Abstract

Introduction

Since 2009, the United States (US) Food and Drug Administration (FDA) Center for Tobacco Products (CTP) has had the authority to regulate the manufacture, distribution, and marketing of tobacco products in order to reduce the death and disease caused by tobacco use. Biomarkers could play an important role across a number of FDA regulatory activities, including assessing new and modified risk tobacco products and identifying and evaluating potential product standards.

Methods

On April 4–5, 2016, FDA/CTP hosted a public workshop focused on biomarkers of potential harm (BOPH) with participants from government, industry, academia, and other organizations. The workshop was divided into five sessions focused on: (1) overview of BOPH; (2) cardiovascular disease (CVD); (3) chronic obstructive pulmonary disease (COPD); (4) cancer; and (5) new areas of research.

Results and Conclusions

The deliberations from the workshop noted some promising BOPH but also highlighted the lack of systematic effort to identify BOPH that would have utility and validity for evaluating tobacco products. Research areas that could further strengthen the applicability of BOPH to tobacco regulatory science include the exploration of composite biomarkers as predictors of disease risk, “omics” biomarkers, and examining biomarkers using existing cohorts, surveys, and experimental studies.

Implications

This paper synthesizes the main findings from the 2016 FDA-sponsored workshop focused on BOPH and highlights research areas that could further strengthen the science around BOPH and their applicability to tobacco regulatory science.

Introduction

The United States (US) Food and Drug Administration (FDA) Center for Tobacco Products (CTP), established in 2009 by the Family Smoking Prevention and Tobacco Control Act (Tobacco Control Act), has the broad authority to regulate the manufacturing, distribution, and marketing of tobacco products with the ultimate goal of reducing harm caused by tobacco use.1 Under the 2009 Tobacco Control Act, FDA immediately regulated cigarettes, roll-your-own tobacco and smokeless tobacco. In 2016, FDA finalized a rule extending the FDA’s authority to regulate all tobacco products, including electronic nicotine delivery systems (ENDS), cigars, hookah tobacco, pipe tobacco and nicotine gels, and any future tobacco products.2 Although the health risks of cigarettes and, to a lesser extent, other traditional tobacco products have been well characterized, less is known about the health risks of more novel combustible and noncombustible tobacco products that have been introduced in recent years. FDA’s regulatory authorities include the review of premarket tobacco product applications and modified risk tobacco product applications, as well as, setting tobacco product standards. FDA uses a population health standard to make regulatory decisions by taking into account the benefits and risks of the product to both users and nonusers of tobacco products and assessing the “net” population-level health impacts of tobacco products.

Biomarkers can play an important role in characterizing the potential health risks of tobacco products, and, therefore, help assess the impact of FDA actions on the population health. In the 2001 Institute of Medicine report Clearing the Smoke, biomarkers of potential harm (BOPH) are defined as the “measurement of an effect due to exposure; these include early biological effects, alterations in morphology, structure, or function, and clinical symptoms consistent with harm; also includes ‘preclinical changes’”.3 Although clinical outcomes such as cancer, cardiovascular disease (CVD), and chronic obstructive pulmonary disease (COPD) are definitive endpoints, they take decades to develop and thus are not always practical to quickly assess in the premarket regulatory setting. BOPH could serve as more intermediate endpoints for assessing potential health risk of new and novel tobacco products in the absence of long-term epidemiological evidence.3 However, as stated in the 2001 IOM report, “Few specific early indicators of biomarkers have been validated as predictive of later disease development.”3 To understand the current state of the science on BOPH, the FDA invited experts in the field to participate in a public workshop. This report is an integration of the different viewpoints expressed at the workshop.

Materials and Methods

On April 4–5, 2016, FDA/CTP hosted a public workshop to gather information on: (1) approaches to assessing and selecting BOPH; (2) the processes of identifying BOPH that may be useful in tobacco product regulation; and (3) areas of research that may further strengthen knowledge about BOPH.4 Previously, FDA/CTP hosted a public workshop on biomarkers of exposure (BOE).5 The BOPH workshop consisted of participants from government, industry, academia, and other organizations (Supplementary Table 1). The five sessions focused on: (1) overview of BOPH; (2) CVD; (3) COPD; (4) cancer; and (5) new areas of research.

Results

Overview of BOPH

BOPH can encompass a number of types of biomarkers, from those that indicate a risk factor for disease (biomarker of risk) to those that are meant to substitute for a clinical endpoint (surrogate endpoint; Table 1). Biomarkers used as validated surrogate endpoints often fail to predict the clinical outcome of interest.6,7 One well-known example is the Cardiac Arrhythmia Suppression Trial in which patients randomized to the study drugs designed to suppress arrhythmia were more (rather than less) likely to die of arrhythmia or shock than placebo patients.8 Fleming and DeMets explained the reasons for such failures as one or more of the following: (1) the biomarker under study is not found on the pathophysiologic pathway that leads to the clinical outcome of interest; (2) the biomarker is used to test an intervention associated with only one pathway when multiple causal pathways to a particular clinical outcome exist; (3) the biomarker is insensitive to or is not a part of the causal pathway of the intervention’s effect or is insensitive to this effect; or (4) the intervention results in additional mechanisms of action (including other harmful effects) independent of the disease process.9

Table 1.

Additional Definitions/Terms for Biomarkers of Potential Harm (BOPH)

BiomarkerDefinitionReference
Biomarkers of riskBiomarker that indicates a risk factor for disease.14
Surrogate endpointAn endpoint that is used in clinical trials as a substitute for a direct measure of how a patient feels, functions, or survives. A surrogate endpoint does not measure the clinical benefit of primary interest in and of itself, but rather is expected to predict that clinical benefit or harm based on epidemiologic, therapeutic, pathophysiologic, or other scientific evidence.99
Reasonably likely surrogate endpointAn endpoint supported by strong mechanistic and/or epidemiologic rationale such that an effect on the surrogate endpoint is expected to be correlated with an endpoint intended to assess clinical benefit in clinical trials, but without sufficient clinical data to show that it is a validated surrogate endpoint. Such endpoints may be used for accelerated approval for drugs and potentially also for approval or clearance of medical devices. In the case of accelerated approval for drugs, postmarketing confirmatory trials have been required to verify and describe the anticipated effect on the irreversible morbidity or mortality or other clinical benefit.99
Pharmacodynamic/response biomarkerA biomarker used to show that a biological response has occurred in an individual who has received an intervention or exposure.99
Predictive biomarkerA biomarker used to identify individuals who are more likely than similar individuals without the biomarker to experience a favorable or unfavorable effect from exposure to a medical product or an environmental agent.99
Biomarkers of effectIndicators of a change in biologic function in response to a chemical exposure.100
BiomarkerDefinitionReference
Biomarkers of riskBiomarker that indicates a risk factor for disease.14
Surrogate endpointAn endpoint that is used in clinical trials as a substitute for a direct measure of how a patient feels, functions, or survives. A surrogate endpoint does not measure the clinical benefit of primary interest in and of itself, but rather is expected to predict that clinical benefit or harm based on epidemiologic, therapeutic, pathophysiologic, or other scientific evidence.99
Reasonably likely surrogate endpointAn endpoint supported by strong mechanistic and/or epidemiologic rationale such that an effect on the surrogate endpoint is expected to be correlated with an endpoint intended to assess clinical benefit in clinical trials, but without sufficient clinical data to show that it is a validated surrogate endpoint. Such endpoints may be used for accelerated approval for drugs and potentially also for approval or clearance of medical devices. In the case of accelerated approval for drugs, postmarketing confirmatory trials have been required to verify and describe the anticipated effect on the irreversible morbidity or mortality or other clinical benefit.99
Pharmacodynamic/response biomarkerA biomarker used to show that a biological response has occurred in an individual who has received an intervention or exposure.99
Predictive biomarkerA biomarker used to identify individuals who are more likely than similar individuals without the biomarker to experience a favorable or unfavorable effect from exposure to a medical product or an environmental agent.99
Biomarkers of effectIndicators of a change in biologic function in response to a chemical exposure.100
Table 1.

Additional Definitions/Terms for Biomarkers of Potential Harm (BOPH)

BiomarkerDefinitionReference
Biomarkers of riskBiomarker that indicates a risk factor for disease.14
Surrogate endpointAn endpoint that is used in clinical trials as a substitute for a direct measure of how a patient feels, functions, or survives. A surrogate endpoint does not measure the clinical benefit of primary interest in and of itself, but rather is expected to predict that clinical benefit or harm based on epidemiologic, therapeutic, pathophysiologic, or other scientific evidence.99
Reasonably likely surrogate endpointAn endpoint supported by strong mechanistic and/or epidemiologic rationale such that an effect on the surrogate endpoint is expected to be correlated with an endpoint intended to assess clinical benefit in clinical trials, but without sufficient clinical data to show that it is a validated surrogate endpoint. Such endpoints may be used for accelerated approval for drugs and potentially also for approval or clearance of medical devices. In the case of accelerated approval for drugs, postmarketing confirmatory trials have been required to verify and describe the anticipated effect on the irreversible morbidity or mortality or other clinical benefit.99
Pharmacodynamic/response biomarkerA biomarker used to show that a biological response has occurred in an individual who has received an intervention or exposure.99
Predictive biomarkerA biomarker used to identify individuals who are more likely than similar individuals without the biomarker to experience a favorable or unfavorable effect from exposure to a medical product or an environmental agent.99
Biomarkers of effectIndicators of a change in biologic function in response to a chemical exposure.100
BiomarkerDefinitionReference
Biomarkers of riskBiomarker that indicates a risk factor for disease.14
Surrogate endpointAn endpoint that is used in clinical trials as a substitute for a direct measure of how a patient feels, functions, or survives. A surrogate endpoint does not measure the clinical benefit of primary interest in and of itself, but rather is expected to predict that clinical benefit or harm based on epidemiologic, therapeutic, pathophysiologic, or other scientific evidence.99
Reasonably likely surrogate endpointAn endpoint supported by strong mechanistic and/or epidemiologic rationale such that an effect on the surrogate endpoint is expected to be correlated with an endpoint intended to assess clinical benefit in clinical trials, but without sufficient clinical data to show that it is a validated surrogate endpoint. Such endpoints may be used for accelerated approval for drugs and potentially also for approval or clearance of medical devices. In the case of accelerated approval for drugs, postmarketing confirmatory trials have been required to verify and describe the anticipated effect on the irreversible morbidity or mortality or other clinical benefit.99
Pharmacodynamic/response biomarkerA biomarker used to show that a biological response has occurred in an individual who has received an intervention or exposure.99
Predictive biomarkerA biomarker used to identify individuals who are more likely than similar individuals without the biomarker to experience a favorable or unfavorable effect from exposure to a medical product or an environmental agent.99
Biomarkers of effectIndicators of a change in biologic function in response to a chemical exposure.100

Thus, the criteria (eg, biomarker characteristics and pathophysiology) for identifying, evaluating, and selecting BOPH require careful consideration.10–12 In the context of tobacco product evaluation, the criteria for biomarker evaluation and selection are described in the Institute of Medicine (IOM) 2001 (Clearing the Smoke) and 2012 (Modified Risk Tobacco Products) reports and are based on those developed by Hill.3,13,14Table 2 presents the questions that might be considered in determining whether a biomarker is an adequate indicator of potential harm, and we provide some examples for the criteria in the following.

Table 2.

Questions for Supporting Adequacy of Biomarker as Indicator of Potential Harm

CriteriaQuestion
Reflective of pathological process as a consequence of exposurePlausibility: Is the data elucidating the biological pathways from exposure to effect plausible?
Coherence: Is the cause and effect interpretation of the data not seriously in conflict with [the] generally known facts of the natural history and biology of the disease?
SensitivityHow sensitive is the biomarker in assessing alterations in biology and in its ability to detect disease?
PredictiveTo what extent are the biomarkers predictive of disease?
TemporalityDoes exposure precede the effect?
ExperimentIs there evidence showing the removal of exposure lessens or removes the effect?
Biological gradientIs the magnitude of exposure proportional to the magnitude of effect?
SpecificityIs the effect specific or are there other known causes?
AnalogyCan inferences be made based on data from other agents?
ConsistencyIs the relationship reproducible and observed by multiple investigators in different populations using different methodologies?
CriteriaQuestion
Reflective of pathological process as a consequence of exposurePlausibility: Is the data elucidating the biological pathways from exposure to effect plausible?
Coherence: Is the cause and effect interpretation of the data not seriously in conflict with [the] generally known facts of the natural history and biology of the disease?
SensitivityHow sensitive is the biomarker in assessing alterations in biology and in its ability to detect disease?
PredictiveTo what extent are the biomarkers predictive of disease?
TemporalityDoes exposure precede the effect?
ExperimentIs there evidence showing the removal of exposure lessens or removes the effect?
Biological gradientIs the magnitude of exposure proportional to the magnitude of effect?
SpecificityIs the effect specific or are there other known causes?
AnalogyCan inferences be made based on data from other agents?
ConsistencyIs the relationship reproducible and observed by multiple investigators in different populations using different methodologies?
Table 2.

Questions for Supporting Adequacy of Biomarker as Indicator of Potential Harm

CriteriaQuestion
Reflective of pathological process as a consequence of exposurePlausibility: Is the data elucidating the biological pathways from exposure to effect plausible?
Coherence: Is the cause and effect interpretation of the data not seriously in conflict with [the] generally known facts of the natural history and biology of the disease?
SensitivityHow sensitive is the biomarker in assessing alterations in biology and in its ability to detect disease?
PredictiveTo what extent are the biomarkers predictive of disease?
TemporalityDoes exposure precede the effect?
ExperimentIs there evidence showing the removal of exposure lessens or removes the effect?
Biological gradientIs the magnitude of exposure proportional to the magnitude of effect?
SpecificityIs the effect specific or are there other known causes?
AnalogyCan inferences be made based on data from other agents?
ConsistencyIs the relationship reproducible and observed by multiple investigators in different populations using different methodologies?
CriteriaQuestion
Reflective of pathological process as a consequence of exposurePlausibility: Is the data elucidating the biological pathways from exposure to effect plausible?
Coherence: Is the cause and effect interpretation of the data not seriously in conflict with [the] generally known facts of the natural history and biology of the disease?
SensitivityHow sensitive is the biomarker in assessing alterations in biology and in its ability to detect disease?
PredictiveTo what extent are the biomarkers predictive of disease?
TemporalityDoes exposure precede the effect?
ExperimentIs there evidence showing the removal of exposure lessens or removes the effect?
Biological gradientIs the magnitude of exposure proportional to the magnitude of effect?
SpecificityIs the effect specific or are there other known causes?
AnalogyCan inferences be made based on data from other agents?
ConsistencyIs the relationship reproducible and observed by multiple investigators in different populations using different methodologies?

Plausibility

“Plausibility” that the potential biomarker can occur along one or more of the physiological pathways to disease is helpful, especially for tobacco-related disease pathways that are well characterized, including cancer and CVD.15 However, plausibility is limited to the current state of the knowledge and may not be known yet for every given biomarker.

Coherence

“Coherence” between different lines of evidence, including laboratory and epidemiological, can also strengthen the case of a given biomarker. For example, in vitro studies on cells exposed to cigarette smoke or cigarette smoke condensate observed the increased expression of oxidative stress markers, whereas studies of humans have also demonstrated evidence of both local and systemic oxidative stress after short-term exposure to cigarette smoke.15

Sensitivity

“Sensitivity” of biomarkers can be assessed by examining the distribution and level of biomarkers in a population without the exposure/disease and by comparing the distribution of the biomarker in a population with the exposure/disease to assess the degree of discrimination.16 For example, in the cross-sectional, multi-center Total Exposure Study (TES, funded by Altria), 21 of the 29 selected BOPH, including many markers specific to cardiovascular risk, had shown significant differences between adult smokers and nonsmokers; seven of these had a greater than 10% difference, demonstrating sensitivity to differences by smoking status for some of the selected BOPH.17

Predictive Potential

The “predictive potential” of biomarkers include determining the extent to which the biomarker is predictive of a disease.16 For example, a pooled study of nine prospective observational studies demonstrated positive associations between diastolic blood pressure (DBP) and risks of stroke and coronary heart disease (CHD) and, most importantly, no evidence of a “threshold” below which lower levels of DBP were not associated with lower risk of stroke and CHD.18

Temporality, Experimentation, and Biological Gradient

“Temporality” can be determined by longitudinal studies but also inferred by experimentation and biological gradient. “Experimentation” includes examining the extent of changes as a result of cessation of the product. This type of study can also determine the “half-life” of the biomarker. The “biological gradient” is determined by the dose–response relationship between the level of harmful and potentially harmful constituents or exposure biomarkers with the biomarker of potential harm, and also by the change in biomarkers of harm with reduction in the extent of use of a product or in constituent yields within or across products.

Specificity, Analogy, and Consistency

Most biomarkers of potential harm have not shown “specificity” to tobacco and can be influenced by diet, cooking methods, ambient air, occupational settings, physical activity, etc.; therefore, these potential confounders should be considered and controlled. For example, in TES, body mass index, a potentially confounding factor, was the most important factor for 12 of the BOPH, demonstrating a lack of specificity to smoking.17 With regards to “analogy,” several nontobacco agents have demonstrated that lowering BOPH will reduce disease risk (eg, hypertension). Finally, some biomarkers have shown “consistency” or similar results across different study methods, tobacco products, and investigators (eg, white blood cells) demonstrating sensitivity to tobacco exposure effects.

Biomarker Development and Testing

Biomarker development and testing may include the following types of studies:16 (1) preclinical experimentation where potential biomarkers reflecting pathophysiology of disease are identified and assays for these biomarkers are validated; (2) cross-sectional biomarker studies involving people with and without the disease or by tobacco use status at a specific point in time; (3) experiments to determine whether levels and changes in tobacco use or exposure including secondhand smoke will affect the measured biomarker; (4) nested case–control studies where biospecimens have been collected prior to disease development and analyzed to determine whether predisease biomarker levels predict eventual disease; and (5) prospective longitudinal studies to determine whether changes in biomarker(s) will be able to predict disease development, either by reaching a certain threshold or continuously (no threshold). Several general factors should be considered when testing and characterizing BOPH in tobacco users: the representativeness of the population, which does not preclude focusing on special populations, including smokers at risk for the disease outcome (eg, genetics) or who are currently experiencing the disease; pharmacokinetic properties (half-life) of the biomarker, which will inform the required duration of a study testing tobacco products; use of a range of biomarkers reflective of a disease condition or different disease conditions; and assessment of confounding environmental, behavioral, and individual factors (eg, diet, environmental/occupational exposures, level of physical activity, sex, age, race, body mass index [BMI], alcohol use).

Pathophysiology and Biomarkers Associated With CVD, COPD, and Cancer

For the remaining sections of this report, definitions of physiological processes and other terms discussed are provided in Supplementary Table 2.

Biomarkers That Cut Across Diseases: Inflammation and Oxidative Stress

Inflammation and oxidative stress play roles across smoking-related CVD, COPD, and cancer and can be induced by smoking along with infection and other processes. Supplementary Table 3 characterizes some commonly studied inflammatory and oxidative stress markers as they relate to smoking and disease risk.

Cardiovascular Disease

CVD generally reflects conditions that involve the narrowing or blockage of blood vessels that can lead to myocardial infarction or stroke. Exposure to constituents of cigarette smoking known to contribute to CVD include: (1) oxidizing chemicals and particulates, both of which can increase inflammation and thrombosis or lead to blood clot formation and endothelial dysfunction; (2) carbon monoxide, which reduces the delivery of oxygen; (3) volatile organic compounds (VOCs) such as acrolein, which can cause cardiovascular toxicity; (4) heavy metals, which can damage endothelial cells; and (5) nicotine, known to activate the sympathetic nervous system and increase heart rate and blood pressure.15

Oxidative Stress and Inflammation

Oxidative stress resulting from smoke inhalation can lead to chronic inflammation, endothelial damage and dysfunction, platelet activation, impaired vasodilatation, adverse effects on blood lipids, pathologic angiogenesis, and enhanced thrombosis.19 Cigarette smoke activates the endothelium by the induction of adhesion molecule expression, as well as macrophages and platelets. In response to in vitro smoke exposure, endothelial cells are known to release inflammatory and proatherogenic cytokines, leading to endothelial dysfunction. Inflammation plays a key role in both atherogenesis and thrombotic events, two key processes that lead to acute coronary events such as a myocardial infarction.20

C-reactive protein (CRP) is a nonspecific marker of inflammation, and increased plasma CRP levels are associated with smoking and increased risk for CVD (Supplementary Table 3). A meta-analysis showed that risk of future coronary heart disease was approximately 50% higher in individuals with CRP levels in the top third of the distribution compared with those with CRP levels in the lower third of the distribution.21 A similar association was also found for interleukin-6 (IL-6), an inflammatory biomarker upstream of CRP.22 Fibrinogen is another inflammatory biomarker associated with cardiovascular events, though also associated with non-CVD deaths, suggesting low specificity for predicting CVD risk.23 Strong statistical associations between these biomarkers and CVD risk do not necessarily translate into their increased ability to predict future risk of disease development.21,24

Endothelial Dysfunction

The endothelium (inner lining of blood vessels) is critical for maintaining blood flow to various organs, including the heart and the brain. Thus, endothelial injury and dysfunction caused by smoking is thought to play a major role in acute cardiovascular events. Oxidants destroy and reduce nitric oxide, an effect considered to be the hallmark of endothelial dysfunction leading to impaired vasodilatation. The established biomarkers of the endothelial dysfunction include: % flow-mediated dilation (FMD) as measured by brachial artery ultrasound imaging, circulating plasma endothelial precursor cells and microvesicles, E-selectin, P-selectin, and von Willebrand factor.25–27 In the cross-sectional Tobacco Exposure Study (TES), plasma von Willebrand factor levels were significantly higher in smokers than in nonsmokers, though no trend was observed for increasing cigarettes per day.28 In a 1-year longitudinal study, smoking cessation was significantly associated with improved % FMD, whereas no change in % FMD occurred for continued smokers.27

Enhanced Thrombosis

Cigarette smoking increases the number and activation of platelets, leading to a prothrombotic and procoagulative state in the vascular wall of smokers. Platelets play a pivotal role in the pathogenesis of thrombosis, atherogenesis, and angiogenesis. Thus, platelet-derived biomarkers are promising, and include measurements of thrombocytosis or an overabundance of platelets, mean platelet volume, and immature platelets, all of which have been associated with adverse cardiac events or mortality.29 Other biomarkers of the hypercoagulable state include markers of platelet activation, red blood cell mass, and fibrinogen have been shown to be higher in smokers than in nonsmokers in cross-sectional studies.17,30

Hemodynamic Stress

Mediated primarily by nicotine, acute smoking leads to hemodynamic changes, including elevated heart rate, blood pressure, and cardiac output.15 As a result, the reserve of blood flow in the heart needed to respond to increased myocardial demand is inappropriately reduced in smokers, contributing to ischemic cardiac events. In an experimental study of 10 healthy volunteers, both heart rate and blood pressure increased with use of all forms of nicotine (cigarettes, oral snuff, chewing tobacco, nicotine gum) and returned to baseline values within 90 min.31 In another experimental study, 10 healthy volunteers smoking different types of cigarettes (usual brand, high- and low-nicotine research cigarettes) resulted in similar increases over a 24-h period in heart rate and blood pressure as compared with abstinence.32 Smoking cessation has been shown to immediately result in lower blood pressure.33 Despite the evidence of acute increases in blood pressure due to smoking, chronic effects on blood pressure, including hypertension, have not been consistently shown to be higher in smokers compared with nonsmokers in the general population.34

Blood levels of natriuretic peptides increase under cardiac dysfunction and can serve as biomarkers of subclinical hemodynamic stress. The increase of plasma B-type natriuretic peptide (BNP) was strongly associated with a variety of CVD risks, including sudden death, heart failure, atrial fibrillation, coronary heart disease, and stroke.35,36 In one study, current smokers had about a threefold increase in BNP levels than never smokers, suggesting smoking is one of the factors that causes subclinical increase in cardiac stress or dysfunction.37

Blood Lipids

Cigarette smoking is associated with changes in the blood lipid profile that can contribute to the development of fatty plaques and atherosclerosis.15 For example, in an analysis based on 54 studies, smokers compared with nonsmokers had significantly higher serum concentrations of total cholesterol (3% difference from values in nonsmokers), triglycerides (9%), and very low-density lipoprotein (VLDL; 10%), as well as decreased high-density lipoprotein (HDL) cholesterol (−6%).38 In a longitudinal study, smokers who quit had significantly increased HDL cholesterol after 1 year.39

Insulin Resistance

Cigarette smoking is associated with diabetes, a major risk factor for CVD.40 Insulin resistance and diabetes can be measured with glycated hemoglobin (hemoglobin A1c (HbA1c)), a glucose tolerance test, and glucose clamping studies. An analysis from the large cross-sectional Scottish Health Survey observed that smokers were twice as likely to have HbA1c in the “pre-diabetic” range as compared with nonsmokers (OR = 2.25, 95% CI = 1.84 to 2.75), suggesting that smoking adversely affects glucose homeostasis.41

Arrhythmogenesis

Smoking is associated with sudden cardiac death and arrhythmia, and catecholamine release is thought to play a role.42 This effect was observed in an experimental study of 12 smokers who received three doses of transdermal nicotine patches (21, 42, and 63 mg/24 h) and placebo (0 mg) each for 5 days, including 4 days of smoking (days 1–4) and smoking abstinence on the fifth day. While on the placebo patch, urinary epinephrine concentrations were higher by about 60% during the smoking period when compared with the nonsmoking period. Though epinephrine concentrations increased in participants on nicotine-containing patches, the dose–response was flat, suggesting that only a small level of nicotine was needed to cause this effect.43

Atherosclerosis

Atherosclerosis involves the narrowing and hardening of the arteries due to the build-up of fatty material or plaque, leading to reduced or blocked blood flow that can progress to adverse events, including heart attack or stroke. Coronary artery calcification (CAC) score is one example of a marker of subclinical atherosclerosis and has been shown to be predictive of coronary heart disease independent of conventional risk factors.44 In the Multi-Ethnic Study of Atherosclerosis (MESA) cohort, both former smoking and current smoking were associated with a CAC > 0 (compared to never smokers).45 One potential limitation of CAC is that it is a marker of an advanced stage of the disease process, and thus may not be reversible.46

Tobacco Product Evaluation With CVD Biomarkers

A cross-sectional study compared several CVD biomarkers among exclusive smokers, exclusive moist snuff consumers, and nonusers of tobacco (n = 48–60 in each condition) and found that the three BOPH that best differentiated the smokers from the two nonsmoking groups were the inflammatory biomarkers: soluble intercellular adhesion molecule (sICAM), IL-8, and IL-12(p70).47,48 However, no measurable differences in sICAM, IL-6, or IL-8 were detected among moist snuff users compared with nontobacco users, a finding that has also been observed in other studies.28,49

In a clinical multi-site switching study, 150 smokers were randomized to switch to a tobacco-heating cigarette (TH), snus (S), or an ultra-low machine yield tobacco-burning cigarette (TB) for 24 weeks with a comparison group of never smokers (n = 30) at baseline.50 At weeks 12 and 24 compared with baseline, significant reductions in sICAM were observed in all three product groups. In both weeks 12 and 24, researchers observed reductions in platelets (TH), increases in glycated hemoglobin (HbA1c; TH, S), and reductions in the F2-isoprostane biomarker (±)5-iPF –VI (TB). Thus, sICAM showed differences across product categories in the cross-sectional study with smokers showing higher levels of sICAM than ST users or nonusers of tobacco and sICAM was reduced when smokers were switched to the other tobacco products in the switching study. Similar results were observed for white blood cells and the F2-isoprostane biomarker iPF–III. Overall, the most consistent and significant improvements in biomarkers were found in smokers who switched to TH. Due to varied compliance across treatment groups, these findings reflect differences in poly-tobacco use rather than complete product switching.

These studies demonstrate the types of experiments that can be conducted to identify biomarkers that might be sensitive to differences across tobacco product categories and as compared with nonusers of tobacco. They also highlight how the biomarkers that show differences across tobacco type in cross-sectional studies are not necessarily the same biomarkers that show differences in switching studies.

Chronic Obstructive Pulmonary Disease

Many of the same components of tobacco smoke that lead to CVD also adversely affect the respiratory system, increasing the risk of COPD. Bronchitis and emphysema are the two most common underlying diseases of COPD.51 Tobacco smoke constituents cause lung injury through a number of mechanisms, including: impairment of the lung’s innate defense system, leading to potential for infection and inflammation (eg, acrolein); toxicity to the cilia or microscopic hairs along air passages (eg, acrolein, formaldehyde); lung irritation (eg, formaldehyde); oxidative damage (eg, nitrogen oxides, cadmium); and disruption of the oxidative metabolism of cells (eg, hydrogen cyanide).52

Forced Expiratory Volume in 1 Second

Forced expiratory volume in 1 second (FEV1) is a reproducible and well-established surrogate endpoint for COPD.53 Decreasing FEV1 is associated with increased COPD exacerbations, hospitalizations, and mortality.54 In a prospective cohort study of about 800 manual workers in London, the authors observed that FEV1 decreased over time for both smokers and nonsmokers, though at a steeper decline in smokers.55 Although smoking cessation did not recover lung function, the rate of FEV1 decline slowed to a normal level in former smokers. FEV1 has long been used to grade COPD and is strongly correlated with disease severity. However, patients with lung abnormalities may still have normal FEV1; thus, FEV1 is not considered to be highly sensitive to detection of disease.56 Additionally, changes in FEV1 may require several years to detect depending on factors such as study design, study size, and patient characteristics.53

Imaging Biomarkers

Some imaging tools, such as computed tomography (CT) scanning and magnetic resonance imaging (MRI), may have greater sensitivity for detecting emphysema and airway disease compared with FEV1.57,58 In the COPDGene cohort of approximately 10000 smokers followed over time, about 40% were considered controls based on FEV1 evaluation, and 60% had various grades of COPD at baseline.56 CT scans were taken at baseline as a type of micro-mapping of the lung, with individual pixels followed over time using a computer program. In a subset of 2300 study participants observed over 5 years, the CT scan detected progression of COPD in about 40% of the participants.56 Additionally, CT scanning provided evidence of airway disease and emphysema in 15% and 20%, respectively, of the smokers who were considered normal based on FEV1. Although preliminary findings suggest that imaging biomarkers may have greater sensitivity for detecting disease than spirometry, additional studies of smokers versus nonsmokers and smoking cessation could help strengthen their potential role in the tobacco regulatory setting.

Inflammatory Markers

The relationship between plasma inflammatory markers and poor clinical outcomes in COPD has been assessed. In the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints (ECLIPSE) cohort of COPD patients, persistent systemic inflammation based on white blood cell count, CRP, IL-6, TNF-alpha, fibrinogen, and IL-8 was associated with increased all-cause mortality and exacerbation.59 In another example, soluble receptor for advanced glycation end products (sRAGE) is thought to be anti-inflammatory by binding to damaged ligands and preventing them from activating cell signaling pathways. In a subset of participants enrolled in COPDGene cohort (n = 588), sRAGE was inversely associated with more severe emphysema.60 Similarly, in the ECLIPSE cohort of COPD patients (n = 1928), higher levels of sRAGE at baseline were associated with less emphysema and less disease progression over time.61

The relationship between markers of inflammation and smoking has also been assessed directly in the lung.62 Neutrophils measured in samples using bronchoaveolar lavage bronchoscopies have been observed to be higher in individuals with chronic bronchitis than asymptomatic smokers, but higher in asymptomatic smokers as compared with normal nonsmokers.63 In another study, the relative amount of M1 macrophages, which are pro-inflammatory, and M2 macrophages, which are anti-inflammatory, were observed to differ between nonsymptomatic smokers and nonsmokers.64

Cystic Fibrosis Transmembrane Conductance Regulator

Chronic bronchitis is characterized by sputum production and inflammation and shares similar symptoms with early cystic fibrosis (CF). Biomarkers for CF lung disease have recently been assessed for utility as BOPH. CF is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) ion channel that reduce activity, leading to airway surface dehydration.65 Tobacco exposure can also inhibit CFTR function, which further supports a potential role in COPD. In a study measuring the CFTR function in the nose using an electrode connected to a voltmeter, the authors observed reduced CFTR-mediated ion transport among nonsymptomatic smokers (n = 4) compared to nonsmokers (n = 12).66 This study also found that acute smoke exposure decreased CFTR ion transport activity for up to 3 h after cigarette smoke exposure.

Cancer

In the central mechanism of tobacco carcinogenesis, carcinogens in cigarette smoke are metabolically activated by cytochrome P450 enzymes and bind to DNA.52 The DNA adducts that are formed can cause miscoding, resulting in uncontrolled cell growth and leading to cancer development. Based on the initiation-promotion-progression mouse model originally developed by Berenblum and Shubik in the late 1940s, mouse experiments have shown that tumor development occurs only in the presence of both initiators that form mutations in cells (eg, benzo[a]pyrene, nitrosamine) and promoters that facilitate survival and growth of the mutated cells (eg, inflammatory agents such as lipopolysaccharide and smoke condensate).67,68

Inflammation and Oxidative Stress

Different lines of evidence support the role of inflammation in lung cancer. COPD, an inflammatory condition, is a risk factor for lung cancer.69 Use of aspirin and nonsteroidal anti-inflammatory drugs is associated with lower lung cancer risk.70 In prospective studies, inflammatory markers, including CRP, IL-6, and IL-8, have been associated with lung cancer risk.71 Another promising inflammatory marker is prostaglandin E2 metabolite (PGE-M), which is associated with a number of cancers including colorectal, lung, breast, and head and neck cancers; increased levels have been observed in smokers in a limited number of studies.72 As described previously, smoking causes oxidative damage. Although F2-isoprostane biomarkers can be reliably measured and are increased with smoking, their relationship with cancer risk is not yet established.

Biomarkers of Exposure (BOE) and DNA Adducts as BOPH

Several BOE, including 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanol (NNAL), N-nitrosonornicotine (NNN), and phenanthrene tetraol, are associated with subsequent cancer risk, as observed in the Shanghai Cohort Study, and thus seem to fulfill many of the criteria for BOPH (Table 2).73,74 For example, NNN causes esophageal tumors in rats which demonstrates biologic plausibility.75 In a nested case–control study, NNN was associated with a subsequent risk of esophageal cancer in a dose-dependent manner, demonstrating, temporality, predictivity, biological gradient, and coherence.74 Genetic variants have been observed to modify the effect of these biomarkers and cancer risk and may be important for stratification in study populations. For example, variation in the α5 nicotinic cholinergic receptor subunit gene (CHRNA5) on chromosome 15q25.1 is associated with smoking intensity and nicotine dependence as well as age at lung cancer diagnosis.76,77 Variants of CYP2A6 also modify nicotine and carcinogen uptake in smokers and are related to lung cancer risk.78 Finally, biomarkers such as DNA adducts play a central role in carcinogenesis, but challenges include low concentrations and nonquantitative methods. Recent developments for detecting DNA adducts in human oral cell DNA include use of nanospray ionization, which concentrates the sample, and quantification by high-resolution mass spectrometry.79

Airway Epithelial Biomarkers

Biomarkers investigated within the context of chemoprevention of lung cancer include bronchial epithelia dysplasia. Typically, biopsies taken from sites along the respiratory tract using bronchoscopy are examined for dysplasia based on a scoring system.80 In a clinical trial of oral iloprost (a prostacyclin analog) in current and former smokers, endobronchial histology was significantly worse in current smokers compared with former smokers at baseline.81 The persistence or progression of bronchial dysplasia may be predictive of lung cancer risk. In a small cohort of 188 participants at a high risk of pulmonary squamous cell carcinoma, having a higher frequency of sites that persisted or progressed to high-grade dysplasia was associated with a higher risk of invasive squamous cell carcinoma (hazards ratio = 7.84, 95% CI = 1.56 to 39.39).82 Less invasive methods are currently being investigated for use in lung cancer screening.83,84

Gene Expression

Smoking alters epithelial cell gene expression throughout the respiratory tract, and there is variability in the gene expression across individuals in response to damage from smoking and risk for developing lung cancer.85 Gene expression of the bronchial airway epithelium, collected using an endoscopic cytobrush, may serve as an early diagnostic biomarker for lung cancer.86 In the Airway Epithelium Gene Expression In the Diagnosis of Lung Cancer (AEGIS) clinical trial, investigators found that their 23 gene-expression classifier showed a higher sensitivity for detecting lung cancer, especially for small peripheral lung lesions (ranging from 90 to 92%), than bronchoscopy (ranging from 55% to 58%).86 In another study comparing non-symptomatic smokers and nonsmokers, active exposure to tobacco smoke altered the expression of 175 genes in the bronchial airway epithelium, and many of these changes reverted back to never smoker levels within a few months of quitting among former smokers. However, about 28 (16%) of the alterations in gene expression persisted after smoking cessation.87 Current studies are underway for detecting changes in gene expression using nasal epithelium samples with respect to smoking cessation and other clinical applications.88 Furthermore, in vitro experiments on bronchial epithelium cells showed that tobacco flavoring and the presence of nicotine induced most of the gene expression changes which may be relevant to ENDS use.85

Novel Biomarkers

Although more traditional approaches are based on a priori hypotheses, a less targeted approach has emerged with the development of high-throughput technologies and concurrent testing of massive numbers of markers, including “omics” biomarkers. Below are examples of studies of “omics” biomarkers as they relate to tobacco use.

Epigenome

Epigenetics involves the heritable changes in gene function that occur without a change in the DNA sequence, such as changes in microRNAs, polycomb proteins, histone modifications, and DNA methylation.89 Epigenetic changes can turn genes on or off and may lead to adverse health outcomes. For example, recent epigenome-wide studies observed that smoking was strongly associated with DNA methylation alterations of the aryl hydrocarbon receptor repressor (AHRR).90 A further study of AHRR methylation in current, former, and never smokers from the MESA cohort found that methylation of cg05575921 was most significantly associated with smoking status (current vs. never).90 Cg05575921 methylation also demonstrated reversibility with cessation, a dose-dependent relationship with smoking, and an association with carotid plaque scores. Another study of methylation profiling in buccal cells from smokers, moist snuff users, and nontobacco users observed that smokers exhibited a greater hypo-methylation pattern of methylation as compared with the moist snuff users and nontobacco users, including six out of seven loci of the AHRR gene that were hypo-methylated.91

Microbiome

The human body can be considered as an ecosystem where different microbiome patterns are present in different parts of the body. Recent investigations focused on whether the microbiome may mediate the adverse health effects of tobacco use. For example, a study conducted in China showed that current smokers appeared to have higher levels of unique microbial species and genera present in the upper gastrointestinal tract microbiome compared with never smokers whereas the levels of microbial species and genera in former smokers did not differ from those in never smokers.92 However, no significant associations between microbiome and pack-years of smoking (current smokers) or years since quitting (former smokers) were observed, though the analysis was based on a small sample. Overall, previous studies have shown inconsistent associations between the microbiome (eg, numbers of unique microbes) and current smoking, and future studies involving larger sample sizes and examining different types of tobacco use could further strengthen this area of research.93

Risk Assessment Using “Omics” Biomarkers

“Omics” biomarkers may enhance risk assessment at agencies such as the US Environmental Protection Agency (EPA) and The National Institute for Occupational Safety and Health (NIOSH). The development of an occupational exposure limit can be facilitated by a thorough understanding of comprehensive exposure assessments including use of “omics” biomarkers.94 As early biomarkers of effect, they may provide information on mode of action and dose–response relationship. However, challenges in using biomarkers of early effect in the risk assessment process include the fact that scientific tools for facilitating biomarker development, such as computational toxicology and systems biology, are still being developed. Additionally, few biomarkers have been validated in human populations, and appropriate interpretation of biomarker data is lacking.95

One tool that can facilitate the interpretation of biomarker data is the adverse outcome pathway (AOP), a conceptual framework developed by EPA scientists that connects molecular-level changes to adverse outcomes in a biological system. As high-throughput toxicity testing has become increasingly used for determining chemical toxicity (in addition to the more traditional animal testing), the AOP was developed to better interpret in vitro perturbations in terms of consequences to the whole organism as well as to identify the full set of toxicity pathways leading to all of the potential adverse outcomes in vivo.96 The AOP framework systematically organizes and summarizes data into the following components: molecular initiating event indicating initial chemical contact, key events such as changes in biological state, and adverse outcomes that are measurable and essential for adverse outcome progression. The advantage of the AOP framework is that it connects molecular initiating events to toxicity pathways without considering specific chemicals, unlike mode of action which is chemical-specific.97 As one potential application of this framework for tobacco evaluation, in vitro and clinical studies based on “omics” techniques could identify candidate biomarkers that differentiate exposure from nonexposure to cigarette smoke and then be screened based on their associations with the established adverse outcomes in order to serve as potential key events.

Future Directions

The deliberations from the workshop noted some promising BOPH but also highlighted the lack of systematic effort to identify BOPH that would have utility and validity for evaluating tobacco products. Because of the important role played by BOPH, a concerted effort would be useful to engage in multi-disciplinary, collaborative, and integrative biomarker research. Additionally, a common framework to guide this research would be helpful in accelerating progress. The following recommendations describe additional research that could further strengthen the utility of BOPH in the tobacco regulatory setting.

Utilize Current Knowledge of Critical and Common Pathways to Disease

Identifying BOPH based on our current knowledge of major smoking-mediated pathways to disease is a reasonable approach. Given that many of the smoking-related diseases share some of the same pathways, such as inflammation and oxidative stress, biomarkers that are relevant across disease areas (eg, CVD, COPD, cancer) may be especially useful to identify. How these biomarkers relate to each other, to tobacco use, and to each disease outcome separately may be important to investigate in future studies.

Explore Composite Biomarkers as Predictors of Disease Risk

For the major tobacco-related diseases (CVD, pulmonary disease, and cancer), a set of biomarkers, rather than a single biomarker, could better represent the multiple mechanisms by which tobacco causes these diseases. One possible approach is to improve disease risk prediction by combining multiple biomarkers into a composite variable. Specific considerations for testing composite biomarkers may include: pre-analytical considerations (eg, stability of individual biomarkers, sample collection times); analytical validation of all individual biomarker assays; factors influencing the variability of individual biomarker levels and the composite measure (eg, age, weight); weighting of individual biomarkers (ie, whether equal or unequal); and identifying a threshold score to define a significant change.98

Consider Novel Technologies

As some key events may be missed by focusing on known biomarkers and pathways, a more comprehensive and nontargeted approach based on novel technologies, including “omics” biomarkers, can be considered. Novel technologies can help identify new biomarkers that are less likely to be correlated with known biomarkers and lead to the development of a composite biomarker profile that is also specific for tobacco use. The main challenge is that few “omics” biomarkers have been fully validated with respect to reproducibility of results in human populations. Additionally, their roles in causal pathways and disease prediction have yet to be established.

Systematically Examine BOPH Using Existing Cohorts, Surveys, and Experimental Studies

BOPH can be further evaluated by leveraging existing studies with available biosamples or biomarker data, including cross-sectional, case-control, prospective cohorts (eg, MESA, Framingham study, Atherosclerosis Risk in Communities study [ARIC], Prostate, Lung, Colorectal and Ovarian [PLCO] Cancer Screening Trial, Population Assessment of Tobacco and Health [PATH] Study), intervention, and smoking cessation studies. In particular, prospective studies could help to establish temporality as well as to identify the magnitude of change needed to observe clinically meaningful changes. To date, most studies of biomarkers and tobacco have been conducted in cohorts of cigarette smokers. Thus, additional studies using existing cohorts of users of other established tobacco products, such as smokeless tobacco, cigar, and hookah, as well as cohorts of nonusers exposed to secondhand smoke, could further strengthen our understanding of how biomarkers relate to tobacco use/exposure and disease risk. Another consideration is focusing on special populations, including smokers at risk for the disease outcome (eg, genetics). A major challenge is that few BOPH are tobacco-specific, and approaches that could help distinguish effects of tobacco use from effects of other confounding factors are important to explore.

Supplementary Material

Supplementary data are available at Nicotine and Tobacco Research online

Funding

This work was supported by funds from the Food and Drug Administration.

Declaration of Interests

None declared.

Disclaimer

The findings and conclusions of this report are those of the authors and do not necessarily represent FDA positions or policies.

Acknowledgments

The authors thank the CTP Office of Science (OS) and the members of the OS Biomarker Workgroup, Aarthi Arab, Jiping Chen, Susan Chemerynski, Carolyn Dresler, Selvin Edwards, Dhanya John, Maocheng Yang, and Ling Yang for their contributions to the planning and execution of the BOPH Workshop.

References

1.

Family Smoking Prevention and Tobacco Control Act
.
2009
.
Pub L No. 111–31, 123 Stat 1776
.

2.

U.S. Food and Drug Administration
.
Deeming tobacco products to be subject to the Federal Food, Drug, and Cosmetic Act, as amended by the Family Smoking Prevention and Tobacco Control Act; restrictions on the sale and distribution of tobacco products and required warning statements for tobacco products
.
2016
. https://www.federalregister.gov/articles/2016/05/10/2016-10685/deeming-tobacco-products-to-be-subject-to-the-federal-food-drug-and-cosmetic-act-as-amended-by-the. Accessed December 19, 2017.

3.

Institute of Medicine
.
Clearing the Smoke: Assessing the Science Base for Tobacco Harm Reduction
.
Washington, DC
:
National Academy Press
;
2001
.

4.

U.S. Food and Drug Administration
.
Biomarkers of potential harm: a public workshop
.
2016
. https://wayback.archive-it.org/7993/20170404191030/https://www.fda.gov/TobaccoProducts/Newsroom/ucm481513.htm.
Accessed December 19, 2017
.

5.

Chang
CM
,
Edwards
SH
,
Arab
A
,
Del Valle-Pinero
AY
,
Yang
L
,
Hatsukami
DK
.
Biomarkers of tobacco exposure: summary of an fda-sponsored public workshop
.
Cancer Epidemiol Biomarkers Prev
.
2017
;
26
(
3
):
291
302
.

6.

Temple
R.
Surrogate markers at FDA – Lessons learned. Paper presented at the Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

7.

Temple
R
.
Are surrogate markers adequate to assess cardiovascular disease drugs
?
JAMA
.
1999
;
282
(
8
):
790
795
.

8.

Echt
DS
,
Liebson
PR
,
Mitchell
LB
et al. 
Mortality and morbidity in patients receiving encainide, flecainide, or placebo. The cardiac arrhythmia suppression trial
.
N Engl J Med
.
1991
;
324
(
12
):
781
788
.

9.

Fleming
TR
,
DeMets
DL
.
Surrogate end points in clinical trials: are we being misled
?
Ann Intern Med
.
1996
;
125
(
7
):
605
613
.

10.

Institute of Medicine
.
Evaluation of Biomarkers and Surrogate Endpoints in Chronic Disease
.
Washington, DC
:
The National Academies Press
;
2010
.

11.

Lassere
MN
.
The biomarker-surrogacy evaluation schema: a review of the biomarker-surrogate literature and a proposal for a criterion-based, quantitative, multidimensional hierarchical levels of evidence schema for evaluating the status of biomarkers as surrogate endpoints
.
Stat Methods Med Res
.
2008
;
17
(
3
):
303
340
.

12.

Shi
Q
,
Sargent
DJ
.
Meta-analysis for the evaluation of surrogate endpoints in cancer clinical trials
.
Int J Clin Oncol
.
2009
;
14
(
2
):
102
111
.

13.

HILL
AB
.
The environment and disease: association or causation
?
Proc R Soc Med
.
1965
;
58
:
295
300
.

14.

Institute of Medicine
.
Scientific Standards for Studies on Modified Risk Tobacco Products
.
Washington, DC
:
The National Academies Press
;
2012
.

15.

U.S. Department of Health and Human Services
.
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, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health
;
2010
.

16.

Vasan
RS
.
Biomarkers of cardiovascular disease: molecular basis and practical considerations
.
Circulation
.
2006
;
113
(
19
):
2335
2362
.

17.

Frost-Pineda
K
,
Liang
Q
,
Liu
J
et al. 
Biomarkers of potential harm among adult smokers and nonsmokers in the total exposure study
.
Nicotine Tob Res
.
2011
;
13
(
3
):
182
193
.

18.

MacMahon
S
,
Peto
R
,
Cutler
J
et al. 
Blood pressure, stroke, and coronary heart disease. Part 1, Prolonged differences in blood pressure: prospective observational studies corrected for the regression dilution bias
.
Lancet
.
1990
;
335
(
8692
):
765
774
.

19.

Mazzone
P
,
Tierney
W
,
Hossain
M
,
Puvenna
V
,
Janigro
D
,
Cucullo
L
.
Pathophysiological impact of cigarette smoke exposure on the cerebrovascular system with a focus on the blood-brain barrier: expanding the awareness of smoking toxicity in an underappreciated area
.
Int J Environ Res Public Health
.
2010
;
7
(
12
):
4111
4126
.

20.

Libby
P
,
Ridker
PM
,
Maseri
A
.
Inflammation and atherosclerosis
.
Circulation
.
2002
;
105
(
9
):
1135
1143
.

21.

Collaboration ERF
.
C-reactive protein, fibrinogen, and cardiovascular disease prediction
.
New Eng J Med
.
2012
;
367
(
14
):
1310
1320
.

22.

Danesh
J
,
Kaptoge
S
,
Mann
AG
et al. 
Long-term interleukin-6 levels and subsequent risk of coronary heart disease: two new prospective studies and a systematic review
.
PLoS Med
.
2008
;
5
(
4
):
e78
.

23.

Fibrinogen Studies Collaboration
.
Plasma fibrinogen level and the risk of major cardiovascular diseases and nonvascular mortality: an individual participant meta-analysis
.
JAMA
.
2005
;
294
(
14
):
1799
1809
.

24.

Ridker
PM
,
Hennekens
CH
,
Buring
JE
,
Rifai
N
.
C-reactive protein and other markers of inflammation in the prediction of cardiovascular disease in women
.
N Engl J Med
.
2000
;
342
(
12
):
836
843
.

25.

Barbieri
SS
,
Zacchi
E
,
Amadio
P
et al. 
Cytokines present in smokers’ serum interact with smoke components to enhance endothelial dysfunction
.
Cardiovasc Res
.
2011
;
90
(
3
):
475
483
.

26.

Csordas
A
,
Bernhard
D
.
The biology behind the atherothrombotic effects of cigarette smoke
.
Nat Rev Cardiol
.
2013
;
10
(
4
):
219
230
.

27.

Johnson
HM
,
Gossett
LK
,
Piper
ME
et al. 
Effects of smoking and smoking cessation on endothelial function: 1-year outcomes from a randomized clinical trial
.
J Am Coll Cardiol
.
2010
;
55
(
18
):
1988
1995
.

28.

Liu
J
,
Liang
Q
,
Frost-Pineda
K
et al. 
Relationship between biomarkers of cigarette smoke exposure and biomarkers of inflammation, oxidative stress, and platelet activation in adult cigarette smokers
.
Cancer Epidemiol Biomarkers Prev
.
2011
;
20
(
8
):
1760
1769
.

29.

Pasalic
L
,
Wang
SS
,
Chen
VM
.
Platelets as biomarkers of coronary artery disease
.
Semin Thromb Hemost
.
2016
;
42
(
3
):
223
233
.

30.

Wennmalm
A
,
Benthin
G
,
Granström
EF
,
Persson
L
,
Petersson
AS
,
Winell
S
.
Relation between tobacco use and urinary excretion of thromboxane A2 and prostacyclin metabolites in young men
.
Circulation
.
1991
;
83
(
5
):
1698
1704
.

31.

Benowitz
NL
,
Porchet
H
,
Sheiner
L
,
Jacob
P
III
.
Nicotine absorption and cardiovascular effects with smokeless tobacco use: comparison with cigarettes and nicotine gum
.
Clin Pharmacol Ther
.
1988
;
44
(
1
):
23
28
.

32.

Benowitz
NL
,
Kuyt
F
,
Jacob
P
III
.
Influence of nicotine on cardiovascular and hormonal effects of cigarette smoking
.
Clin Pharmacol Ther
.
1984
;
36
(
1
):
74
81
.

33.

Minami
J
,
Ishimitsu
T
,
Matsuoka
H
.
Effects of smoking cessation on blood pressure and heart rate variability in habitual smokers
.
Hypertension
.
1999
;
33
(
1 Pt 2
):
586
590
.

34.

Messner
B
,
Bernhard
D
.
Smoking and cardiovascular disease: mechanisms of endothelial dysfunction and early atherogenesis
.
Arterioscler Thromb Vasc Biol
.
2014
;
34
(
3
):
509
515
.

35.

Di Angelantonio
E
,
Chowdhury
R
,
Sarwar
N
et al. 
B-type natriuretic peptides and cardiovascular risk: systematic review and meta-analysis of 40 prospective studies
.
Circulation
.
2009
;
120
(
22
):
2177
2187
.

36.

Wang
TJ
,
Larson
MG
,
Levy
D
et al. 
Plasma natriuretic peptide levels and the risk of cardiovascular events and death
.
N Engl J Med
.
2004
;
350
(
7
):
655
663
.

37.

Otsuka
T
,
Kawada
T
,
Seino
Y
,
Ibuki
C
,
Katsumata
M
,
Kodani
E
.
Relation of smoking status to serum levels of N-terminal pro-brain natriuretic peptide in middle-aged men without overt cardiovascular disease
.
Am J Cardiol
.
2010
;
106
(
10
):
1456
1460
.

38.

Craig
WY
,
Palomaki
GE
,
Haddow
JE
.
Cigarette smoking and serum lipid and lipoprotein concentrations: an analysis of published data
.
BMJ
.
1989
;
298
(
6676
):
784
788
.

39.

Gepner
AD
,
Piper
ME
,
Johnson
HM
,
Fiore
MC
,
Baker
TB
,
Stein
JH
.
Effects of smoking and smoking cessation on lipids and lipoproteins: outcomes from a randomized clinical trial
.
Am Heart J
.
2011
;
161
(
1
):
145
151
.

40.

Willi
C
,
Bodenmann
P
,
Ghali
WA
,
Faris
PD
,
Cornuz
J
.
Active smoking and the risk of type 2 diabetes: a systematic review and meta-analysis
.
JAMA
.
2007
;
298
(
22
):
2654
2664
.

41.

Vlassopoulos
A
,
Lean
ME
,
Combet
E
.
Influence of smoking and diet on glycated haemoglobin and ‘pre-diabetes’ categorisation: a cross-sectional analysis
.
BMC Public Health
.
2013
;
13
:
1013
.

42.

Sandhu
RK
,
Jimenez
MC
,
Chiuve
SE
et al. 
Smoking, smoking cessation, and risk of sudden cardiac death in women
.
Circ Arrhythm Electrophysiol
.
2012
;
5
(
6
):
1091
1097
.

43.

Zevin
S
,
Jacob
P
III
,
Benowitz
NL
.
Dose-related cardiovascular and endocrine effects of transdermal nicotine
.
Clin Pharmacol Ther
.
1998
;
64
(
1
):
87
95
.

44.

Polonsky
TS
,
McClelland
RL
,
Jorgensen
NW
et al. 
Coronary artery calcium score and risk classification for coronary heart disease prediction
.
JAMA
.
2010
;
303
(
16
):
1610
1616
.

45.

McEvoy
JW
,
Nasir
K
,
DeFilippis
AP
et al. 
Relationship of cigarette smoking with inflammation and subclinical vascular disease: the Multi-Ethnic Study of Atherosclerosis
.
Arterioscler Thromb Vasc Biol
.
2015
;
35
(
4
):
1002
1010
.

46.

Schmermund
A
,
Achenbach
S
,
Budde
T
et al. 
Effect of intensive versus standard lipid-lowering treatment with atorvastatin on the progression of calcified coronary atherosclerosis over 12 months: a multicenter, randomized, double-blind trial
.
Circulation
.
2006
;
113
(
3
):
427
437
.

47.

Campbell
LR
,
Brown
BG
,
Jones
BA
,
Marano
KM
,
Borgerding
MF
.
Study of cardiovascular disease biomarkers among tobacco consumers, part 1: biomarkers of exposure
.
Inhal Toxicol
.
2015
;
27
(
3
):
149
156
.

48.

Nordskog
BK
,
Brown
BG
,
Marano
KM
,
Campell
LR
,
Jones
BA
,
Borgerding
MF
.
Study of cardiovascular disease biomarkers among tobacco consumers, part 2: biomarkers of biological effect
.
Inhal Toxicol
.
2015
;
27
(
3
):
157
166
.

49.

Lip
GY
,
Blann
A
.
von Willebrand factor: a marker of endothelial dysfunction in vascular disorders
?
Cardiovasc Res
.
1997
;
34
(
2
):
255
265
.

50.

Ogden
MW
,
Marano
KM
,
Jones
BA
,
Morgan
WT
,
Stiles
MF
.
Switching from usual brand cigarettes to a tobacco-heating cigarette or snus: Part 3. Biomarkers of biological effect
.
Biomarkers
.
2015
;
20
(
6-7
):
404
410
.

51.

Barr
RG.
Imaging Biomarkers of Potential Harm: CT and MRI. Paper Presented at the Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

52.

U.S. Department of Health and Human Services
.
The Health Consequences of Smoking -- 50 Years of Progress: a Report of the Surgeon General
.
Atlanta, GA
:
U.S. Department of Health and Human Services, Centers for Disease Control and Prevention, National Center for Chronic Disease Prevention and Health Promotion, Office on Smoking and Health
;
2014
.

53.

Torjusen
E.
Forced Expiratory Volume in 1 Second: a Multifunctional Biomarker of Lung Function. Paper Presented at: Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

54.

Pompeo
F
,
Brooke
E
,
Kawamura
A
,
Mushtaq
A
,
Sim
E
.
The pharmacogenetics of NAT: structural aspects
.
Pharmacogenomics
.
2002
;
3
(
1
):
19
30
.

55.

Fletcher
C
,
Peto
R
.
The natural history of chronic airflow obstruction
.
Br Med J
.
1977
;
1
(
6077
):
1645
1648
.

56.

Crapo
JD.
Tobacco use and COPD: Overview. Paper Presented at Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

57.

Galbán
CJ
,
Han
MK
,
Boes
JL
et al. 
Computed tomography-based biomarker provides unique signature for diagnosis of COPD phenotypes and disease progression
.
Nat Med
.
2012
;
18
(
11
):
1711
1715
.

58.

Kirby
M
,
Parraga
G
.
COPD imaging: new tools to tackle an old problem
?
COPD
.
2014
;
11
(
5
):
477
479
.

59.

Agusti
A
,
Edwards
LD
,
Rennard
SI
et al. 
Persistent systemic inflammation is associated with poor clinical outcomes in COPD: a novel phenotype
.
PLoS One
.
2012
;
7
(
5
):
e37483
.

60.

Carolan
BJ
,
Hughes
G
,
Morrow
J
et al. 
The association of plasma biomarkers with computed tomography-assessed emphysema phenotypes
.
Respir Res
.
2014
;
15
:
127
.

61.

Coxson
HO
,
Dirksen
A
,
Edwards
LD
et al. 
The presence and progression of emphysema in COPD as determined by CT scanning and biomarker expression: a prospective analysis from the ECLIPSE study
.
Lancet Respir Med
.
2013
;
1
(
2
):
129
136
.

62.

Hatsukami
DK
,
Benowitz
NL
,
Rennard
SI
,
Oncken
C
,
Hecht
SS
.
Biomarkers to assess the utility of potential reduced exposure tobacco products
.
Nicotine Tob Res
.
2006
;
8
(
4
):
600
622
.

63.

Thompson
AB
,
Daughton
D
,
Robbins
RA
,
Ghafouri
MA
,
Oehlerking
M
,
Rennard
SI
.
Intraluminal airway inflammation in chronic bronchitis. Characterization and correlation with clinical parameters
.
Am Rev Respir Dis
.
1989
;
140
(
6
):
1527
1537
.

64.

Tarran
R.
Tobacco Exposure-Induced Biomarkers of Harm in Human Airway Epithelia. Paper Presented at: Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

65.

Ghosh
A
,
Boucher
RC
,
Tarran
R
.
Airway hydration and COPD
.
Cell Mol Life Sci
.
2015
;
72
(
19
):
3637
3652
.

66.

Clunes
LA
,
Davies
CM
,
Coakley
RD
et al. 
Cigarette smoke exposure induces CFTR internalization and insolubility, leading to airway surface liquid dehydration
.
FASEB J
.
2012
;
26
(
2
):
533
545
.

67.

Hoffmann
D
,
Schmeltz
I
,
Hecht
S
,
Wynder
R
,
Ts’o
P
,
Gelboin
H.
Polycyclic hydrocarbons and cancer
.
New York, NY
:
Academic Press
;
1978
.

68.

Melkamu
T
,
Qian
X
,
Upadhyaya
P
,
O’Sullivan
MG
,
Kassie
F
.
Lipopolysaccharide enhances mouse lung tumorigenesis: a model for inflammation-driven lung cancer
.
Vet Pathol
.
2013
;
50
(
5
):
895
902
.

69.

Durham
AL
,
Adcock
IM
.
The relationship between COPD and lung cancer
.
Lung Cancer
.
2015
;
90
(
2
):
121
127
.

70.

Smith
CJ
,
Perfetti
TA
,
King
JA
.
Perspectives on pulmonary inflammation and lung cancer risk in cigarette smokers
.
Inhal Toxicol
.
2006
;
18
(
9
):
667
677
.

71.

Brenner
DR
,
Fanidi
A
,
Grankvist
K
et al. 
Inflammatory cytokines and lung cancer risk in 3 prospective studies
.
Am J Epidemiol
.
2017
;
185
(
2
):
86
95

72.

Duffield-Lillico
AJ
,
Boyle
JO
,
Zhou
XK
et al. 
Levels of prostaglandin E metabolite and leukotriene E(4) are increased in the urine of smokers: evidence that celecoxib shunts arachidonic acid into the 5-lipoxygenase pathway
.
Cancer Prev Res (Phila)
.
2009
;
2
(
4
):
322
329
.

73.

Yuan
JM
,
Gao
YT
,
Murphy
SE
et al. 
Urinary levels of cigarette smoke constituent metabolites are prospectively associated with lung cancer development in smokers
.
Cancer Res
.
2011
;
71
(
21
):
6749
6757
.

74.

Yuan
JM
,
Knezevich
AD
,
Wang
R
,
Gao
YT
,
Hecht
SS
,
Stepanov
I
.
Urinary levels of the tobacco-specific carcinogen N’-nitrosonornicotine and its glucuronide are strongly associated with esophageal cancer risk in smokers
.
Carcinogenesis
.
2011
;
32
(
9
):
1366
1371
.

75.

Stepanov
I
,
Sebero
E
,
Wang
R
,
Gao
YT
,
Hecht
SS
,
Yuan
JM
.
Tobacco-specific N-nitrosamine exposures and cancer risk in the Shanghai Cohort Study: remarkable coherence with rat tumor sites
.
Int J Cancer
.
2014
;
134
(
10
):
2278
2283
.

76.

Chen
LS
,
Hung
RJ
,
Baker
T
et al. 
CHRNA5 risk variant predicts delayed smoking cessation and earlier lung cancer diagnosis--a meta-analysis
.
J Natl Cancer Inst
.
2015
;
107
(
5
):pii:
djv100
.

77.

Le Marchand
L
,
Derby
KS
,
Murphy
SE
et al. 
Smokers with the CHRNA lung cancer-associated variants are exposed to higher levels of nicotine equivalents and a carcinogenic tobacco-specific nitrosamine
.
Cancer Res
.
2008
;
68
(
22
):
9137
9140
.

78.

Yuan
JM
,
Nelson
HH
,
Butler
LM
et al. 
Genetic determinants of cytochrome P450 2A6 activity and biomarkers of tobacco smoke exposure in relation to risk of lung cancer development in the Shanghai cohort study
.
Int J Cancer
.
2016
;
138
(
9
):
2161
2171
.

79.

Hecht
SS.
Biomarkers of Potential Harm Associated With Cigarette Smoking. Paper Presented at the Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

80.

Travis
W.
D.
,
Brambilla
E.
,
Burke
A.
P.
,
Marx
A.
,
Nicholson
A.
G
.
WHO classification of tumours of the lung, pleara, thymus and heart
, 4th Edition.
Lyon
:
IARC
;
2015
.

81.

Keith
RL
,
Blatchford
PJ
,
Kittelson
J
et al. 
Oral iloprost improves endobronchial dysplasia in former smokers
.
Cancer Prev Res (Phila)
.
2011
;
4
(
6
):
793
802
.

82.

Merrick
DT
,
Gao
D
,
Miller
YE
et al. 
Persistence of bronchial dysplasia is associated with development of invasive squamous cell carcinoma
.
Cancer Prev Res (Phila)
.
2016
;
9
(
1
):
96
104
.

83.

Meyer
MG
,
Hayenga
JW
,
Neumann
T
et al. 
The Cell-CT 3-dimensional cell imaging technology platform enables the detection of lung cancer using the noninvasive LuCED sputum test
.
Cancer Cytopathol
.
2015
;
123
(
9
):
512
523
.

84.

Wilbur
DC
,
Meyer
MG
,
Presley
C
et al. 
Automated 3-dimensional morphologic analysis of sputum specimens for lung cancer detection: Performance characteristics support use in lung cancer screening
.
Cancer Cytopathol
.
2015
;
123
(
9
):
548
556
.

85.

Spira
A
,
Beane
J
,
Shah
V
et al. 
Effects of cigarette smoke on the human airway epithelial cell transcriptome
.
Proc Natl Acad Sci U S A
.
2004
;
101
(
27
):
10143
10148
.

86.

Silvestri
GA
,
Vachani
A
,
Whitney
D
et al. 
A bronchial genomic classifier for the diagnostic evaluation of lung cancer
.
New Eng J Med
.
2015
;
373
(
3
):
243
251
.

87.

Beane
J
,
Sebastiani
P
,
Liu
G
,
Brody
JS
,
Lenburg
ME
,
Spira
A
.
Reversible and permanent effects of tobacco smoke exposure on airway epithelial gene expression
.
Genome Biology
.
2007
;
8
(
9
):
1
17
.

88.

Zhang
X
,
Sebastiani
P
,
Liu
G
et al. 
Similarities and differences between smoking-related gene expression in nasal and bronchial epithelium
.
Physiol Genomics
.
2010
;
41
(
1
):
1
8
.

89.

Clark
SJ
,
Statham
A
,
Stirzaker
C
,
Molloy
PL
,
Frommer
M
.
DNA methylation: bisulphite modification and analysis
.
Nat Protoc
.
2006
;
1
(
5
):
2353
2364
.

90.

Reynolds
LM
,
Wan
M
,
Ding
J
et al. 
DNA methylation of the aryl hydrocarbon receptor repressor associations with cigarette smoking and subclinical atherosclerosis
.
Circ Cardiovasc Genet
.
2015
;
8
(
5
):
707
716
.

91.

Prasad
GL.
Biomarkers of Potential harm: Tools to Differentiate Tobacco Product Classes. Paper Presented at the Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

92.

Vogtmann
E
,
Flores
R
,
Yu
G
et al. 
Association between tobacco use and the upper gastrointestinal microbiome among Chinese men
.
Cancer Causes Control
.
2015
;
26
(
4
):
581
588
.

93.

Vogtmann
E.
Tobacco use and the human microbiome. Paper presented at the Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

94.

DeBord
DG
,
Burgoon
L
,
Edwards
SW
et al. 
Systems biology and biomarkers of early effects for occupational exposure limit setting
.
J Occup Environ Hyg
.
2015
;
12
(
Suppl 1
):
S41
S54
.

95.

DeBord
DG.
Use of Biomarkers of Early Effect in Occupational Exposure Limit Setting. Paper Presented at the Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

96.

Edwards
SW
,
Tan
YM
,
Villeneuve
DL
,
Meek
ME
,
McQueen
CA
.
Adverse Outcome Pathways-Organizing Toxicological Information to Improve Decision Making
.
J Pharmacol Exp Ther
.
2016
;
356
(
1
):
170
181
.

97.

Ankley
GT
,
Bennett
RS
,
Erickson
RJ
et al. 
Adverse outcome pathways: a conceptual framework to support ecotoxicology research and risk assessment
.
Environ Toxicol Chem
.
2010
;
29
(
3
):
730
741
.

98.

Amur
SG.
Biomarker qualification at CDER, FDA. Paper presented at the Center for Tobacco Products’ Biomarkers of Potential Harm Workshop
;
April 4–5
,
2016
;
Silver Spring, MD
.

99.

U.S. Department of Health and Human Services
.
BEST (Biomarkers, EndpointS, and other Tools) Resource
.
2016
; http://www.ncbi.nlm.nih.gov/books/NBK326791/.
Accessed December 19, 2017
.

100.

U.S. Environmental Protection Agency
.
Defining Pesticide Biomarkers
.
2016
; https://www.epa.gov/pesticide-science-and-assessing-pesticide-risks/defining-pesticide-biomarkers.
Accessed December 19, 2017
.

This work is written by (a) US Government employee(s) and is in the public domain in the US.

Supplementary data

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.