • Bazzano, L.A., et al., 2003. Relationship between cigarette smoking and novel risk factors for cardiovascular disease in the United States. Annals of internal medicine, 138, 891–897.
  • Becklake, M.R., 1986. Concepts of normality applied to the measurement of lung function. The American journal of medicine, 80, 1158–1164.
  • Belda, J., et al., 2000. Induced sputum cell counts in healthy adults. American journal of respiratory and critical care medicine, 161, 475–478.
  • Bellisarii, F.L., Gallina, S., and De Caterina, R., 2001. Tumor necrosis factor-alpha and cardiovascular diseases. Italian heart journal, 2, 408–417.
  • Benowitz, N.L., et al., 2012. Smoking behavior and exposure to tobacco toxicants during 6 months of smoking progressively reduced nicotine content cigarettes. Cancer epidemiology, biomarkers & prevention, 21, 761–769.
  • Bermudez, E.A., et al., 2002. Relation between markers of systemic vascular inflammation and smoking in women. American journal of cardiology, 89, 1117–1119.
  • Bhatt, S.P., et al., 2016. Association between functional small airway disease and FEV1 decline in chronic obstructive pulmonary disease. American journal of respiratory and critical care medicine, 194, 178–184.
  • Bialas, A.J., et al., 2016. The role of mitochondria and oxidative/antioxidative imbalance in pathobiology of chronic obstructive pulmonary disease. Oxidative medicine and cellular longevity, 2016, 7808576.
  • Bielinski, S.J., et al., 2015. P-selectin and subclinical and clinical atherosclerosis: the Multi-Ethnic Study of Atherosclerosis (MESA). Atherosclerosis, 240, 3–9.
  • Biomarkers Definitions Working Group, 2001. Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clinical pharmacology & therapeutics, 69, 89–95.
  • Bird, Y. and Staines-Orozco, H., 2016. Pulmonary effects of active smoking and secondhand smoke exposure among adolescent students in Juarez, Mexico. International journal of chronic obstructive pulmonary disease, 11, 1459–1467.
  • Bjerg, A., et al., 2013. Higher risk of wheeze in female than male smokers. Results from the Swedish GA 2 LEN study. PLoS one, 8, e54137.
  • Bjorkhem, I., 2002. Do oxysterols control cholesterol homeostasis? The journal of clinical investigation, 110, 725–730.
  • Bjorkhem, I., Meaney, S., and Diczfalusy, U., 2002. Oxysterols in human circulation: which role do they have?. Current opinion in lipidology, 13, 247–253.
  • Blann, A.D., 2006. Plasma von Willebrand factor, thrombosis, and the endothelium: the first 30 years. Thrombosis and haemostasis, 95, 49–55.
  • Blann, A.D., et al., 1997a. Soluble P-selectin in hyperlipidaemia with and without symptomatic vascular disease: relationship with von Willebrand factor. Blood Coagulation & Fibrinolysis, 8, 200–204.
  • Blann, A.D., et al., 1998. The influence of acute smoking on leucocytes, platelets and the endothelium. Atherosclerosis, 141, 133–139.
  • Blann, A.D. and Mccollum, C.N., 1993. Adverse influence of cigarette smoking on the endothelium. Thrombosis and haemostasis, 70, 707–711.
  • Blann, A.D., Nadar, S.K., and Lip, G.Y., 2003. The adhesion molecule P-selectin and cardiovascular disease. European heart journal, 24, 2166–2179.
  • Blann, A.D., Steele, C., and Mccollum, C.N., 1997b. The influence of smoking and of oral and transdermal nicotine on blood pressure, and haematology and coagulation indices. Thrombosis and haemostasis, 78, 1093–1096.
  • Blann, A.D., Steele, C., and Mccollum, C.N., 1997c. The influence of smoking on soluble adhesion molecules and endothelial cell markers. Thrombosis research, 85, 433–438.
  • Blidberg, K., et al., 2013. Adhesion molecules in subjects with COPD and healthy non-smokers: a cross sectional parallel group study. Respiratory research, 14, 47.
  • Boeing, H., et al., 2000. Association between glycated hemoglobin and diet and other lifestyle factors in a nondiabetic population: cross-sectional evaluation of data from the Potsdam cohort of the European Prospective Investigation into Cancer and Nutrition Study. The American journal of clinical nutrition, 71, 1115–1122.
  • Boltri, J.M., et al., 2005. Hemoglobin A1c levels in diagnosed and undiagnosed black, Hispanic, and white persons with diabetes: results from NHANES 1999-2000. Ethnicity & disease, 15, 562–567.
  • Borrill, Z.L., et al., 2008. Non-invasive biomarkers and pulmonary function in smokers. International journal of chronic obstructive pulmonary disease, 3, 171–183.
  • Bostrom, L., Linder, L.E., and Bergstrom, J., 1999. Smoking and crevicular fluid levels of IL-6 and TNF-alpha in periodontal disease. Journal of clinical periodontology, 26, 352–357.
  • Bottino, D.A., et al., 2015. Relationship between biomarkers of inflammation, oxidative stress and endothelial/microcirculatory function in successful aging versus healthy youth: a transversal study. BMC Geriatrics, 15, 41.
  • Bouloukaki, I., et al., 2009. Maintained smoking cessation for 6 months equilibrates the percentage of sputum CD8+ lymphocyte cells with that of nonsmokers. Mediators of inflammation, 2009, 812102.
  • Bozinovski, S., et al., 2016. COPD and squamous cell lung cancer: aberrant inflammation and immunity is the common link. British journal of pharmacology, 173, 635–648.
  • Brennan, M.L., et al., 2003. Prognostic value of myeloperoxidase in patients with chest pain. The new England journal of medicine, 349, 1595–1604.
  • Brignac-Huber, L., Reed, J.R., and Backes, W.L., 2011. Organization of NADPH-cytochrome P450 reductase and CYP1A2 in the endoplasmic reticulum–microdomain localization affects monooxygenase function. Molecular pharmacology, 79, 549–557.
  • Brilakis, E.S., et al., 2008. Influence of race and sex on lipoprotein-associated phospholipase A2 levels: observations from the Dallas Heart Study. Atherosclerosis, 199, 110–115.
  • Brindicci, C., et al., 2005. Exhaled nitric oxide from lung periphery is increased in COPD. The European respiratory journal, 26, 52–59.
  • Burgel, P.R., 2011. The role of small airways in obstructive airway diseases. European respiratory review, 20, 23–33.
  • Busse, P.J., et al., 2017. Effect of aging on sputum inflammation and asthma control. The journal of allergy and clinical immunology, 139, 1808–1818.e6.
  • Butkiewicz, A.M., et al., 2006. Does smoking affect thrombocytopoiesis and platelet activation in women and men? Advances in medical science, 51, 123–126.
  • Cahill, K., et al., 2013. Pharmacological interventions for smoking cessation: an overview and network meta-analysis. The Cochrane database of systematic review, 5, CD009329.
  • Calapai, G., et al., 2009. A cross-sectional investigation of biomarkers of risk after a decade of smoking. Inhalation toxicology, 21, 1138–1143.
  • Campagna, D., et al., 2016. Changes in breathomics from a 1-year randomized smoking cessation trial of electronic cigarettes. European journal of clinical investigation, 46, 698–706.
  • Campion, E.W., Delabry, L.O., and Glynn, R.J., 1988. The effect of age on serum albumin in healthy males: report from the Normative Aging Study. Journal of gerontology, 43, M18–M20.
  • Carmella, S.G., et al., 2009. Effects of smoking cessation on eight urinary tobacco carcinogen and toxicant biomarkers. Chemical research in toxicology, 22, 734–741.
  • Carpagnano, G.E., et al., 2007. IL-2, TNF-alpha, and leptin: local versus systemic concentrations in NSCLC patients. Oncological research, 16, 375–381.
  • Carpagnano, G.E., et al., 2013. Aging and airway inflammation. Aging clinical and experimental research, 25, 239–245.
  • Carroll, A., 2016. Utilizing selection biomarkers in clinical trials: is this the future of drug development? Biomarkers in medicine, 10, 939–941.
  • Casale, R., et al., 1991. Impaired pulmonary function in schoolchildren exposed to passive smoking. Detection by questionnaire and urinary cotinine levels. Respiration, 58, 198–203.
  • Caslake, M.J. and Packard, C.J., 2003. Lipoprotein-associated phospholipase A2 (platelet-activating factor acetylhydrolase) and cardiovascular disease. Current opinion in lipidology, 14, 347–352.
  • Catella, F., et al., 1986. 11-Dehydrothromboxane B2: a quantitative index of thromboxane A2 formation in the human circulation. Proceedings of the national academy of sciences of the United States of America, 83, 5861–5865.
  • Cellai, A.P., et al., 2014. Hyperhomocysteinemia in patients with pulmonary embolism is associated with impaired plasma fibrinolytic capacity. Journal of thrombosis and thrombolysis, 38, 45–49.
  • Celli, B.R., et al., 2012. Inflammatory biomarkers improve clinical prediction of mortality in chronic obstructive pulmonary disease. American journal of respiratory and critical care medicine, 185, 1065–1072.
  • Centers for Disease Control and Prevention (US); National Center for Chronic Disease Prevention and Health Promotion (US); Office on Smoking and Health (US), 2010. How tobacco smoke causes disease: the biology and behavioral basis for smoking-attributable disease: a report of the surgeon general. Atlanta (GA): Centers for Disease Control and Prevention.
  • Cerveri, I., et al., 2008. Underestimation of airflow obstruction among young adults using FEV1/FVC <70% as a fixed cut-off: a longitudinal evaluation of clinical and functional outcomes. Thorax, 63, 1040–1045.
  • Chalmers, G.W., et al., 2001. Smoking and airway inflammation in patients with mild asthma. Chest, 120, 1917–1922.
  • Chambers, D.C., Boldy, D.A., and Ayres, J.G., 1999. Chronic respiratory symptoms, von Willebrand factor and longitudinal decline in FEV1. Respiratory medicine, 93, 726–733.
  • Chamnan, P., et al., 2013. Change in HbA1c over 3 years does not improve the prediction of cardiovascular disease over and above HbA1c measured at a single time point. Diabetologia, 56, 1004–1011.
  • Chan, S.C., et al., 2008. Upregulation of ICAM-1 expression in bronchial epithelial cells by airway secretions in bronchiectasis. Respiratory medicine, 102, 287–298.
  • Chang, M.H., et al., 2011. Racial/ethnic variation in the association of lipid-related genetic variants with blood lipids in the US adult population. Circulation: cardiovascular genetics, 4, 523–533.
  • Chase, H.P., et al., 1991. Cigarette smoking increases the risk of albuminuria among subjects with type I diabetes. JAMA, 265, 614–617.
  • Chaudhuri, R., et al., 2014. Effects of a FLAP inhibitor, GSK2190915, in asthmatics with high sputum neutrophils. Pulmonary pharmacology & therapeutics, 27, 62–69.
  • Chen, M. and Geng, J.G., 2006. P-selectin mediates adhesion of leukocytes, platelets, and cancer cells in inflammation, thrombosis, and cancer growth and metastasis. Archivum immunologiae et therapiae experimentalis, 54, 75–84.
  • Chen, S., et al., 2015. Relationship between increase of serum homocysteine caused by smoking and oxidative damage in elderly patients with cardiovascular disease. International journal of clinical and experimental medicine, 8, 4446–4454.
  • Chevrier, I., et al., 2003. Myeloperoxidase: new polymorphisms and relation with lung cancer risk. Pharmacogenetics, 13, 729–739.
  • Choi, Y.J., et al., 2013. Changes in smoking behavior and adherence to preventive guidelines among smokers after a heart attack. Journal of geriatric cardiology, 10, 146–150.
  • Church, T.R., et al., 2009. A prospectively measured serum biomarker for a tobacco-specific carcinogen and lung cancer in smokers. Cancer epidemiology, biomarkers & prevention, 18, 260–266.
  • Cibella, F., et al., 2016. Lung function and respiratory symptoms in a randomized smoking cessation trial of electronic cigarettes. Clinical science (London), 130, 1929–1937.
  • Cirillo, D.J., Agrawal, Y., and Cassano, P.A., 2002. Lipids and pulmonary function in the Third National Health and Nutrition Examination Survey. American journal of epidemiology, 155, 842–848.
  • Clair, C., et al., 2011. Relationships of cotinine and self-reported cigarette smoking with hemoglobin A1c in the U.S.: results from the National Health and Nutrition Examination Survey, 1999-2008. Diabetes care, 34, 2250–2255.
  • Coates, A.L., et al., 2016. Reference Equations for Spirometry in the Canadian Population. Annals of the American thoracic society, 13, 833–841.
  • Cohen, S.I., Deane, M., and Goldsmith, J.R., 1969. Carbon monoxide and survival from myocardial infarction. Archives of environmental health, 19, 510–517.
  • Cohen, W., et al., 2012. ABO blood group and von Willebrand factor levels partially explained the incomplete penetrance of congenital thrombophilia. Arteriosclerosis, thrombosis, and vascular biology, 32, 2021–2028.
  • Committee on Human Biomonitoring for Environmental Toxicants. 2006. Human biomonitoring for environmental chemicals. Washington: National Academies Press.
  • Committee on Scientific Standards for Studies on Modified Risk Tobacco Products. 2012. Scientific standards for studies on modified risk tobacco products (2012). Washington: The National Academies Press.
  • Conese, M., et al., 2014. Evaluation of genome-wide expression profiles of blood and sputum neutrophils in cystic fibrosis patients before and after antibiotic therapy. PLoS one, 9, e104080.
  • Conlan, M.G., et al., 1993. Associations of factor VIII and von Willebrand factor with age, race, sex, and risk factors for atherosclerosis. The Atherosclerosis Risk in Communities (ARIC) Study. Thrombosis and haemostasis, 70, 380–385.
  • Cooper, J.K. and Gardner, C., 1989. Effect of aging on serum albumin. Journal of the American geriatrics society, 37, 1039–1042.
  • Corradi, L., et al., 1993. Association between smoking and micro-albuminuria in hypertensive patients with type 2 diabetes mellitus. Journal of hypertension supplement, 11, S190–S191.
  • Cosio, M., et al., 1978. The relations between structural changes in small airways and pulmonary-function tests. New England journal of medicine, 298, 1277–1281.
  • Cowman, J., et al., 2015. Age-related changes in platelet function are more profound in women than in men. Scientific reports, 5, 12235.
  • Craig, W.Y., Palomaki, G.E., and Haddow, J.E., 1989. Cigarette smoking and serum lipid and lipoprotein concentrations: an analysis of published data. BMJ (clinical research ed.), 298, 784–788.
  • Crapo, R.O. and Jensen, R.L., 2003. Standards and interpretive issues in lung function testing. Respiratory care, 48, 764–772.
  • Czoli, C.D. and Hammond, D., 2015. TSNA Exposure: Levels of NNAL Among Canadian Tobacco Users. Nicotine &Amp; tobacco research, 17, 825–830.
  • Dahl, M., et al., 2007. C-reactive protein as a predictor of prognosis in chronic obstructive pulmonary disease. American journal of respiratory and critical care medicine, 175, 250–255.
  • Dalaveris, E., et al., 2009. VEGF, TNF-alpha and 8-isoprostane levels in exhaled breath condensate and serum of patients with lung cancer. Lung cancer, 64, 219–225.
  • Danesh, J., et al., 1998. Association of fibrinogen, C-reactive protein, albumin, or leukocyte count with coronary heart disease: meta-analyses of prospective studies. JAMA, 279, 1477–1482.
  • Danesh, J., et al., 2004. C-reactive protein and other circulating markers of inflammation in the prediction of coronary heart disease. New England journal of medicine, 350, 1387–1397.
  • Davidson, M.B. and Schriger, D.L., 2010. Effect of age and race/ethnicity on HbA1c levels in people without known diabetes mellitus: implications for the diagnosis of diabetes. Diabetes research and clinical practice, 87, 415–421.
  • De Bree, A., et al., 2002. Homocysteine determinants and the evidence to what extent homocysteine determines the risk of coronary heart disease. Pharmacological reviews, 54, 599–618.
  • Dedonder-Decoopman, E., et al., 1980. Plasma levels of VLDL- + LDL-cholesterol, HDL-cholesterol, triglycerides and apoproteins B and A-I in a healthy population–influence of several risk factors. Atherosclerosis, 37, 559–568.
  • Delporte, C., et al., 2013. Low-density lipoprotein modified by myeloperoxidase in inflammatory pathways and clinical studies. Mediators of inflammation, 2013, 971579.
  • Deneva-Koycheva, T.I., et al., 2011. Serum levels of siCAM-1, sVCAM-1, sE-selectin, sP-selectin in healthy Bulgarian people. Folia medicine (Plovdiv), 53, 22–28.
  • Deng, Z.C., et al., 2014. C-reactive protein as a prognostic marker in chronic obstructive pulmonary disease. Experimental and therapeutic medicine, 7, 443–446.
  • Derin, D., et al., 2008. Serum levels of apoptosis biomarkers, survivin and TNF-alpha in nonsmall cell lung cancer. Lung cancer (Amsterdam, Netherlands), 59, 240–245.
  • Diaz-Guzman, E. and Mannino, D.M., 2014. Epidemiology and prevalence of chronic obstructive pulmonary disease. Clinics in chest medicine, 35, 7–16.
  • Dicpinigaitis, P.V., et al., 2016. Effect of e-cigarette use on cough reflex sensitivity. Chest, 149, 161–165.
  • Diczfalusy, U., et al., 2008. 4Beta-hydroxycholesterol is a new endogenous CYP3A marker: relationship to CYP3A5 genotype, quinine 3-hydroxylation and sex in Koreans, Swedes and Tanzanians. Pharmacogenetics and genomics, 18, 201–208.
  • Dogne, J.M., Hanson, J., and Pratico, D., 2005. Thromboxane, prostacyclin and isoprostanes: therapeutic targets in atherogenesis. Trends in pharmacological science, 26, 639–644.
  • Dominguez-Rodriguez, A., and Abreu-Gonzalez, P., 2011. Current role of myeloperoxidase in routine clinical practice. Expert review of cardiovascular therapy, 9, 223–230.
  • Dotevall, A., et al., 1992. Cigarette smoking increases thromboxane A2 formation without affecting platelet survival in young healthy females. Thrombosis and haemostasis, 68, 583–588.
  • Drenos, F., Miller, G.J., and Humphries, S.E., 2007. Increase of plasma fibrinogen levels and variability with age in a sample of middle aged healthy men. Annals of human genetics, 71, 43–53.
  • Drucker, E. and Krapfenbauer, K., 2013. Pitfalls and limitations in translation from biomarker discovery to clinical utility in predictive and personalised medicine. EPMA journal, 4, 7.
  • Eeg-Olofsson, K., et al., 2010. New aspects of HbA1c as a risk factor for cardiovascular diseases in type 2 diabetes: an observational study from the Swedish National Diabetes Register (NDR). Journal of internal medicine, 268, 471–482.
  • Eikelboom, J.W., et al., 2002. Aspirin-resistant thromboxane biosynthesis and the risk of myocardial infarction, stroke, or cardiovascular death in patients at high risk for cardiovascular events. Circulation, 105, 1650–1655.
  • Ekim, M., et al., 2015. Study on relationships among deep vein thrombosis, homocysteine & related B group vitamins. Pakistan journal of medical sciences, 31, 398–402.
  • El-Sammak, M., et al., 2004. Elevated plasma homocysteine is positively associated with age independent of C677T mutation of the methylenetetrahydrofolate reductase gene in selected Egyptian subjects. International journal of medical science, 1, 181–192.
  • Ernst, E. and Resch, K.L., 1993. Fibrinogen as a cardiovascular risk factor: a meta-analysis and review of the literature. Annals of internal medicine, 118, 956–963.
  • Everett, C.J., et al., 2007. Smoking, fibrinogen and cancer mortality. Journal of the national medical association, 99, 328–333.
  • Fatima, N., et al., 2017. Emerging role of Interleukins IL-23/IL-17 axis and biochemical markers in the pathogenesis of type 2 diabetes: association with age and gender in human subjects. International journal of biological macromolecules, 105, 1279–1288.
  • Faux, S.P., et al., 2009. The role of oxidative stress in the biological responses of lung epithelial cells to cigarette smoke. Biomarkers, 14Suppl1, 90–96.
  • Ference, B.A. and Mahajan, N., 2013. Effect of long-term exposure to lower low-density lipoprotein cholesterol beginning early in life on the risk of coronary heart disease: a Mendelian randomization analysis. Journal of the American college of cardiology, 61, 1931–1932.
  • Fernandez, J.A., et al., 2012. Systemic inflammation in 222.841 healthy employed smokers and nonsmokers: white blood cell count and relationship to spirometry. Tobacco induced diseases, 10, 7.
  • Fimognari, F.L., et al., 2009. Hyperhomocysteinaemia and poor vitamin B status in chronic obstructive pulmonary disease. Nutrition, metabolism and cardiovascular disease, 19, 654–659.
  • Firouzjahi, A., et al., 2013. Serum C-reactive protein level and distribution in chronic obstructive pulmonary disease versus healthy controls: a case-control study from Iran. Inflammation, 36, 1122–1128.
  • Fleming, T.R. and Demets, D.L., 1996. Surrogate end points in clinical trials: are we being misled?. Annals of internal medicine, 125, 605–613.
  • Flenley, D.C., 1988. Chronic obstructive pulmonary disease. Disease-a-month, 34, 537–599.
  • Ford, E.S., et al., 2013. COPD surveillance-United States, 1999-2011. Chest, 144, 284–305.
  • Forey, B.A., et al., 2013. The effect of quitting smoking on HDL-cholesterol – a review based on within-subject changes. Biomarker research, 1, 26
  • Foss, O.P. and Lund-Larsen, P.G., 1986. Serum thiocyanate and smoking: interpretation of serum thiocyanate levels observed in a large health study. Scandinavian journal of clinical and laboratory investigation, 46, 245–251.
  • Franchini, M. and Lippi, G., 2007. The role of von Willebrand factor in hemorrhagic and thrombotic disorders. Critical reviews in clinical laboratory sciences, 44, 115–149.
  • Franchini, M. and Mannucci, P.M., 2008. Von Willebrand factor: another janus-faced hemostasis protein. Seminars in thrombosis and hemostasis, 34, 663–669.
  • Frankel, D.S., et al., 2008. Von Willebrand factor, type 2 diabetes mellitus, and risk of cardiovascular disease: the Framingham offspring study. Circulation, 118, 2533–2539.
  • Fratta Pasini, A., et al., 2013. Lysophosphatidylcholine and carotid intima-media thickness in young smokers: a role for oxidized LDL-induced expression of PBMC lipoprotein-associated phospholipase A2? PLoS one, 8, e83092.
  • Freedman, B.I., et al., 2005. Relationship between albuminuria and cardiovascular disease in Type 2 diabetes. Journal of the American society of nephrology, 16, 2156–2161.
  • Freeman, D.J., et al., 1993. Smoking and plasma lipoproteins in man: effects on low density lipoprotein cholesterol levels and high density lipoprotein subfraction distribution. European journal of clinical investigation, 23, 630–640.
  • Friedman, G.D., et al., 1973. Smoking habits and the leukocyte count. Archives of environmental health, 26, 137–143.
  • Frondelius, K., et al., 2017. Lifestyle and dietary determinants of serum apolipoprotein A1 and apolipoprotein B concentrations: cross-sectional analyses within a Swedish cohort of 24,984 individuals. Nutrients, 9, 211.
  • Frost-Pineda, K., et al., 2011. Biomarkers of potential harm among adult smokers and nonsmokers in the total exposure study. Nicotine & tobacco research, 13, 182–193.
  • Fu, A. and Nair, K.S., 1998. Age effect on fibrinogen and albumin synthesis in humans. The American journal of physiology, 275, E1023–E1030.
  • Garcia-Larsen, V., et al., 2014. Ventilatory function and cardiovascular disease risk factors: a cross-sectional study in young adults. BMC pulmonary medicine, 14, 206.
  • Garey, K.W., et al., 2004. Markers of inflammation in exhaled breath condensate of young healthy smokers. Chest, 125, 22–26.
  • Ge, P.C., et al., 2016. Synergistic Effect of Lipoprotein-associated phospholipase A2 with classical risk factors on coronary heart disease: a multi-ethnic study in China. Cellular physiology and biochemistry, 40, 953–968.
  • Geng, J.G., Chen, M., and Chou, K.C., 2004. P-selectin cell adhesion molecule in inflammation, thrombosis, cancer growth and metastasis. Current medicinal chemistry, 11, 2153–2160.
  • Gerber, P.A., et al., 2013. Smoking is associated with impaired long-term glucose metabolism in patients with type 1 diabetes mellitus. Nutrition, metabolism and cardiovascular disease, 23, 102–108.
  • Gessner, C., et al., 2005. Exhaled breath condensate cytokine patterns in chronic obstructive pulmonary disease. Respiratory medicine, 99, 1229–1240.
  • Gijsberts, C.M., et al., 2015. Biomarkers of coronary artery disease differ between Asians and Caucasians in the general population. Global heart, 10, 301–311 e11.
  • Gillissen, A., 2011. [COPD: smoker's cough? No, life-threatening!]. MMW fortschritte der medizin, 153, 32.
  • Gnasso, A., et al., 1984. Acute influence of smoking on plasma lipoproteins. Klinische wochenschrift, 62Suppl2, 36–42.
  • Godinho Netto, A.C., et al., 2016. Fraction of exhaled nitric oxide measurements in the diagnoses of asthma in elderly patients. Clinical interventions in aging, 11, 623–629.
  • Golbahar, J., Rezaian, G., and Bararpour, H., 2004. Distribution of plasma total homocysteine concentrations in the healthy Iranians. Clinical biochemistry, 37, 149–151.
  • Gold, D.R., et al., 1996. Effects of cigarette smoking on lung function in adolescent boys and girls. The new England journal of medicine, 335, 931–937.
  • Goniewicz, M.L., et al., 2009. Elimination kinetics of the tobacco-specific biomarker and lung carcinogen 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol. Cancer epidemiology, biomarkers & prevention, 18, 3421–3425.
  • Gordon, S.M. and Davidson, W.S., 2012. Apolipoprotein A-I mimetics and high-density lipoprotein function. Current opinion in endocrinology, diabetes, and obesity, 19, 109–114.
  • Greenblatt, D.J., 1979. Reduced serum albumin concentration in the elderly: a report from the Boston Collaborative Drug Surveillance Program. Journal of the American geriatrics society, 27, 20–22.
  • Gross, M.D., et al., 2012. Circulating soluble intercellular adhesion molecule 1 and subclinical atherosclerosis: the Coronary Artery Risk Development in Young Adults Study. Clinical chemistry, 58, 411–420.
  • Gryglewski, R.J., Dembinska-Kiec, A., and Korbut, R., 1978. A possible role of thromboxane A2 (TXA2) and prostacyclin (PGI2) in circulation. Acta biologica et medica germanica, 37, 715–723.
  • Guerra, S., et al., 2009. Chronic bronchitis before age 50 years predicts incident airflow limitation and mortality risk. Thorax, 64, 894–900.
  • Gulliford, M.C. and Ukoumunne, O.C., 2001. Determinants of glycated haemoglobin in the general population: associations with diet, alcohol and cigarette smoking. European journal of clinical nutrition, 55, 615–623.
  • Gupta, V. and Singh, D., 2013. Critical assessment of the value of sputum neutrophils. COPD, 10, 107–114.
  • Gutierrez, C.M., et al., 2014. [New spirometric reference equations for healthy Chilean adults]. Revista medica de Chile, 142, 143–152.
  • Guven, F.M., et al., 2013. The importance of myeloperoxidase enzyme activity in the pathogenesis of Crimean-Congo haemorrhagic fever. Journal of medical microbiology, 62, 441–445.
  • Guy, H.J., et al., 1994. Inhomogeneity of pulmonary ventilation during sustained microgravity as determined by single-breath washouts. Journal of applied physiology (1985), 76, 1719–1729.
  • Hadfield, K.A., et al., 2013. Myeloperoxidase-derived oxidants modify apolipoprotein A-I and generate dysfunctional high-density lipoproteins: comparison of hypothiocyanous acid (HOSCN) with hypochlorous acid (HOCl). Biochemical journal, 449, 531–542.
  • Haffner, S.M., et al., 1985. Epidemiological correlates of high density lipoprotein subfractions, apolipoproteins A-I, A-II, and D, and lecithin cholesterol acyltransferase. Effects of smoking, alcohol, and adiposity. Arteriosclerosis, 5, 169–177.
  • Halvorsen, B., et al., 2007. Effect of smoking cessation on markers of inflammation and endothelial cell activation among individuals with high risk for cardiovascular disease. Scandinavian journal of clinical and laboratory investigation, 67, 604–611.
  • Hamilton, G. and Rath, B., 2015. Smoking, inflammation and small cell lung cancer: recent developments. Wiener medizinische wochenschrift (1946), 165, 379–386.
  • Hare, J.M., et al., 2002. Exhaled nitric oxide: a marker of pulmonary hemodynamics in heart failure. Journal of the American college of cardiology, 40, 1114–1119.
  • Harman, S.M., et al., 2003. Urinary excretion of three nucleic acid oxidation adducts and isoprostane F(2)alpha measured by liquid chromatography-mass spectrometry in smokers, ex-smokers, and nonsmokers. Free radical biology & medicine, 35, 1301–1309.
  • Hatsukami, D.K., et al., 2005. Effects of cigarette reduction on cardiovascular risk factors and subjective measures. Chest, 128, 2528–2537.
  • Hecht, S.S., 1998. Biochemistry, biology, and carcinogenicity of tobacco-specific N-nitrosamines. Chemical research in toxicology, 11, 559–603.
  • Hecht, S.S., 2008. Progress and challenges in selected areas of tobacco carcinogenesis. Chemical research in toxicology, 21, 160–171.
  • Hecht, S.S., et al., 2015. Evaluation of toxicant and carcinogen metabolites in the urine of e-cigarette users versus cigarette smokers. Nicotine & tobacco research, 17, 704–709.
  • Hecht, S.S., et al., 2013. Tobacco smoke biomarkers and cancer risk among male smokers in the Shanghai cohort study. Cancer letters, 334, 34–38.
  • Hedblad, B., et al., 2006. COHb% as a marker of cardiovascular risk in never smokers: results from a population-based cohort study. Scandinavian journal of public health, 34, 609–615.
  • Hedblad, B., et al., 2005. Heterogeneity of cardiovascular risk among smokers is related to degree of carbon monoxide exposure. Atherosclerosis, 179, 177–183.
  • Hegde, S., et al., 2016. Estimation and correlation of salivary thiocyanate levels in periodontally healthy subjects, smokers, nonsmokers, and gutka-chewers with chronic periodontitis. Indian journal of dental research, 27, 12–14.
  • Hertsgaard, L.A., et al., 2008. Exposure to a tobacco-specific lung carcinogen in adolescent versus adult smokers. Cancer epidemiology, biomarkers & prevention, 17, 3337–3343.
  • Higashimoto, Y., et al., 2008. Systemic inflammation in chronic obstructive pulmonary disease and asthma: similarities and differences. Respirology, 13, 128–133.
  • Higuchi, T., et al., 2016. Current cigarette smoking is a reversible cause of elevated white blood cell count: cross-sectional and longitudinal studies. Preventive medicine reports, 4, 417–422.
  • Hijazi, Z., et al., 2013. Biomarkers in atrial fibrillation: a clinical review. European heart journal, 34, 1475–1480.
  • Hilton, S., et al., 2016. E-cigarettes, a safer alternative for teenagers? A UK focus group study of teenagers' views. BMJ open, 6, e013271.
  • Ho, E., et al., 2013a. Biological markers of oxidative stress: applications to cardiovascular research and practice. Redox biology, 1, 483–491.
  • Ho, G., et al., 2013b. Biomarkers of tobacco smoke exposure and asthma severity in adults. American journal of preventive medicine, 45, 703–709.
  • Hoffmeyer, F., et al., 2009. Leukotriene B4, 8-iso-prostaglandin F2 alpha, and pH in exhaled breath condensate from asymptomatic smokers. Journal of physiology and pharmacology, 60(Suppl 5), 57–60.
  • Hogan, S.L., et al., 2007. Association of cigarette smoking with albuminuria in the United States: the third National Health and Nutrition Examination Survey. Renal failure, 29, 133–142.
  • Hogg, J.C., et al., 2004. The nature of small-airway obstruction in chronic obstructive pulmonary disease. The new England journal of medicine, 350, 2645–2653.
  • Hogg, J.C., Macklem, P.T., and Thurlbeck, W.M., 1968. Site and nature of airway obstruction in chronic obstructive lung disease. The new England journal of medicine, 278, 1355–1360.
  • Hogg, J.C., Mcdonough, J.E., and Suzuki, M., 2013. Small airway obstruction in COPD: new insights based on micro-CT imaging and MRI imaging. Chest, 143, 1436–1443.
  • Hogman, M., et al., 2002. Increased nitric oxide elimination from the airways after smoking cessation. Clinical science (London), 103, 15–19.
  • Hokanson, J.M., et al., 2006. Integrated tobacco cessation counseling in a diabetes self-management training program: a randomized trial of diabetes and reduction of tobacco. The diabetes educator, 32, 562–570.
  • Holdenrieder, S., 2016. Biomarkers along the continuum of care in lung cancer. Scandinavian journal of clinical and laboratory investigation. supplementum, 245, S40–S45.
  • Holt, P.G., 1987. Immune and inflammatory function in cigarette smokers. Thorax, 42, 241–249.
  • Holz, O., et al., 2014. Potential prognostic value of biomarkers in lavage, sputum and serum in a five year clinical follow-up of smokers with and without COPD. BMC pulmonary medicine, 14, 30.
  • Holzer, M., et al., 2013. Aging affects high-density lipoprotein composition and function. Biochimica et biophysica acta, 1831, 1442–1448.
  • Hovingh, G.K., Bochem, A.E., and Kastelein, J.J., 2010. Apolipoprotein A-I mimetic peptides. Current opinion in lipidology, 21, 481–486.
  • Hovingh, G.K., Rader, D.J., and Hegele, R.A., 2015. HDL re-examined. Current opinion in lipidology, 26, 127–132.
  • Hoy, A., et al., 2002. Growing significance of myeloperoxidase in non-infectious diseases. Clinical chemistry and laboratory medicine, 40, 2–8.
  • Hozawa, A., et al., 2006. Does the impact of smoking on coronary heart disease differ by low-density lipoprotein cholesterol level?: the Atherosclerosis Risk in Communities (ARIC) Study. Circulation journal, 70, 1105–1110.
  • Hu, G., et al., 2010. Risk of COPD from exposure to biomass smoke: a metaanalysis. Chest, 138, 20–31.
  • Hu, R., et al., 2005. Smoking, homocysteine and degree of arteriolar retinopathy. Atherosclerosis, 183, 95–100.
  • Hwang, S.J., et al., 1997. Circulating adhesion molecules VCAM-1, ICAM-1, and E-selectin in carotid atherosclerosis and incident coronary heart disease cases: the Atherosclerosis Risk in Communities (ARIC) study. Circulation, 96, 4219–4225.
  • Ikeda, Y., et al., 1997. Effect of smoking on the prevalence of albuminuria in Japanese men with non-insulin-dependent diabetes mellitus. Diabetes research and clinical practice, 36, 57–61.
  • Ikitimur, B. and Karadag, B., 2010. Role of myeloperoxidase in cardiology. Future cardiology, 6, 693–702.
  • Ikonomidis, I., et al., 2005. Cigarette smoking is associated with increased circulating proinflammatory and procoagulant markers in patients with chronic coronary artery disease: effects of aspirin treatment. American heart journal, 149, 832–839.
  • Il'yasova, D., et al., 2012. Racial differences in urinary F2-isoprostane levels and the cross-sectional association with BMI. Obesity (Silver Spring), 20, 2147–2150.
  • Imaizumi, S., et al., 2011. Dysfunctional high-density lipoprotein and the potential of apolipoprotein A-1 mimetic peptides to normalize the composition and function of lipoproteins. Circulation journal, 75, 1533–1538.
  • Imaizumi, T., et al., 1990. Activity of platelet-activating factor (PAF) acetylhydrolase in plasma from healthy habitual cigarette smokers. Heart and vessels, 5, 81–86.
  • Imes, C.C. and Austin, M.A., 2013. Low-density lipoprotein cholesterol, apolipoprotein B, and risk of coronary heart disease: from familial hyperlipidemia to genomics. Biological research for nursing, 15, 292–308.
  • Inonu, H., et al., 2012. Oxidative stress levels in exhaled breath condensate associated with COPD and smoking. Respiratory care, 57, 413–419.
  • Ishikawa, S., et al., 1997. Factor VII and fibrinogen levels examined by age, sex, and other atherosclerotic risk factors in a Japanese population. The Jichi Medical School Cohort Study. Thrombosis and haemostasis, 77, 890–893.
  • Issaq, H.J., Waybright, T.J., and Veenstra, T.D., 2011. Cancer biomarker discovery: opportunities and pitfalls in analytical methods. Electrophoresis, 32, 967–975.
  • Iwaoka, M., et al., 2014. Twelve weeks of smoking cessation therapy with varenicline increases the serum levels of apolipoprotein A-I only in the success group. Journal of cardiology, 64, 318–323.
  • Jain, R.B., 2016. Trends and variability in the levels of urinary thiocyanate, perchlorate, and nitrate by age, gender, race/ethnicity, smoking status, and exposure to environmental tobacco smoke over 2005-2012. Science of the total environment, 557–558, 221–230.
  • Janssen, L.J., 2001. Isoprostanes: an overview and putative roles in pulmonary pathophysiology. American journal of physiology. Lung cellular and molecular physiology, 280, L1067–L1082.
  • Jha, P., et al., 2013. 21st-century hazards of smoking and benefits of cessation in the United States. The new England journal of medicine, 368, 341–350.
  • Ji, J., et al., 2014. Compartment differences of inflammatory activity in chronic obstructive pulmonary disease. Respiratory research, 15, 104.
  • Jiang, Y., Wang, X., and Hu, D., 2017. Mitochondrial alterations during oxidative stress in chronic obstructive pulmonary disease. International journal of chronic obstructive pulmonary disease, 12, 1153–1162.
  • Johns, D.P., Walters, J.A., and Walters, E.H., 2014. Diagnosis and early detection of COPD using spirometry. Journal of thoracic disease, 6, 1557–1569.
  • Joo, M.J., et al., 2013. Use of spirometry in the diagnosis of COPD: a qualitative study in primary care. COPD, 10, 444–449.
  • Kamat, G.V., et al., 2010. A cross-sectional study to detect the prevalence of hyperhomocysteinemia in cases of deep vein thrombosis. The Indian journal of surgery, 72, 323–326.
  • Kannel, W.B., et al., 1987. Fibrinogen and risk of cardiovascular disease. The Framingham Study. JAMA, 258, 1183–1186.
  • Kanner, R.E., et al., 1999. Effects of randomized assignment to a smoking cessation intervention and changes in smoking habits on respiratory symptoms in smokers with early chronic obstructive pulmonary disease: the Lung Health Study. The American journal of medicine, 106, 410–416.
  • Kappelmayer, J., et al., 2004. The emerging value of P-selectin as a disease marker. Clinical chemistry and laboratory medicine, 42, 475–486.
  • Kar, D., et al., 2016. Relationship of cardiometabolic parameters in non-smokers, current smokers, and quitters in diabetes: a systematic review and meta-analysis. Cardiovascular diabetology, 15, 158.
  • Kawada, T., 2015. Relationships between the smoking status and plasma fibrinogen, white blood cell count and serum C-reactive protein in Japanese workers. Diabetes & Metabolic Syndrome, 9, 180–182.
  • Kaysen, G.A. and Levin, N.W., 2002. Why measure serum albumin levels? Journal of renal nutrition, 12, 148–150.
  • Keatings, V.M., et al., 1996. Differences in interleukin-8 and tumor necrosis factor-alpha in induced sputum from patients with chronic obstructive pulmonary disease or asthma. American journal of respiratory and critical care medicine, 153, 530–534.
  • Khan, N.A., et al., 2016. The effect of folic acid supplementation on hyperhomocysteinemia and pulmonary function parameters in chronic obstructive pulmonary disease: a pilot study. Journal of clinical and diagnostic research, 10, OC17–OC21.
  • Kharitonov, S.A. and Barnes, P.J., 2002. Biomarkers of some pulmonary diseases in exhaled breath. Biomarkers: biochemical indicators of exposure, response, and susceptibility to chemicals, 7, 1–32.
  • Kharitonov, S.A. and Barnes, P.J., 2006. Exhaled biomarkers. Chest, 130, 1541–1546.
  • Kharitonov, S.A., et al., 1995. Acute and chronic effects of cigarette smoking on exhaled nitric oxide. American journal of respiratory and critical care medicine, 152, 609–612.
  • Khera, A., et al., 2005. Race and gender differences in C-reactive protein levels. Journal of the American college of cardiology, 46, 464–469.
  • Kim, H.O., et al., 2011. Serum cytokine profiles in healthy young and elderly population assessed using multiplexed bead-based immunoassays. Journal of translational medicine, 9, 113.
  • King, C.C., et al., 2017. Longitudinal impact of smoking and smoking cessation on inflammatory markers of cardiovascular disease risk. Arteriosclerosis, thrombosis, and vascular biology, 37, 374–379.
  • Kleiner, G., et al., 2013. Cytokine levels in the serum of healthy subjects. Mediators of inflammation, 2013, 434010
  • Kleniewska, A., et al., 2016. Comparison of biomarkers in serum and induced sputum of patients with occupational asthma and chronic obstructive pulmonary disease. Journal of occupational health, 58, 333–339.
  • Koenig, W., 2003. Fibrin(ogen) in cardiovascular disease: an update. Thrombosis and haemostasis, 89, 601–609.
  • Koeth, R.A., Haselden, V., and Tang, W.H., 2013. Myeloperoxidase in cardiovascular disease. Advances in clinical chemistry, 62, 1–32.
  • Kontush, A. and Chapman, M.J., 2006. Antiatherogenic small, dense HDL-guardian angel of the arterial wall? Nature clinical practice. Cardiovascular medicine, 3, 144–153.
  • Korhonen, T., et al., 2011. Smoking cessation program with exercise improves cardiovascular disease biomarkers in sedentary women. Journal of womens health (Larchmt), 20, 1051–1064.
  • Kotani, K., et al., 2010. Influence of smoking habits on the GA/HbA1c ratio in patients with type 1 diabetes mellitus. Medical principles and practice: International journal of the Kuwait University, health science centre, 19, 415–417.
  • Kryfti, M., et al., 2015. Effects of smoking cessation on serum leptin and adiponectin levels. Tobacco induced diseases, 13, 30
  • Kubala, L., et al., 2008. Plasma levels of myeloperoxidase are not elevated in patients with stable coronary artery disease. Clinica chimica acta; International journal of clinical chemistry, 394, 59–62.
  • Kuban, P. and Foret, F., 2013. Exhaled breath condensate: determination of non-volatile compounds and their potential for clinical diagnosis and monitoring. A review. Analytica chimica acta, 805, 1–18.
  • Kucharska-Newton, A.M., et al., 2008. HDL-cholesterol and the incidence of lung cancer in the Atherosclerosis Risk in Communities (ARIC) study. Lung cancer (Amsterdam, Netherlands), 61, 292–300.
  • Kumari, M., Marmot, M., and Brunner, E., 2000. Social determinants of von willebrand factor: the Whitehall II study. Arteriosclerosis, thrombosis, and vascular biology, 20, 1842–1847.
  • Kverneland, A.H., et al., 2016. Age and gender leucocytes variances and references values generated using the standardized ONE-Study protocol. Cytometry part A, 89, 543–564.
  • Lammertyn, L., et al., 2015. Ethnic-specific relationships between haemostatic and oxidative stress markers in black and white South Africans: the SABPA study. Clinical and experimental hypertension, 37, 511–517.
  • Lange, P., et al., 2003. Chronic bronchitis in an elderly population. Age and ageing, 32, 636–642.
  • Lappas, A.S., et al., 2016. Immediate effects of cigar smoking on respiratory mechanics and exhaled biomarkers; differences between young smokers with mild asthma and otherwise healthy young smokers. Tobacco induced diseases, 14, 29.
  • Lathe, R., Sapronova, A., and Kotelevtsev, Y., 2014. Atherosclerosis and Alzheimer–diseases with a common cause? Inflammation, oxysterols, vasculature. BMC geriatrics, 14, 36.
  • Lavange, L., et al., 2017. Spirometry reference equations from the HCHS/SOL (Hispanic Community Health Study/Study of Latinos). American journal of respiratory and critical care medicine, 196, 993–1003.
  • Lee, D.S., et al., 2008. Clinical and genetic correlates of soluble P-selectin in the community. Journal of thrombosis and haemostasis, 6, 20–31.
  • Lee, J.S., et al., 2017. Triglyceride and HDL-C dyslipidemia and risks of coronary heart disease and ischemic stroke by glycemic dysregulation status: the strong heart study. Diabetes care, 40, 529–537.
  • Lee, P.N., et al., 2014. The effect of quitting smoking on white blood cell count: a review based on within-subject changes. Sutton, Surrey: P. N. Lees Statistics and Computing.
  • Lee, P.N. and Forey, B.A., 2013. Indirectly estimated absolute lung cancer mortality rates by smoking status and histological type based on a systematic review. BMC cancer, 13, 189.
  • Lee, S.J., et al., 1993. Serum levels of tumor necrosis factor-alpha and interleukin-6 in ocular cicatricial pemphigoid. Investigative ophthalmology & visual science, 34, 3522–3525.
  • Lee, W.J., et al., 2016. Soluble ICAM-1, independent of IL-6, is associated with prevalent frailty in community-dwelling elderly Taiwanese people. PLoS one, 11, e0157877.
  • Leman, L.J., Maryanoff, B.E., and Ghadiri, M.R., 2014. Molecules that mimic apolipoprotein A-I: potential agents for treating atherosclerosis. Journal of medicinal chemistry, 57, 2169–2196.
  • Lensmar, C., et al., 1998. Smoking alters the phenotype of macrophages in induced sputum. Respiratory medicine, 92, 415–420.
  • Lenting, P.J., et al., 2012. von Willebrand factor: the old, the new and the unknown. Journal of thrombosis and haemostasis, 10, 2428–2437.
  • Lenting, P.J., Denis, C.V., and Wohner, N., 2013. Von Willebrand factor and thrombosis: risk factor, actor and pharmacological target. Current vascular pharmacology, 11, 448–456.
  • Leone, A., 2015. Toxics of tobacco smoke and cardiovascular system: from functional to cellular damage. Current pharmaceutical design, 21, 4370–4379.
  • Leoni, V., et al., 2013. Diagnostic power of 24S-hydroxycholesterol in cerebrospinal fluid: candidate marker of brain health. Journal of Alzheimer’s diseases, 36, 739–747.
  • Leung, J.M. and Sin, D.D., 2013. Biomarkers in airway diseases. Canadian respiratory journal, 20, 180–182.
  • Levitzky, Y.S., et al., 2008. Relation of smoking status to a panel of inflammatory markers: the Framingham offspring. Atherosclerosis, 201, 217–224.
  • Li, D., et al., 2017. Lipoprotein-associated phospholipase A2 in coronary heart disease: review and meta-analysis. Clinica chimica acta, 465, 22–29.
  • Life Sciences Research Office., 2007. Biological effects assessment in the evaluation of potential reduced-risk tobacco products. Bethesda: Life Sciences Research Office, Inc.
  • Lim, E., Miyamura, J., and Chen, J.J., 2015. Racial/ethnic-specific reference intervals for common laboratory tests: a comparison among Asians, blacks, hispanics, and white. Hawaii journal of medicine & public health, 74, 302–310.
  • Lim, E.M., et al., 2010. Race-specific WBC and neutrophil count reference intervals. International journal of laboratory hematology, 32, 590–597.
  • Lind, P., et al., 2004. Risk of myocardial infarction and stroke in smokers is related to plasma levels of inflammation-sensitive proteins. Arteriosclerosis, thrombosis, and vascular biology, 24, 577–582.
  • Lip, G.Y. and Blann, A.D., 1995. von Willebrand factor and its relevance to cardiovascular disorders. British heart journal, 74, 580–583.
  • Liu, J., et al., 2011. Relationship between biomarkers of cigarette smoke exposure and biomarkers of inflammation, oxidative stress, and platelet activation in adult cigarette smokers. Cancer epidemiology, biomarkers & prevention, 20, 1760–1769.
  • Lobbes, M.B., et al., 2010. Leukocyte counts, myeloperoxidase, and pregnancy-associated plasma protein a as biomarkers for cardiovascular disease: towards a multi-biomarker approach. International journal of vascular medicine, 2010, 726207.
  • Lock-Johansson, S., Vestbo, J., and Sorensen, G.L., 2014. Surfactant protein D, Club cell protein 16, Pulmonary and activation-regulated chemokine, C-reactive protein, and Fibrinogen biomarker variation in chronic obstructive lung disease. Respiratory research, 15, 147.
  • Lopez-Campos, J.L., et al., 2012. Increased levels of soluble ICAM-1 in chronic obstructive pulmonary disease and resistant smokers are related to active smoking. Biomarkers in medicine, 6, 805–811.
  • Loscalzo, J., 2001. Nitric oxide insufficiency, platelet activation, and arterial thrombosis. Circulation research, 88, 756–762.
  • Lowe, F.J., Gregg, E.O., and Mcewan, M., 2009. Evaluation of biomarkers of exposure and potential harm in smokers, former smokers and never-smokers. Clinical chemistry and laboratory medicine, 47, 311–320.
  • Lowe, G.D. and Pepys, M.B., 2006. C-reactive protein and cardiovascular disease: weighing the evidence. Current atherosclerosis reports, 8, 421–428.
  • Lowe, G.D., et al., 2001. C-reactive protein, fibrin D-dimer, and incident ischemic heart disease in the Speedwell study: are inflammation and fibrin turnover linked in pathogenesis?. Arteriosclerosis, thrombosis, and vascular biology, 21, 603–610.
  • Lp-PLA(2) Studies Collaboration, et al., 2010. Lipoprotein-associated phospholipase A(2) and risk of coronary disease, stroke, and mortality: collaborative analysis of 32 prospective studies. Lancet, 375, 1536–1544.
  • Lu, J., et al., 2012. Profiling plasma peptides for the identification of potential ageing biomarkers in Chinese Han adults. PLoS one, 7, e39726.
  • Lüdicke, F., et al., 2016. Evaluation of biomarkers of exposure in smokers switching to a carbon-heated tobacco product: a controlled, randomized, open-label 5-day exposure study. Nicotine & tobacco research, 18, 1606–1613.
  • Lüdicke, F., et al., 2017. Effects of switching to the menthol tobacco heating system 2.2, Smoking abstinence, or continued cigarette smoking on clinically relevant risk markers: a randomized, controlled, open-label, multicenter study in sequential confinement and ambulatory settings (Part 2). Nicotine & tobacco research. doi: 10.1093/ntr/ntx028. [Epub ahead of print]
  • Ludviksdottir, D., et al., 1999. Effects of nicotine nasal spray on atherogenic and thrombogenic factors during smoking cessation. Journal of internal medicine, 246, 61–66.
  • Lupia, E., et al., 2010. Thrombopoietin contributes to enhanced platelet activation in cigarette smokers. Atherosclerosis, 210, 314–319.
  • Luthje, S., et al., 2013. Plasma membrane electron pathways and oxidative stress. Antioxidants & redox signaling, 18, 2163–2183.
  • Lutsey, P.L., et al., 2006. Plasma hemostatic factors and endothelial markers in four racial/ethnic groups: the MESA study. Journal of thrombosis and haemostasis, 4, 2629–2635.
  • Macnee, W., et al., 1989. The effect of cigarette smoking on neutrophil kinetics in human lungs. The new England journal of medicine, 321, 924–928.
  • Madjid, M., et al., 2004. Leukocyte count and coronary heart disease: implications for risk assessment. Journal of the American college of cardiology, 44, 1945–1956.
  • Maestrelli, P., et al., 1996. Integrin upregulation on sputum neutrophils in smokers with chronic airway obstruction. American journal of respiratory and critical care medicine, 154, 1296–1300.
  • Malik, I., et al., 2001. Soluble adhesion molecules and prediction of coronary heart disease: a prospective study and meta-analysis. Lancet, 358, 971–976.
  • Malinovschi, A., et al., 2012. The value of exhaled nitric oxide to identify asthma in smoking patients with asthma-like symptoms. Respiratory medicine, 106, 794–801.
  • Mandraffino, G., et al., 2017. Biglycan expression, earlier vascular damage and pro-atherogenic profile improvement after smoke cessation in young people. Atherosclerosis, 257, 109–115.
  • Marano, K.M., et al., 2015. Study of cardiovascular disease biomarkers among tobacco consumers. Part 3: evaluation and comparison with the US National Health and Nutrition Examination Survey. Inhalation toxicology, 27, 167–173.
  • Marti-Carvajal, A.J., et al., 2013. Homocysteine-lowering interventions for preventing cardiovascular events. The Cochrane database of systematic reviews, 1, CD006612.
  • Martin, F., et al., 2017. Quantifying the risk-reduction potential of new modified risk tobacco products. Regulatory toxicology and pharmacology. doi: 10.1016/j.yrtph.2017.12.011. [Epub ahead of print].
  • Martin Leroy, C., et al., 2012. Reduced exposure evaluation of an Electrically Heated Cigarette Smoking System. Part 7: a one-month, randomized, ambulatory, controlled clinical study in Poland. Regulatory toxicology and pharmacology, 64, S74–S84.
  • Martini, F., et al., 2005. Plasma von Willebrand factor antigen levels in non-small cell lung cancer patients. Anticancer research, 25, 403–407.
  • Masarei, J.R., et al., 1991. Effect of smoking cessation on serum apolipoprotein A-I and A-II concentrations. Pathology, 23, 98–102.
  • Maskey-Warzechowska, M., et al., 2017. Acute response to cigarette smoking assessed in exhaled breath condensate in patients with chronic obstructive pulmonary disease and healthy smokers. Advances in experimental medicine and biology, 944, 73–80.
  • Maurya, P.K., et al., 2016. The role of oxidative and nitrosative stress in accelerated aging and major depressive disorder. Progress in neuropsychopharmacology & biological psychiatry, 65, 134–144.
  • Mcadam, B.F., et al., 2005. Contribution of cyclooxygenase-2 to elevated biosynthesis of thromboxane A2 and prostacyclin in cigarette smokers. Circulation, 112, 1024–1029.
  • Mceligot, A.J., et al., 2006. Responsiveness of homocysteine concentrations to food and supplemental folate intakes in smokers and never-smokers enrolled in a diet intervention trial. Nicotine & tobacco research, 8, 57–66.
  • Mcgrath, C.R., Hitchcock, D.C., and Van Assendelft, O.W., 1982. Total white blood cell counts for persons ages 1-74 years with differential leukocyte counts for adults ages 25-74 years: United States, 1971-75. Vital and health statistics, 11, 1–36.
  • Mcguinness, A.J. and Sapey, E., 2017. Oxidative stress in COPD: sources, markers, and potential mechanisms. Journal of clinical medicine, 6, E21.
  • Mclain, J.H., Alsterda, A.J., and Arora, R.R., 2016. Cholesteryl ester transfer protein inhibitors: trials and tribulations. Journal of cardiovascular pharmacology and therapy. pii: 1074248416662349. [Epub ahead of print]
  • Mehrotra, N., et al., 2010. Predictors of mortality in elderly subjects with obstructive airway disease: the PILE score. Annals of epidemiology, 20, 223–232.
  • Merten, M. and Thiagarajan, P., 2004. P-selectin in arterial thrombosis. Zeitschrift fur kardiologie, 93, 855–863.
  • Messner, B. and Bernhard, D., 2014. Smoking and cardiovascular disease: mechanisms of endothelial dysfunction and early atherogenesis. Arteriosclerosis, thrombosis, and vascular biology, 34, 509–515.
  • Metcalf, P.A., et al., 1993. Albuminuria in people at least 40 years old: effect of alcohol consumption, regular exercise, and cigarette smoking. Clinical chemistry, 39, 1793–1797.
  • Meuwese, M.C., et al., 2007. Serum myeloperoxidase levels are associated with the future risk of coronary artery disease in apparently healthy individuals: the EPIC-Norfolk Prospective Population Study. Journal of the American college of cardiology, 50, 159–165.
  • Miao Jonasson, J., et al., 2012. HbA1C and cancer risk in patients with type 2 diabetes: a nationwide population-based prospective cohort study in Sweden. PLoS one, 7, e38784.
  • Miles, E.A., et al., 2001. Influence of age and dietary fish oil on plasma soluble adhesion molecule concentrations. Clinical science, 100, 91–100.
  • Miller, M.A. and Cappuccio, F.P., 2007. Ethnicity and inflammatory pathways: implications for vascular disease, vascular risk and therapeutic intervention. Current medicinal chemistry, 14, 1409–1425.
  • Mirabelli, M.C., et al., 2016. Lung function decline over 25 years of follow-up among black and white adults in the ARIC study cohort. Respiratory medicine, 113, 57–64.
  • Mol, M.J., et al., 1997. Plasma levels of lipid and cholesterol oxidation products and cytokines in diabetes mellitus and cigarette smoking: effects of vitamin E treatment. Atherosclerosis, 129, 169–176.
  • Molassiotis, A., et al., 2010a. Interventions for cough in cancer. The Cochrane database of systematic reviews, 5, CD007881.
  • Molassiotis, A., et al., 2010b. Pharmacological and non-pharmacological interventions for cough in adults with respiratory and non-respiratory diseases: a systematic review of the literature. Respiratory medicine, 104, 934–944.
  • Mons, U., et al., 2016. Effect of smoking reduction and cessation on the plasma levels of the oxidative stress biomarker glutathione – post-hoc analysis of data from a smoking cessation trial. Free radical biology & medicine, 91, 172–177.
  • Moore, P.K., 1982. Prostaglandins, prostacyclin and thromboxanes. Biochemical education, 10, 82–87.
  • Moreira, M.A., et al., 2013. Pulmonary changes on HRCT scans in nonsmoking females with COPD due to wood smoke exposure. Jornal Brasileiro de pneumologia, 39, 155–163.
  • Morgan, P.E., et al., 2011. High plasma thiocyanate levels in smokers are a key determinant of thiol oxidation induced by myeloperoxidase. Free radical biology & medicine, 51, 1815–1822.
  • Morita, H., et al., 2005. Only two-week smoking cessation improves platelet aggregability and intraplatelet redox imbalance of long-term smokers. Journal of the American college of cardiology, 45, 589–594.
  • Morrow, J.D., 2005. Quantification of isoprostanes as indices of oxidant stress and the risk of atherosclerosis in humans. Arteriosclerosis, thrombosis, and vascular biology, 25, 279–286.
  • Morrow, J.D., et al., 1995. Increase in circulating products of lipid peroxidation (F2-isoprostanes) in smokers. Smoking as a cause of oxidative damage. New England journal of medicine, 332, 1198–1203., 2nd,
  • Morrow, J.D., et al., 1990. A series of prostaglandin F2-like compounds are produced in vivo in humans by a non-cyclooxygenase, free radical-catalyzed mechanism. Proceedings of the national academy of sciences of the United States of America, 87, 9383–9387.
  • Muenchhoff, J., et al., 2017. Plasma apolipoproteins and physical and cognitive health in very old individuals. Neurobiology of Aging, 55, 49–60.
  • Nadif, R., et al., 2010. Passive and active smoking and exhaled nitric oxide levels according to asthma and atopy in adults. Annals of allergy, asthma, & immunology, 104, 385–393.
  • Nakahata, N., 2008. Thromboxane A2: physiology/pathophysiology, cellular signal transduction and pharmacology. Pharmacology &Amp; therapeutics, 118, 18–35.
  • Nakos, G., et al., 2005. Phospholipases A2 and platelet-activating-factor acetylhydrolase in patients with acute respiratory distress syndrome. Critical care medicine, 33, 772–779.
  • Nakos, G., Tsangaris, I., and Lekka, M.E., 2004. Platelet-activating factor acetylhydrolase and acute respiratory distress syndrome: unanswered questions. Critical care medicine, 32, 1434–1435.
  • Nalls, M.A., et al., 2008. Admixture mapping of white cell count: genetic locus responsible for lower white blood cell count in the Health ABC and Jackson Heart studies. The American journal of human genetics, 82, 81–87.
  • Nash, M.C., et al., 1996. Normal levels of soluble E-selectin, soluble intercellular adhesion molecule-1 (sICAM-1), and soluble vascular cell adhesion molecule-1 (sVCAM-1) decrease with age. Clinical and experimental immunology, 103, 167–170.
  • Neubauer, H., et al., 2009. Upregulation of platelet CD40, CD40 ligand (CD40L) and P-Selectin expression in cigarette smokers: a flow cytometry study. Blood coagulation & fibrinolysis, 20, 694–698.
  • Niccoli, G., Dato, I., and Crea, F., 2010. Myeloperoxidase may help to differentiate coronary plaque erosion from plaque rupture in patients with acute coronary syndromes. Trends in cardiovascular medicine, 20, 276–281.
  • Nicholls, S.J. and Hazen, S.L., 2005. Myeloperoxidase and cardiovascular disease. Arteriosclerosis, thrombosis, and vascular biology, 25, 1102–1111.
  • Nilsson, G., et al., 2009. Ten-year survival in 75-year-old men and women: predictive ability of total cholesterol, HDL-C, and LDL-C. Current gerontology and geriatrics research, 2009, 158425.
  • Nilsson, P.M., et al., 1995. Increased level of hemoglobin A1c, but not impaired insulin sensitivity, found in hypertensive and normotensive smokers. Metabolism, 44, 557–561.
  • Nordskog, B.K., et al., 2015. Study of cardiovascular disease biomarkers among tobacco consumers, part 2: biomarkers of biological effect. Inhalation toxicology, 27, 157–166.
  • Nowak, J., et al., 1987. Biochemical evidence of a chronic abnormality in platelet and vascular function in healthy individuals who smoke cigarettes. Circulation, 76 (1), 6–14.
  • Nussbaum, C., et al., 2013. Myeloperoxidase: a leukocyte-derived protagonist of inflammation and cardiovascular disease. Antioxidants & redox signaling, 18, 692–713.
  • Nutt, D.J., et al., 2014. Estimating the harms of nicotine-containing products using the MCDA approach. European addiction research, 20, 218–225.
  • Nyberg, M., et al., 2014. Biomarkers of vascular function in premenopausal and recent postmenopausal women of similar age: effect of exercise training. American journal of physiology. Regulatory, integrative and comparative physiology, 306, R510–R517.
  • Nygard, O., et al., 1997. Plasma homocysteine levels and mortality in patients with coronary artery disease. The new England journal of medicine, 337, 230–236.
  • Nygard, O., et al., 1998. Major lifestyle determinants of plasma total homocysteine distribution: the Hordaland Homocysteine Study. The American journal of clinical nutrition, 67, 263–270.
  • Nygard, O., et al., 1995. Total plasma homocysteine and cardiovascular risk profile. The Hordaland Homocysteine Study. JAMA, 274, 1526–1533.
  • O'callaghan, P., et al., 2002. Smoking and plasma homocysteine. European heart journal, 23, 1580–1586.
  • O'callaghan, P.A., et al., 2005. New and old cardiovascular risk factors: C-reactive protein, homocysteine, cysteine and von Willebrand factor increase risk, especially in smokers. European journal of cardiovascular prevention & rehabilitation, 12, 542–547.
  • Obersby, D., et al., 2013. Plasma total homocysteine status of vegetarians compared with omnivores: a systematic review and meta-analysis. British journal of nutrition, 109, 785–794.
  • Ohkuma, T., et al., 2015. Dose- and time-dependent association of smoking and its cessation with glycemic control and insulin resistance in male patients with type 2 diabetes mellitus: the Fukuoka Diabetes Registry. PLoS one, 10, e0122023.
  • Olkkonen, V.M., 2012. Macrophage oxysterols and their binding proteins: roles in atherosclerosis. Current opinion in lipidology, 23, 462–470.
  • Olsen, B.N., et al., 2012. Side-chain oxysterols: from cells to membranes to molecules. Biochimica et biophysica acta, 1818, 330–336.
  • Orth, S.R., 2002. Cigarette smoking: an important renal risk factor – far beyond carcinogenesis. Tobacco induced diseases, 1, 137–155.
  • Osterud, B., et al., 1999. Haemostatic parameters related to lipids and adhesion molecules. Blood coagulation & fibrinolysis, 10, 465–470.
  • Otokozawa, S., et al., 2010. Direct assessment of plasma low density lipoprotein and high density lipoprotein cholesterol levels and coronary heart disease: results from the Framingham Offspring Study. Atherosclerosis, 213, 251–255.
  • Paalani, M., et al., 2011. Determinants of inflammatory markers in a bi-ethnic population. Ethnicity &Amp; disease, 21, 142–149.
  • Pacilli, A., et al., 2013. Role of relationship between HbA1c, fibrinogen and HDL-cholesterol on cardiovascular disease in patients with type 2 diabetes mellitus. Atherosclerosis, 228, 247–248.
  • Pallasaho, P., et al., 2014. Combined effect of smoking and occupational exposure to dusts, gases or fumes on the incidence of COPD. COPD, 11, 88–95.
  • Paone, G., et al., 2011. Analysis of sputum markers in the evaluation of lung inflammation and functional impairment in symptomatic smokers and COPD patients. Disease markers, 31, 91–100.
  • Paramo, J.A., et al., 2004. Validation of plasma fibrinogen as a marker of carotid atherosclerosis in subjects free of clinical cardiovascular disease. Haematologica, 89, 1226–1231.
  • Park, H.Y., et al., 2013. The relation of serum myeloperoxidase to disease progression and mortality in patients with chronic obstructive pulmonary disease (COPD). PLoS one, 8, e61315.
  • Paulinska, P., Spiel, A., and Jilma, B., 2009. Role of von Willebrand factor in vascular disease. Hamostaseologie, 29, 32–38.
  • Payne, A.B., et al., 2014. High factor VIII, von Willebrand factor, and fibrinogen levels and risk of venous thromboembolism in blacks and whites. Ethnicity & disease, 24, 169–174.
  • Pearson, T.A., et al., 2003. Markers of inflammation and cardiovascular disease: application to clinical and public health practice: a statement for healthcare professionals from the Centers for Disease Control and Prevention and the American Heart Association. Circulation, 107, 499–511.
  • Pena-Pereira, F., Lavilla, I., and Bendicho, C., 2016. Paper-based analytical device for instrumental-free detection of thiocyanate in saliva as a biomarker of tobacco smoke exposure. Talanta, 147, 390–396.
  • Pepys, M.B., 2005. CRP or not CRP? That is the question. Arteriosclerosis, thrombosis, and vascular biology, 25, 1091–1094.
  • Persson, M., et al., 2007. The epidemiology of Lp-PLA(2): distribution and correlation with cardiovascular risk factors in a population-based cohort. Atherosclerosis, 190, 388–396.
  • Petrescu, F., Voican, S.C., and Silosi, I., 2010. Tumor necrosis factor-alpha serum levels in healthy smokers and nonsmokers. International journal of chronic obstructive pulmonary disease, 5, 217–222.
  • Pezzuto, A., et al., 2013. Short-term effectiveness of smoking-cessation treatment on respiratory function and CEA level. Journal of comparative effectiveness research, 2, 335–343.
  • Pignatti, P., et al., 2011. Age-related increase of airway neutrophils in older healthy nonsmoking subjects. Rejuvenation research, 14, 365–370.
  • Pinto-Sietsma, S.J., et al., 2000. Smoking is related to albuminuria and abnormal renal function in nondiabetic persons. Annals of internal medicine, 133, 585–591.
  • Polley, L., et al., 2008. Impact of cough across different chronic respiratory diseases: comparison of two cough-specific health-related quality of life questionnaires. Chest, 134, 295–302.
  • Poste, G., 2011. Bring on the biomarkers. Nature, 469, 156–157.
  • Pownall, H.J. and Gotto, A.M., Jr., 2016. New insights into the high-density lipoprotein dilemma. Trends in endocrinology and metabolism, 27, 44–53.
  • Prasad, G.L., et al., 2016. A cross-sectional study of biomarkers of exposure and effect in smokers and moist snuff consumers. Clinical chemistry and laboratory medicine, 54, 633–642.
  • Putcha, N., et al., 2014. Chronic productive cough is associated with death in smokers with early COPD. COPD, 11, 451–458.
  • Quanjer, P.H., et al., 2012a. Age- and height-based prediction bias in spirometry reference equations. The European respiratory journal, 40, 190–197.
  • Quanjer, P.H., et al., 2012b. Multi-ethnic reference values for spirometry for the 3-95-yr age range: the global lung function 2012 equations. The European respiratory journal, 40, 1324–1343.
  • Quanjer, P.H., et al., 2014. Measurement of FEF25-75% and FEF75% does not contribute to clinical decision making. The European respiratory journal, 43, 1051–1058.
  • Racherla, S. and Arora, R., 2012. Utility of Lp-PLA2 in lipid-lowering therapy. American journal of therapeutics, 19, 115–120.
  • Ramirez-Venegas, A., et al., 2014. FEV1 decline in patients with chronic obstructive pulmonary disease associated with biomass exposure. American journal of respiratory and critical care medicine, 190, 996–1002.
  • Rangemark, C., et al., 1992. Tobacco use and urinary excretion of thromboxane A2 and prostacyclin metabolites in women stratified by age. Circulation, 86, 1495–1500.
  • Rangemark, C., Ciabattoni, G., and Wennmalm, A., 1993. Excretion of thromboxane metabolites in healthy women after cessation of smoking. Arteriosclerosis and thrombosis, 13, 777–782.
  • Rangemark, C. and Wennmalm, A., 1991. Cigarette smoking and urinary excretion of markers for platelet/vessel wall interaction in healthy women. Clinical science (London, England: 1979), 81, 11–15.
  • Rangemark, C. and Wennmalm, A., 1996. Smoke-derived nitric oxide and vascular prostacyclin are unable to counteract the platelet effect of increased thromboxane formation in healthy female smokers. Clinical physiology, 16, 301–315.
  • Refsum, H., et al., 2006. The Hordaland Homocysteine Study: a community-based study of homocysteine, its determinants, and associations with disease. Journal of nutrition, 136, 1731S–1740S.
  • Reilly, M., et al., 1996. Modulation of oxidant stress in vivo in chronic cigarette smokers. Circulation, 94, 19–25.
  • Richter, E., et al., 2009. Biotransformation of 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone in lung tissue from mouse, rat, hamster, and man. Chemical research in toxicology, 22, 1008–1017.
  • Ridker, P.M., Buring, J.E., and Rifai, N., 2001. Soluble P-selectin and the risk of future cardiovascular events. Circulation, 103, 491–495.
  • Ridker, P.M., et al., 1998. Plasma concentration of soluble intercellular adhesion molecule 1 and risks of future myocardial infarction in apparently healthy men. Lancet, 351, 88–92.
  • Ridker, P.M., et al., 2000. Elevation of tumor necrosis factor-alpha and increased risk of recurrent coronary events after myocardial infarction. Circulation, 101, 2149–2153.
  • Riise, G.C., et al., 1994. Circulating cell adhesion molecules in bronchial lavage and serum in COPD patients with chronic bronchitis. The European respiratory journal, 7, 1673–1677.
  • Robbins, R.A., et al., 1997. Smoking cessation is associated with an increase in exhaled nitric oxide. Chest, 112, 313–318.
  • Roethig, H.J., et al., 2008. A 12-month, randomized, controlled study to evaluate exposure and cardiovascular risk factors in adult smokers switching from conventional cigarettes to a second-generation electrically heated cigarette smoking system. The journal of clinical pharmacology, 48, 580–591.
  • Roethig, H.J., et al., 2010. Short term effects of reduced exposure to cigarette smoke on white blood cells, platelets and red blood cells in adult cigarette smokers. Regulatory toxicology and pharmacology, 57, 333–337.
  • Roethig, H.J., et al., 2007. Short-term clinical exposure evaluation of a second-generation electrically heated cigarette smoking system. The journal of clinical pharmacology, 47, 518–530.
  • Rosenberg, S.R. and Kalhan, R., 2012. Biomarkers in chronic obstructive pulmonary disease. Translational research: the journal of laboratory and clinical medicine, 159, 228–237.
  • Rudolph, T.K., Rudolph, V., and Baldus, S., 2008. Contribution of myeloperoxidase to smoking-dependent vascular inflammation. Proceedings of the American thoracic society, 5, 820–823.
  • Rudolph, T.K., et al., 2012. Myeloperoxidase deficiency preserves vasomotor function in humans. European heart journal, 33, 1625–1634.
  • Rufino, R., et al., 2017. Spirometry reference values in the Brazilian population. Brazilian journal of medical and biological research, 50, e5700
  • Ruggiero, C., et al., 2007. White blood cell count and mortality in the Baltimore longitudinal study of aging. Journal of the American college of cardiology, 49, 1841–1850.
  • Runarsdottir, S.B., et al., 2013. Prevalence of airflow obstruction in nonsmoking older individuals using different spirometric criteria: the AGES Reykjavik Study. COPD, 10, 493–499.
  • Rusznak, C., et al., 2000. Effect of cigarette smoke on the permeability and IL-1beta and sICAM-1 release from cultured human bronchial epithelial cells of never-smokers, smokers, and patients with chronic obstructive pulmonary disease. American journal of respiratory cell and molecular biology, 23, 530–536.
  • Saareks, V., et al., 2001. Effects of smoking cessation and nicotine substitution on systemic eicosanoid production in man. Naunyn-Schmiedeberg’s archives of pharmacology, 363, 556–561.
  • Sadler, J.E., 1998. Biochemistry and genetics of von Willebrand factor. Annual review of biochemistry, 67, 395–424.
  • Salive, M.E., et al., 1992. Serum albumin in older persons: relationship with age and health status. Journal of clinical epidemiology, 45, 213–221.
  • Sapey, E., et al., 2008. Inter-relationships between inflammatory markers in patients with stable COPD with bronchitis: intra-patient and inter-patient variability. Thorax, 63, 493–499.
  • Sato, K., et al., 2014. Association between plasma adiponectin levels and decline in forced expiratory volume in 1 s in a general Japanese population: the Takahata study. International journal of medical science, 11, 758–764.
  • Sayyah-Melli, M., et al., 2016. The effect of high dose folic acid throughout pregnancy on homocysteine (hcy) concentration and pre-eclampsia: a randomized clinical trial. PLoS one, 11, e0154400.
  • Sbarouni, E., Georgiadou, P., and Voudris, V., 2011. Gender-specific differences in biomarkers responses to acute coronary syndromes and revascularization procedures. Biomarkers, 16, 457–465.
  • Scherer, G., 2006. Carboxyhemoglobin and thiocyanate as biomarkers of exposure to carbon monoxide and hydrogen cyanide in tobacco smoke. Experimental and toxicologic pathology, 58, 101–124.
  • Schindhelm, R.K., et al., 2009. Myeloperoxidase: a useful biomarker for cardiovascular disease risk stratification?. Clinical chemistry, 55, 1462–1470.
  • Schwab, K.O., et al., 2008. Marked smoking-associated increase of cardiovascular risk in childhood type 1 diabetes. International journal of adolescent medicine and health, 20, 285–292.
  • Schwartz, G.G., et al., 2012. Effects of dalcetrapib in patients with a recent acute coronary syndrome. New England journal of medicine, 367, 2089–2099.
  • Seemungal, T.A., et al., 2007. Plasma homocysteine is elevated in COPD patients and is related to COPD severity. International journal of chronic obstructive pulmonary disease, 2, 313–321.
  • Shankar, S., Suchetha, K.N., and Madhu, L.N., 2012. Variations in serum myeloperoxidase levels with respect to hyperglycemia, duration of diabetes, BMI, sex and aging in type 2 diabetes mellitus. International journal of research in pharmaceutical and biomedical sciences, 3, 652–655.
  • Shao, B., 2012. Site-specific oxidation of apolipoprotein A-I impairs cholesterol export by ABCA1, a key cardioprotective function of HDL. Biochimica et biophysica acta, 1821, 490–501.
  • Shao, B., et al., 2005. Acrolein impairs ATP binding cassette transporter A1-dependent cholesterol export from cells through site-specific modification of apolipoprotein A-I. Journal of biological chemistry, 280, 36386–36396.
  • Sharifov, O.F., et al., 2013. Anti-inflammatory mechanisms of apolipoprotein A-I mimetic peptide in acute respiratory distress syndrome secondary to sepsis. PLoS one, 8, e64486.
  • Sharma, G. and Berger, J.S., 2011. Platelet activity and cardiovascular risk in apparently healthy individuals: a review of the data. Journal of thrombosis and thrombolysis, 32, 201–208.
  • Sharma, G. and Goodwin, J., 2006. Effect of aging on respiratory system physiology and immunology. Clinical interventions in aging, 1, 253–260.
  • Sharma, S. and Lipincott, W., 2017. Biomarkers in Alzheimer's disease-recent update. Current Alzheimer research, doi: 10.2174/1567205014666170220141822. [Epub ahead of print]
  • Sherman, C.B., et al., 1992. Longitudinal lung function decline in subjects with respiratory symptoms. The American review of respiratory disease, 146, 855–859.
  • Shibata, Y., et al., 2013. Relationship between plasma fibrinogen levels and pulmonary function in the Japanese population: the Takahata study. International journal of medical science, 10, 1530–1536.
  • Shiels, M.S., et al., 2014. Cigarette smoking and variations in systemic immune and inflammation markers. Journal of the national cancer institute, 106, dju294.
  • Shinkai, H., 2012. Cholesteryl ester transfer-protein modulator and inhibitors and their potential for the treatment of cardiovascular diseases. Vascular health and risk management, 8, 323–331.
  • Shiue, I., 2015. Urinary thiocyanate concentrations are associated with adult cancer and lung problems: US NHANES, 2009-2012. Environmental science and pollution research international, 22, 5952–5960.
  • Siemons, L., et al., 2014. How age and sex affect the erythrocyte sedimentation rate and C-reactive protein in early rheumatoid arthritis. BMC musculoskeletal disorders, 15, 368.
  • Sies, H., 2014. Role of metabolic H2O2 generation: redox signaling and oxidative stress. The journal of biological chemistry, 289, 8735–8741.
  • Simon, D., et al., 1989. Epidemiological features of glycated haemoglobin A1c-distribution in a healthy population. The telecom study. Diabetologia, 32, 864–869.
  • Simpson, J.L., et al., 2013. Influence of age, past smoking, and disease severity on TLR2, neutrophilic inflammation, and MMP-9 levels in COPD. Mediators of inflammation, 2013, 462934.
  • Singh, D., et al., 2010. Sputum neutrophils as a biomarker in COPD: findings from the ECLIPSE study. Respiratory research, 11, 77
  • Sirnio, P., et al., 2017. Decreased serum apolipoprotein A1 levels are associated with poor survival and systemic inflammatory response in colorectal cancer. Scientific reports, 7, 5374.
  • Skloot, G.S., 2017. The effects of aging on lung structure and function. Clinics in geriatric medicine, 33, 447–457.
  • Skoog, T., et al., 2002. Plasma tumour necrosis factor-alpha and early carotid atherosclerosis in healthy middle-aged men. European heart journal, 23, 376–383.
  • Skulachev, V.P., 2012. Mitochondria-targeted antioxidants as promising drugs for treatment of age-related brain diseases. Journal of Alzheimer’s disease, 28, 283–289.
  • Smith, J.D., 2010. Apolipoprotein A-I and its mimetics for the treatment of atherosclerosis. Current opinion in investigational drugs (London, England: 2000), 11, 989–996.
  • Sobczak, A., et al., 2007. [The influence of tobacco smoke on homocysteine level in plasma of healthy males]. Przeglad lekarsi, 64, 679–684.
  • Sobczak, A.J., 2003. The effects of tobacco smoke on the homocysteine level – a risk factor of atherosclerosis. Addiction biology, 8, 147–158.
  • Sonneveld, M.A., et al., 2015. Von Willebrand factor in relation to coronary plaque characteristics and cardiovascular outcome. Results of the ATHEROREMO-IVUS study. Thrombosis and haemostasis, 113, 577–584.
  • Spanevello, A., et al., 2000. Induced sputum cellularity. Reference values and distribution in normal volunteers. American journal of respiratory and critical care medicine, 162, 1172–1174.
  • Sparrow, D., et al., 1984. The relationship of the peripheral leukocyte count and cigarette smoking to pulmonary function among adult men. Chest, 86, 383–386.
  • Spiel, A.O., Gilbert, J.C., and Jilma, B., 2008. von Willebrand factor in cardiovascular disease: focus on acute coronary syndromes. Circulation, 117, 1449–1459.
  • Stafforini, D.M., 2009. Biology of platelet-activating factor acetylhydrolase (PAF-AH, lipoprotein associated phospholipase A2). Cardiovascular drugs and therapy, 23, 73–83.
  • Stepanov, I., et al., 2009. Evidence for endogenous formation of N'-nitrosonornicotine in some long-term nicotine patch users. Nicotine & tobacco research, 11, 99–105.
  • Stephen, R., et al., 2014. Albuminuria: when urine predicts kidney and cardiovascular disease. Cleveland clinic journal of medicine, 81, 41–50.
  • Stiles, A.R., et al., 2014. Genetic, anatomic, and clinical determinants of human serum sterol and vitamin D levels. Proceedings of the national academy of sciences of the United States of America, 111, E4006–E4014.
  • Stone, H., et al., 2012. Variability of sputum inflammatory mediators in COPD and alpha1-antitrypsin deficiency. European respiratory journal, 40, 561–569.
  • Strassburg, A., et al., 2004. Effect of age on plasma homocysteine concentrations in young and elderly subjects considering serum vitamin concentrations and different lifestyle factors. International journal of vitamin and nutrition research, 74, 129–136.
  • Stratton, K., et al., 2001. Clearing the smoke: the science base for tobacco harm reduction-executive summary. Tobacco control, 10, 189–195.
  • Strimbu, K. and Tavel, J.A., 2010. What are biomarkers?. Current opinion in HIV and AIDS, 5, 463–466.
  • Stubbe, I., Eskilsson, J., and Nilsson-Ehle, P., 1982. High-density lipoprotein concentrations increase after stopping smoking. British medical journal (Clinical Research Ed.), 284, 1511–1513.
  • Subar, A.F., Harlan, L.C., and Mattson, M.E., 1990. Food and nutrient intake differences between smokers and non-smokers in the US. American journal of public health, 80, 1323–1329.
  • Sugiura, H., et al., 2012. Increased 25-hydroxycholesterol concentrations in the lungs of patients with chronic obstructive pulmonary disease. Respirology, 17, 533–540.
  • Sumino, H., et al., 1998. Decreased basal production of nitric oxide in patients with heart disease. Chest, 113, 317–322.
  • Sun, K., et al., 2017. Associations between homocysteine metabolism related SNPs and carotid intima-media thickness: a Chinese sib pair study. Journal of thrombosis and thrombolysis, 43, 401–410.
  • Sun, W., et al., 2016. Common genetic polymorphisms influence blood biomarker measurements in COPD. PLoS Genetics, 12, e1006011.
  • Sundy, J.S., et al., 2007. Smoking is associated with an age-related decline in exhaled nitric oxide. The European respiratory journal, 30, 1074–1081.
  • Swan, G.E., et al., 1991. Quantitative analysis of sputum cytologic differences between smokers and nonsmokers. Diagnostic cytopathology, 7, 569–575.
  • Szadkowski, A. and Myers, C.R., 2008. Acrolein oxidizes the cytosolic and mitochondrial thioredoxins in human endothelial cells. Toxicology, 243, 164–176.
  • Takizawa, H., et al., 2000. Increased expression of inflammatory mediators in small-airway epithelium from tobacco smokers. American journal of physiology-lung cellular and molecular physiology, 278, L906–L913.
  • Tall, A.R., 1998. An overview of reverse cholesterol transport. European heart journal, 19(SupplA), A31–A35.
  • Tan, D.B.A., et al., 2017. Elevated levels of circulating exosome in COPD patients are associated with systemic inflammation. Respiratory medicine, 132, 261–264.
  • Tanni, S.E., et al., 2010. Smoking status and tumor necrosis factor-alpha mediated systemic inflammation in COPD patients. Journal of inflammation, 7, 29.
  • Teixeira, P.C., Cutler, P., and Vuilleumier, N., 2012. Autoantibodies to apolipoprotein A-1 in cardiovascular diseases: current perspectives. Clinical and developmental immunology, 2012, 868251.
  • Tell, G.S., et al., 1985. The relationship of white cell count, platelet count, and hematocrit to cigarette smoking in adolescents: the Oslo Youth Study. Circulation, 72, 971–974.
  • Tembe, N., et al., 2014. Reference values for clinical laboratory parameters in young adults in Maputo, Mozambique. PLoS one, 9, e97391.
  • Thanan, R., et al., 2014. Oxidative stress and its significant roles in neurodegenerative diseases and cancer. International journal of molecular sciences, 16, 193–217.
  • Thelle, D.S., Forde, O.H., and Arnesen, E., 1982. Distribution of high-density lipoprotein cholesterol according to age, sex, and ethnic origin: cardiovascular disease study in Finnmark 1977. Journal of epidemiology and community health, 36, 243–247.
  • Thomas, R.A., et al., 2004. The influence of age on induced sputum differential cell counts in normal subjects. Chest, 126, 1811–1814.
  • Thukkani, A.K., et al., 2003. Identification of alpha-chloro fatty aldehydes and unsaturated lysophosphatidylcholine molecular species in human atherosclerotic lesions. Circulation, 108, 3128–3133.
  • Toljamo, T., et al., 2010. Early detection of COPD combined with individualized counselling for smoking cessation: a two-year prospective study. Scandinavian journal of primary health care, 28, 41–46.
  • Tommola, M., et al., 2016. The effect of smoking on lung function: a clinical study of adult-onset asthma. The European respiratory journal, 48, 1298–1306.
  • Tonstad, S. and Cowan, J.L., 2009. C-reactive protein as a predictor of disease in smokers and former smokers: a review. International journal of clinical practice, 63, 1634–1641.
  • Toren, K., et al., 2017. Reference values of fractional excretion of exhaled nitric oxide among non-smokers and current smokers. BMC pulmonary medicine, 17, 118.
  • Tran, B. and Kalhan, R., 2008. The emerging role of systemic inflammation in chronic obstructive pulmonary disease. Clinical pulmonary medicine, 15, 22–125.
  • Tricker, A.R., 1997. N-nitroso compounds and man: sources of exposure, endogenous formation and occurrence in body fluids. European journal of cancer prevention, 6, 226–268.
  • Tricker, A.R., et al., 2012. Reduced exposure evaluation of an electrically heated cigarette smoking system. Part 4: eight-day randomized clinical trial in Korea. Regulatory toxicology and pharmacology, 64, S45–S53.
  • Tsai, J.S., et al., 2012. Changes of serum adiponectin and soluble intercellular adhesion molecule-1 concentrations after smoking cessation. Clinical chemistry and laboratory medicine, 50, 1063–1069.
  • Tsao, M.S., et al., 2015. Predictive versus prognostic value of lung adenocarcinoma classification. Journal of clinical oncology, 33, 225–226.
  • Tselepis, A.D., et al., 2009. Smoking induces lipoprotein-associated phospholipase A2 in cardiovascular disease free adults: the ATTICA Study. Atherosclerosis, 206, 303–308.
  • Tsimihodimos, V., et al., 2002. Atorvastatin preferentially reduces LDL-associated platelet-activating factor acetylhydrolase activity in dyslipidemias of type IIA and type IIB. Arteriosclerosis, thrombosis, and vascular biology, 22, 306–311.
  • Tsoumakidou, M., Tzanakis, N., and Siafakas, N.M., 2003. Induced sputum in the investigation of airway inflammation of COPD. Respiratory medicine, 97, 863–871.
  • Tsuda, M. and Kurashima, Y., 1991. Tobacco smoking, chewing, and snuff dipping: factors contributing to the endogenous formation of N-nitroso compounds. Critical reviews in toxicology, 21, 243–253.
  • Uedelhoven, W.M., et al., 1991. Smoking alters thromboxane metabolism in man. Biochimica et biophysica acta, 1081 (2), 197–201.
  • Urrutia, I., et al., 2005. Smoking habit, respiratory symptoms and lung function in young adults. European journal of public health, 15, 160–165.
  • US Department of Health and Human Services. 2014. The health consequences of smoking-50 years of progress: a report of the surgeon general. Atlanta (GA): US Department of Health and Human Services.
  • van't Erve, T.J., et al., 2016. Reinterpreting the best biomarker of oxidative stress: the 8-iso-prostaglandin F2alpha/prostaglandin F2alpha ratio shows complex origins of lipid peroxidation biomarkers in animal models. Free radical biology & medicine, 95, 65–73.
  • van't Erve, T.J., et al., 2017. Classifying oxidative stress by F2-isoprostane levels across human diseases: a meta-analysis. Redox biology, 12, 582–599.
  • van't Erve, T.J., et al., 2015. Reinterpreting the best biomarker of oxidative stress: the 8-iso-PGF(2alpha)/PGF(2alpha) ratio distinguishes chemical from enzymatic lipid peroxidation. Free radical biology & medicine, 83, 245–251.
  • Van Dijk, W., et al., 2015. Clinical relevance of fixed ratio vs lower limit of normal of FEV1/FVC in COPD: patient-reported outcomes from the CanCOLD cohort. Annals of family medicine, 13, 41–48.
  • Van Galen, K.P., et al., 2012. Von Willebrand factor deficiency and atherosclerosis. Blood reviews, 26, 189–196.
  • Van Staden, S.R., et al., 2013. Carboxyhaemoglobin levels, health and lifestyle perceptions in smokers converting from tobacco cigarettes to electronic cigarettes. South African medical journal, 103, 865–868.
  • Vardavas, C.I., et al., 2008. Smoking status in relation to serum folate and dietary vitamin intake. Tobacco induced diseases, 4, 8
  • Vartanian, L.S. and Gurevich, S.M., 1989. [NADH- and NADPH-dependent formation of superoxide radicals in liver nuclei]. Biokhimiia, 54, 1020–1025.
  • Veering, B.T., et al., 1990. The effect of age on serum concentrations of albumin and alpha 1-acid glycoprotein. British journal of clinical pharmacology, 29, 201–206.
  • Verbanck, S., et al., 2004. Noninvasive assessment of airway alterations in smokers: the small airways revisited. American journal of respiratory and critical care medicine, 170, 414–419.
  • Verleden, G.M., et al., 1999. The effect of cigarette smoking on exhaled nitric oxide in mild steroid-naive asthmatics. Chest, 116, 59–64.
  • Vestbo, J., 2014. Chronic bronchitis: so much more than just a smoker's cough. The international journal of tuberculosis and lung disease, 18, 760.
  • Vestbo, J., et al., 2016. Global Strategy for the diagnosis, management and prevention of chronic obstructive pulmonary disease. Gold Executive Summary. American journal of respiratory and critical care medicine, 187, 347–365.
  • Viegi, G., et al., 1988a. Variability of maximal expiratory flow-volume curve in young volley players. Respiration, 54, 33–41.
  • Viegi, G., et al., 1988b. Prevalence of respiratory symptoms in an unpolluted area of northern Italy. The European respiratory journal, 1, 311–318.
  • Vischer, U.M., 2006. von Willebrand factor, endothelial dysfunction, and cardiovascular disease. Journal of thrombosis and haemostasis, 4, 1186–1193.
  • Vittos, O., et al., 2012. Lipoprotein-associated phospholipase A2 (Lp-PLA2): a review of its role and significance as a cardiovascular biomarker. Biomarkers, 17, 289–302.
  • Vlassopoulos, A., Lean, M.E., and Combet, E., 2013. Influence of smoking and diet on glycated haemoglobin and 'pre-diabetes' categorisation: a cross-sectional analysis. BMC public health, 13, 1013.
  • Voight, B.F., et al., 2012. Plasma HDL cholesterol and risk of myocardial infarction: a Mendelian randomisation study. Lancet, 380, 572–580.
  • Vollmer, W.M., et al., 1989. Respiratory symptoms, lung function, and mortality in a screening center cohort. American journal of epidemiology, 129, 1157–1169.
  • Voulgari, C., Katsilambros, N., and Tentolouris, N., 2011. Smoking cessation predicts amelioration of microalbuminuria in newly diagnosed type 2 diabetes mellitus: a 1-year prospective study. Metabolism, 60, 1456–1464.
  • Vuilleumier, N., Montecucco, F., and Hartley, O., 2014. Autoantibodies to apolipoprotein A-1 as a biomarker of cardiovascular autoimmunity. World journal of cardiology, 6, 314–326.
  • Vuilleumier, N., et al., 2004. Presence of autoantibodies to apolipoprotein A-1 in patients with acute coronary syndrome further links autoimmunity to cardiovascular disease. Journal of autoimmunity, 23, 353–360.
  • Wadhwani, N.S., et al., 2016. Increased homocysteine levels exist in women with preeclampsia from early pregnancy. The journal of maternal-fetal & neonatal medicine, 29, 2719–2725.
  • Wang, H., et al., 2001. Serum thiocyanate concentration as an indicator of smoking in relation to deaths from cancer. Environmental health and preventive medicine, 6, 88–91.
  • Wang, Q., et al., 2015. Serum apolipoprotein A-1 quantification by LC-MS with a SILAC internal standard reveals reduced levels in smokers. Bioanalysis, 7, 2895–2911.
  • Wang, Z., et al., 2017. Influences of ABO blood group, age and gender on plasma coagulation factor VIII, fibrinogen, von Willebrand factor and ADAMTS13 levels in a Chinese population. PeerJ, 5, e3156.
  • Wang, Z., et al., 2007. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nature medicine, 13, 1176–1184.
  • Wannamethee, S.G., et al., 2005. Associations between cigarette smoking, pipe/cigar smoking, and smoking cessation, and haemostatic and inflammatory markers for cardiovascular disease. European heart journal, 26, 1765–1773.
  • Weber, A.A., et al., 2000. Habitual smoking causes an abnormality in platelet thromboxane A2 metabolism and results in an altered susceptibility to aspirin effects. Platelets, 11, 177–182.
  • Wei, B., et al., 2016. Assessing exposure to tobacco-specific carcinogen NNK using its urinary metabolite NNAL measured in US population: 2011-2012. Journal of exposure science and environmental epidemiology, 26, 249–256.
  • Wei, Y.F., et al., 2014. The impact of metabolic parameters on the change of pulmonary function in obese patients. Surgery of obesity and related diseases, 10, 23–28.
  • Wennmalm, A., et al., 1991. Relation between tobacco use and urinary excretion of thromboxane A2 and prostacyclin metabolites in young men. Circulation, 83, 1698–1704.
  • Wennmalm, A., et al., 1993. Maintained hyperexcretion of thromboxane A2 metabolite in healthy young cigarette smokers: results from a prospective study in randomly sampled males with stable smoking habits. Clinical physiology, 13, 257–264.
  • Whereat, A.F., 1970. Is atherosclerosis a disorder of intramitochondrial respiration? Annals of internal medicine, 73, 125–127.
  • Whincup, P.H., et al., 2002. von Willebrand factor and coronary heart disease: prospective study and meta-analysis. European heart journal, 23, 1764–1770.
  • Willemse, B.W., et al., 2005. Effect of 1-year smoking cessation on airway inflammation in COPD and asymptomatic smokers. The European respiratory journal, 26, 835–845.
  • Witkowska, A.M., 2005. Soluble ICAM-1: a marker of vascular inflammation and lifestyle. Cytokine, 31, 127–134.
  • Woloshin, S. and Schwartz, L.M., 2005. Distribution of C-reactive protein values in the United States. The new England journal of medicine, 352, 1611–1613.
  • Woodruff, P.G., et al., 2016. Clinical significance of symptoms in smokers with preserved pulmonary function. The new England journal of medicine, 374, 1811–1821.
  • Wu, L., et al., 2017. Effect of age on the diagnostic efficiency of HbA1c for diabetes in a Chinese middle-aged and elderly population: the Shanghai Changfeng Study. PLoS one, 12, e0184607.
  • Wu, W., et al., 2005. Determination of carcinogenic tobacco-specific nitrosamines in mainstream smoke from U.S.-brand and non-U.S.-brand cigarettes from 14 countries. Nicotine & tobacco research, 7, 443–451.
  • Wu, Y., et al., 2015. Increased serum TRAIL and DR5 levels correlated with lung function and inflammation in stable COPD patients. International journal of chronic obstructive pulmonary disease, 10, 2405–2412.
  • Wyczalkowska-Tomasik, A., et al., 2016. Inflammatory markers change with age, but do not fall beyond reported normal ranges. Archivum immunologiae et therapiae experimentalis, 64, 249–254.
  • Xia, Y., et al., 2011. Tobacco-specific nitrosamine 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL) in smokers in the United States: NHANES 2007-2008. Biomarkers, 16, 112–119.
  • Xie, Y., et al., 2017. Association of plasma homocysteine, vitamin B12 and folate levels with cognitive function in Parkinson's disease: a meta-analysis. Neuroscience letters, 636, 190–195.
  • Xu, X., et al., 2016. A population-based study of smoking, serum cotinine and exhaled nitric oxide among asthmatics and a healthy population in the USA. Inhalation toxicology, 28, 724–730.
  • Yanbaeva, D.G., et al., 2007. Systemic effects of smoking. Chest, 131, 1557–1566.
  • Yang, L., et al., 2017. Association between Lp-PLA2 and coronary heart disease in Chinese patients. The journal of international medical research, 45, 159–169.
  • Yates, D.H., Breen, H., and Thomas, P.S., 2001. Passive smoke inhalation decreases exhaled nitric oxide in normal subjects. American journal of respiratory and critical care medicine, 164, 1043–1046.
  • Yeung, M.C. and Buncio, A.D., 1984. Leukocyte count, smoking, and lung function. The american journal of medicine, 76, 31–37.
  • Yoon, H.I. and Sin, D.D., 2011. Biomarkers of therapeutic response in patients with chronic obstructive pulmonary disease: a critical review of the literature. Drugs, 71, 1821–1837.
  • Yuan, J., et al., 2015. Differences of plasma IL-1 and TNF-α in healthy Chinese population. Open medicine (Warsaw, Poland), 10, 306–310.
  • Yuan, J.M., et al., 2011. Urinary levels of the tobacco-specific carcinogen N'-nitrosonornicotine and its glucuronide are strongly associated with esophageal cancer risk in smokers. Carcinogenesis, 32, 1366–1371.
  • Yuan, J.M., et al., 2009. Urinary levels of tobacco-specific nitrosamine metabolites in relation to lung cancer development in two prospective cohorts of cigarette smokers. Cancer research, 69, 2990–2995.
  • Zaynagetdinov, R., et al., 2016. Chronic NF-kappaB activation links COPD and lung cancer through generation of an immunosuppressive microenvironment in the lungs. Oncotarget, 7, 5470–5482.
  • Zelova, H. and Hosek, J., 2013. TNF-alpha signalling and inflammation: interactions between old acquaintances. Inflammation research, 62, 641–651.
  • Zezulka, A.V., Gill, J.S., and Beevers, D.G., 1987. 'Neutropenia' in black west Indians. Postgraduate medical journal, 63, 257–261.
  • Zhang, F., et al., 2016. The C-reactive protein/albumin ratio predicts long-term outcomes of patients with operable non-small cell lung cancer. Oncotarget, 8 (5), 8835–8842.
  • Zheng, L., et al., 2004. Apolipoprotein A-I is a selective target for myeloperoxidase-catalyzed oxidation and functional impairment in subjects with cardiovascular disease. Journal of clinical investigation, 114, 529–541.
  • Zhou, Z., et al., 2014. Possible race and gender divergence in association of genetic variations with plasma von Willebrand factor: a study of ARIC and 1000 genome cohorts. PLoS one, 9, e84810.
  • Zhu, L., et al., 2016. A new prognostic score based on the systemic inflammatory response in patients with inoperable non-small-cell lung cancer. Oncotargets and therapy, 9, 4879–4886.
  • Zil, A.R. and Rahman, M.A., 2006. Serum thiocyanate levels in smokers, passive smokers and never smokers. Journal of Pakistan medical association, 56, 323–326.
  • Zuiker, R.G., et al., 2010. Sputum induction with hypertonic saline reduces fractional exhaled nitric oxide in chronic smokers and non-smokers. Respiratory medicine, 104, 917–920.
  • Zuiker, R.G., et al., 2015. Reproducibility of biomarkers in induced sputum and in serum from chronic smokers. Pulmonary pharmacology & therapeutics, 33, 81–86.