Skip to main content
Advertisement
  • Loading metrics

Current and Former Smoking and Risk for Venous Thromboembolism: A Systematic Review and Meta-Analysis

  • Yun-Jiu Cheng ,

    Contributed equally to this work with: Yun-Jiu Cheng, Zhi-Hao Liu

    Affiliation Department of Cardiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China

  • Zhi-Hao Liu ,

    Contributed equally to this work with: Yun-Jiu Cheng, Zhi-Hao Liu

    Affiliation Department of Gastroenterology, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China

  • Feng-Juan Yao,

    Affiliation Department of Ultrasonography, the First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China

  • Wu-Tao Zeng,

    Affiliation Department of Cardiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China

  • Dong-Dan Zheng,

    Affiliation Department of Cardiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China

  • Yu-Gang Dong,

    Affiliation Department of Cardiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China

  • Su-Hua Wu

    *wusuhua@hotmail.com

    Affiliation Department of Cardiology, First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China

Abstract

Background

Smoking is a well-established risk factor for atherosclerotic disease, but its role as an independent risk factor for venous thromboembolism (VTE) remains controversial. We conducted a meta-analysis to summarize all published prospective studies and case-control studies to update the risk for VTE in smokers and determine whether a dose–response relationship exists.

Methods and Findings

We performed a literature search using MEDLINE (source PubMed, January 1, 1966 to June 15, 2013) and EMBASE (January 1, 1980 to June 15, 2013) with no restrictions. Pooled effect estimates were obtained by using random-effects meta-analysis. Thirty-two observational studies involving 3,966,184 participants and 35,151 VTE events were identified. Compared with never smokers, the overall combined relative risks (RRs) for developing VTE were 1.17 (95% CI 1.09–1.25) for ever smokers, 1.23 (95% CI 1.14–1.33) for current smokers, and 1.10 (95% CI 1.03–1.17) for former smokers, respectively. The risk increased by 10.2% (95% CI 8.6%–11.8%) for every additional ten cigarettes per day smoked or by 6.1% (95% CI 3.8%–8.5%) for every additional ten pack-years. Analysis of 13 studies adjusted for body mass index (BMI) yielded a relatively higher RR (1.30; 95% CI 1.24–1.37) for current smokers. The population attributable fractions of VTE were 8.7% (95% CI 4.8%–12.3%) for ever smoking, 5.8% (95% CI 3.6%–8.2%) for current smoking, and 2.7% (95% CI 0.8%–4.5%) for former smoking. Smoking was associated with an absolute risk increase of 24.3 (95% CI 15.4–26.7) cases per 100,000 person-years.

Conclusions

Cigarette smoking is associated with a slightly increased risk for VTE. BMI appears to be a confounding factor in the risk estimates. The relationship between VTE and smoking has clinical relevance with respect to individual screening, risk factor modification, and the primary and secondary prevention of VTE.

Please see later in the article for the Editors' Summary

Editors' Summary

Background

Blood normally flows throughout the human body, supplying its organs and tissues with oxygen and nutrients. But, when an injury occurs, proteins called clotting factors make the blood gel (coagulate) at the injury site. The resultant clot (thrombus) plugs the wound and prevents blood loss. Occasionally, a thrombus forms inside an uninjured blood vessel and partly or completely blocks the blood flow. Clot formation inside one of the veins deep within the body, usually in a leg, is called deep vein thrombosis (DVT) and can cause pain, swelling, and redness in the affected limb. DVT can be treated with drugs that stop the blood clot from getting larger (anticoagulants) but, if left untreated, part of the clot can break off and travel to the lungs, where it can cause a life-threatening pulmonary embolism. DVT and pulmonary embolism are collectively known as venous thromboembolism (VTE). Risk factors for VTE include having an inherited blood clotting disorder, oral contraceptive use, prolonged inactivity (for example, during a long-haul plane flight), and having surgery. VTEs are present in about a third of all people who die in hospital and, in non-bedridden populations, about 10% of people die within 28 days of a first VTE event.

Why Was This Study Done?

Some but not all studies have reported that smoking is also a risk factor for VTE. A clear demonstration of a significant association (a relationship unlikely to have occurred by chance) between smoking and VTE might help to reduce the burden of VTE because smoking can potentially be reduced by encouraging individuals to quit smoking and through taxation policies and other measures designed to reduce tobacco consumption. In this systematic review and meta-analysis, the researchers examine the link between smoking and the risk of VTE in the general population and investigate whether heavy smokers have a higher risk of VTE than light smokers. A systematic review uses predefined criteria to identify all the research on a given topic; meta-analysis is a statistical method for combining the results of several studies.

What Did the Researchers Do and Find?

The researchers identified 32 observational studies (investigations that record a population's baseline characteristics and subsequent disease development) that provided data on smoking and VTE. Together, the studies involved nearly 4 million participants and recorded 35,151 VTE events. Compared with never smokers, ever smokers (current and former smokers combined) had a relative risk (RR) of developing VTE of 1.17. That is, ever smokers were 17% more likely to develop VTE than never smokers. For current smokers and former smokers, RRs were 1.23 and 1.10, respectively. Analysis of only studies that adjusted for body mass index (a measure of body fat and a known risk factor for conditions that affect the heart and circulation) yielded a slightly higher RR (1.30) for current smokers compared with never smokers. For ever smokers, the population attributable fraction (the proportional reduction in VTE that would accrue in the population if no one smoked) was 8.7%. Notably, the risk of VTE increased by 10.2% for every additional ten cigarettes smoked per day and by 6.1% for every additional ten pack-years. Thus, an individual who smoked one pack of cigarettes per day for 40 years had a 26.7% higher risk of developing VTE than someone who had never smoked. Finally, smoking was associated with an absolute risk increase of 24.3 cases of VTE per 100,000 person-years.

What Do These Findings Mean?

These findings indicate that cigarette smoking is associated with a statistically significant, slightly increased risk for VTE among the general population and reveal a dose-relationship between smoking and VTE risk. They cannot prove that smoking causes VTE—people who smoke may share other unknown characteristics (confounding factors) that are actually responsible for their increased risk of VTE. Indeed, these findings identify body mass index as a potential confounding factor that might affect the accuracy of estimates of the association between smoking and VTE risk. Although the risk of VTE associated with smoking is smaller than the risk associated with some well-established VTE risk factors, smoking is more common (globally, there are 1.1 billion smokers) and may act synergistically with some of these risk factors. Thus, smoking behavior should be considered when screening individuals for VTE and in the prevention of first and subsequent VTE events.

Additional Information

Please access these Web sites via the online version of this summary at http://dx.doi.org/10.1371/journal.pmed.1001515.

  • The US National Heart Lung and Blood Institute provides information on deep vein thrombosis (including an animation about how DVT causes pulmonary embolism), and information on pulmonary embolism
  • The UK National Health Service Choices website has information on deep vein thrombosis, including personal stories, and on pulmonary embolism; SmokeFree is a website provided by the UK National Health Service that offers advice on quitting smoking
  • The non-profit organization US National Blood Clot Alliance provides detailed information about deep vein thrombosis and pulmonary embolism for patients and professionals and includes a selection of personal stories about these conditions
  • The World Health Organization provides information about the dangers of tobacco (in several languages)
  • Smokefree.gov, from the US National Cancer Institute, offers online tools and resources to help people quit smoking
  • MedlinePlus has links to further information about deep vein thrombosis, pulmonary embolism, and the dangers of smoking (in English and Spanish)

Introduction

Although cigarette smoking has been responsible for approximately 5 million deaths every year, there are still an estimated 1.1 billion smokers worldwide [1],[2]. The magnitude of this public health challenge is growing, and estimates suggest that as many as 8 million people may die from smoking-related diseases by 2030 [2]. Venous thromboembolism (VTE) is a serious medical event and associated with a substantial risk of mortality [3]. In ambulatory population-based cohorts, the estimated 28-d mortality for the first episode of VTE is 11% [4]. Autopsy studies have found that VTE exists in about one-third of deaths in hospitals and 13% of all autopsies showed signs of fatal pulmonary embolism (PE) [5],[6].

Smoking is a well-established risk factor for atherosclerotic disease, but its role as an independent risk factor or effect modifier for VTE remains controversial. Several prospective studies reported smoking to be an independent risk factor [7],[8], whereas others failed to detect a significant relationship between smoking and VTE [9],[10]. A recent meta-analysis showed a statistically nonsignificant odds ratio (OR) for VTE of 1.18 (95% CI 0.95–1.46) for smokers compared with non-smokers [11]. However, the meta-analysis (involving a total of ten studies) included only about one-third of the data currently available. In addition, six of the ten studies included were clinical trials of oral contraceptives, in which the samples may not be representative of the general population. Furthermore, the VTE risk may be underestimated due to lack of distinction between former and current smokers and no adjustment for cardiovascular risk factors.

Smoking can be potentially reduced by individual and population-related measures; therefore, demonstrating the link between smoking and the risk of VTE may help reduce the burden of this disease. Therefore, we conducted a meta-analysis with the following aims: (1) to estimate the link between smoking and risk of VTE in the general population; (2) to measure the smoking-VTE relationship according to different degrees of adjustment for confounding factors, study designs, study populations, sex category, and type of VTE; and (3) to study dose-response patterns of tobacco exposure on the risk of VTE.

Methods

Search Strategy

This meta-analysis follows PRISMA guidelines (Text S1). We searched the publications listed in the electronic databases MEDLINE (source PubMed, January 1, 1966 to June 15, 2013) and EMBASE (January 1, 1980 to June 15, 2013) using the following text and key words in combination both as MeSH terms and text word “thromboembolism”, “venous thrombosis”, “pulmonary embolism”, deep-vein thrombosis”, “risk factors”, “smoke”, “cigarette”, “tobacco” or “smoking”. We searched articles published in any language and scrutinized references from these studies to identify other relevant studies.

Study Selection

To minimize differences between studies, we imposed the following methodological restrictions for the inclusion criteria: (1) Studies that contained the minimum information necessary to estimate the relative risk (RR) associated with smoking, including case-control and cohort studies published as original articles; (2) Studies in which populations were representative of the general population and not those with selected participants on the basis of risk factor for VTE, such as tumor, surgery, or use of oral contraceptives. In instances of multiple publications, the most up-to-date or comprehensive information was used.

Data Abstraction

Articles were reviewed and cross-checked independently by two authors (YJC and ZHL). Because there is no standardized quality scoring system for observational studies, we selected a priori several important design characteristics that may affect study quality, including method of case confirmation, percentage of patients completing planned follow-up, smoking as the primary analysis of interest, selection criteria for control participants, matching criteria, and control for confounding. Percentage agreement between the two authors on the quality review ranged from 88% to 100%. Any disagreements were resolved by consensus. Data on the following characteristics were independently extracted: study size, number of patients who developed VTE, total person-years of follow-up, study population, publication year, study design, sampling framework, study location (defined as Europe, North America, or Asia); gender category, site of VTE studied (deep vein thrombosis [DVT] or PE), type of VTE studied (unprovoked or provoked), ascertainment of VTE (validated or not validated), smoking category (ever, current, or former), and reported adjustment for potential confounders. When available, we used the most comprehensively adjusted risk estimates.

Data Analysis

RR was used as a measure of the relationship between smoking and the risk of VTE. For case-control studies, the OR was used as a surrogate measure of the corresponding RR. Because the absolute risk of VTE is low, the OR approximates the RR [12].

Summary RRs (95% CI) were calculated by pooling the study-specific estimates using a random-effects model that included between-study heterogeneity (parallel analyses used fixed-effects models), because significant heterogeneity was anticipated among studies. Pooled RRs were expressed with 95% CIs. We calculated the I2 (95% CI) statistic to assess heterogeneity across studies, applying the following interpretation for I2: <50% = low heterogeneity; 50%–75% = moderate heterogeneity; >75% = high heterogeneity [13].

We calculated the population attributable fraction (PAF) as {prevalence of smoking×(RR−1)/[prevalence of smoking×(RR−1)+1]}, where RR indicates pooled RRs [14]. On the basis of population-based cohort studies, the average prevalence of three categories of smoking was estimated by weighting by the sample size of each study.

Subgroup analyses and meta-regression models were carried out to investigate potential sources of between-study heterogeneity. When several risk estimates were present in a single study (i.e., separate estimates for current and former smokers), we adjusted the pooled estimates for intra-study or within-study correlation [15].

In the dose-response analysis, we considered cigarettes per day and pack-years as explanatory variables. Because for many studies continuous exposures were reported as categorical data with a range, we assigned the mid-point in each category to the corresponding RR for each study. When the highest category was open ended, we considered 60 cigarettes per day and 60 pack-years as the maximum (for example, one study reported >20 cigarettes per day as an open range; we considered 40 cigarettes per day as the mid-point in this category). We used generalized least squares (GLST) regression models to assess the pooled dose-response relation between smoking and risk of VTE across studies that had heterogeneous categorizations of smoking [16]. Linear models were fitted and evaluated on the logarithm of the RRs.

To enable the total person-years of observation to be calculated, we included data from reports that specified one or more of the following: total person-time of follow-up; sample size and mean (or median) follow-up per patient; or sample size and cumulative incidence rate. The principal summary measure was event rate expressed per 100,000 patient-years of follow-up. Weighted meta-analytic prevalence estimates for outcomes were calculated with the variance-stabilizing Freeman-Tukey double-arcsine transformation with an inverse-variance random-effects model [17].

Small study bias, consistent with publication bias, was assessed with funnel plot, by Begg's adjusted rank correlation test and by Egger's regression asymmetry test [18]. We used STATA, version 11.0 (Stata Corp) for all analyses. Statistical tests were two sided and used a significance level of p<0.05.

Results

Study Selection

With the search strategy, 1,531 unique citations were initially retrieved. Of these, 231 articles were considered of interest and full text was retrieved for detailed evaluation. One hundred ninety-nine of these 231 articles were subsequently excluded and finally 32 articles were included in the meta-analysis (Figure 1).

thumbnail
Figure 1. Flowchart of the selection of studies included in meta-analysis.

https://doi.org/10.1371/journal.pmed.1001515.g001

Study Characteristics

Thirty-two independent observational studies reporting 3,966,184 individuals and 35,151 incident cases were identified [7],[8],[19][48]. Fifteen studies were based in Europe, eight in North America, and nine in Asia. No studies were based in Africa or South America. Studies were published between January 1980 and March 2013. Thirteen studies were prospective cohort studies and 19 were case-control studies. 15 studies recruited participants from population registers and 15 were hospital-based.

The methodological quality of the included studies was generally good. Of the primary studies, 100% had described independent, consecutive sampling of their cohort. Average follow-up duration ranged from 5.0 to 20.1 y. Patients were followed up for an average of over 10 y in a majority of studies (84.6%). The proportion of patients with complete follow-up to the end of the study was given for 11 studies and ranged from 70.5% to >99%. The sizes of the cohorts ranged from 855 to 2,314,701 (in total 3,926,048), with the two largest studies recruiting participants over 1 million (Table 1) [26],[29]. Nineteen case-control studies were designed to evaluate risk factors for VTE, and eight of them used either hospital discharge data or data from registries. In 12 of the 19 incident case-control studies, controls were matched for age and/or sex only (Table 2).

thumbnail
Table 1. Cohort studies reporting incidence risk estimates.

https://doi.org/10.1371/journal.pmed.1001515.t001

thumbnail
Table 2. Case-control studies reporting incidence risk estimates.

https://doi.org/10.1371/journal.pmed.1001515.t002

Of all the studies, two included only patients with DVT [36],[44] and four investigated only patients with PE [19],[31],[35],[40]. Four cohort studies [8],[22],[27],[28] and four case-control studies [33],[38],[39],[48] compared the prevalence of smoking between patients with unprovoked VTE and provoked VTE. Eight studies investigated only women [19],[23],[25],[29],[30],[32],[34],[48] and three studies included only men [20],[22],[24]. The association between smoking and VTE was the primary outcome of interest for 20 studies, whereas it was a secondary question in 12 studies. The ascertainment of VTE varied across studies; 24 studies based on medical record, radiology or autopsy (validated), and eight confirmed by questionnaire or patient registry (not validated) (Tables 1 and 2).

Adjusted RRs could be determined for all cohort studies and nine of the case-control studies. Most risk estimates were adjusted for age (19 studies) and sex (11 studies). Eighteen studies (56.3%) reported an adjusted estimate for at least one of the cardiovascular risk factors: BMI (11 cohort and seven case-control studies), cholesterol (three cohort and one case-control studies), diabetes (three cohort and three case-control studies), hypertension (four cohort and four case-control studies), alcohol consumption (four cohort studies), or physical activity (four cohort studies). Detailed information on adjustments is reported in Tables 1 and 2.

Smoking and Risk of VTE

Figures 2, S1, and S2 showed the results from the random-effects model (parallel analysis with fixed-effects model) combining the RRs for VTE. Overall, the ever smokers compared with the reference group experienced a significantly increased risk for developing VTE (RR: 1.17 [95% CI 1.09–1.25, p<0.001]). The pooled RRs for current versus never smokers and former versus never smokers were 1.23 (95% CI 1.14–1.33, p<0.001) and 1.10 (95% CI 1.03–1.17, p = 0.002), respectively.

thumbnail
Figure 2. Forest plot for VTE incidence: risk estimates for ever versus never smokers.

The size of each square is proportional to the study's weight (inverse of variance).

https://doi.org/10.1371/journal.pmed.1001515.g002

There was evidence of moderate heterogeneity of RRs across these studies. The findings from the sensitivity analyses based on different inclusion and exclusion criteria were presented in Table 3. Risk estimates changed little after analyses with fixed effects models, inclusion of the studies with adjusted RRs, or exclusion of the two largest and the outlier studies, yet moderate heterogeneity was still present. However, when the analysis was confined to those large prospective cohort studies (high quality), the overall combined RR did not materially change, but heterogeneity was decreased to 34.68% for ever smokers, 10.61% for current smokers, and 0% for former smokers.

thumbnail
Table 3. Sensitivity and heterogeneity analysis of pooled relative risks of VTE for smokers.

https://doi.org/10.1371/journal.pmed.1001515.t003

Because the study by Ray et al. [32] only reported risk estimates for current but not for former smokers, in order to allow an unbiased comparison between the two smoking classes, we also computed the RR for current smokers (RR: 1.25 [95% CI 1.17–1.35]) from the remaining 14 studies reporting both estimates. Compared with former smokers, current smokers experienced a significant higher risk for developing VTE (p = 0.02). Neither funnel plots nor Egger and Begg tests showed evidence of publication bias for ever smokers (Egger, p = 0.88; Begg, p = 0.21), current smokers (Egger, p = 0.06; Begg, p = 0.11), and former smokers (Egger, p = 0.41; Begg, p = 0.83) (Figure 3).

thumbnail
Figure 3. (A–C) Funnel plots showing associations of smoking with VTE.

https://doi.org/10.1371/journal.pmed.1001515.g003

Stratified Analyses

To explore study heterogeneity, we performed stratified analyses across a number of key study characteristics and clinical factors (Table 4). The finding of increased VTE risk in smokers was consistently observed in most of the stratified analyses. Study design, geographical area, or publication year were not significant sources of heterogeneity. In addition, the RRs in studies in which VTE cases were validated with imaging examination or medical record were not systematically different from studies in which they were not (Table 4; Figures S3, S4, S5). Level of adjustment in the primary studies seemed to be associated with the results (p = 0.03 for ever smokers and p<0.001 for current smokers). Studies with no adjustments for cardiovascular risk factors found no significant association between smoking and risk of VTE, while the analysis of studies adjusted for cardiovascular risk factors, especially for BMI, yielded relatively higher RRs for ever and current smokers. There was evidence of moderate heterogeneity for former smokers (I2: 57.10% [95% CI 20.40%–76.88%], p = 0.01), but not for ever smokers (I2: 30.44% [95% CI 0%–60.69%], p = 0.11) or current smokers (I2: 19.73% [95% CI 0%–57.71%]). In a case-control study by Zhu et al. [43], the adjusted risk estimate for former smokers (RR: 3.25 [95% CI 1.92–5.49]) was much higher than the pooled risk estimate. After excluding this single study, there was no evidence of heterogeneity (I2: 0% [95% CI 0%–58.32%], p = 0.44) and the pooled risk estimate still reached statistical significance (RR: 1.09 [95% CI 1.05–1.12]) (Figure 4). The risk for developing VTE was significantly higher in current smokers than former smokers after adjustment for BMI (p<0.001).

thumbnail
Figure 4. Cardiovascular risk factor-adjusted relative risk for VTE.

The size of each square is proportional to the study's weight (inverse of variance). Cardiovascular risk factors (BCDHAP): B, BMI, body weight or waist circumference; C, cholesterol; D, diabetes; H, hypertension; A, alcohol consumption; P, physical activity. For example, Sweetland S 2013 adjusted for body mass index, diabetes, hypertension, alcohol consumption, physical activity, and three other non-cardiovascular risk factors, but not for cholesterol.

https://doi.org/10.1371/journal.pmed.1001515.g004

thumbnail
Table 4. Stratified analysis of pooled relative risks of VTE for smokers and heterogeneity analysisa.

https://doi.org/10.1371/journal.pmed.1001515.t004

We undertook meta-regression to further identify the relationship between BMI and smoking-VTE risk. Although baseline BMI did not seem to be significantly correlated with the smoking-VTE risk for ever smokers, BMI-adjusted risk estimates were significantly higher than unadjusted ones (p = 0.02) (Figure S6).

We also evaluated whether a difference existed between men and women, DVT and PE, and unprovoked and provoked VTE in the smoking-VTE relationship. The stratified analyses shown in Table 4 suggest no modification of the relationship by these characteristics. To allow an unbiased comparison, we also calculated the RRs from studies reporting both estimates for men and women, DVT and PE, and unprovoked and provoked VTE. Similar pooled risks were again observed in both sexes (p = 0.95) [7],[8],[26],[37], sites of VTE (p = 0.31) [29],[33],[37], and types of VTE (p = 0.38) [8],[22],[27],[28],[33],[38],[39],[48].

Dose-Response Relationship and Incidence of VTE

After evaluating dose-response patterns for cigarettes per day and pack-years for ever versus never smokers, we observed a linear increase in VTE risk with increasing smoking consumption. The risk increased by 10.2% (95% CI 8.6%–11.8%) for every additional ten cigarettes per day or by 6.1% (95% CI 3.8%–8.5%) for every additional ten pack-years (for example, an individual who smoked one pack of cigarettes per day for 40 y or two packs per day for 20 y has a relative increased risk of 26.7% [95% CI 16.0%–38.4%] for developing VTE compared with someone who never smoked) (Figure 5).

thumbnail
Figure 5. Linear dose-response relationship between relative risk of VTE incidence and tobacco consumption with cigarettes per day (A) and pack-years (B) as the explanatory variables.

The solid line represents point estimates of association between tobacco consumption and VTE risk; dashed lines are 95% CIs. Circles present the dose-specific RR estimates reported in each study. The area of each circle is proportional to the inverse variance of the RR. The dotted line represents the null hypothesis of no association. The vertical axis is on a log scale.

https://doi.org/10.1371/journal.pmed.1001515.g005

From eight population-based studies that reported information on person-years in smokers and nonsmokers, we could calculate absolute annual rates of VTE cases from the general population: 176.3 cases per 100,000 person-years in smokers and 152.0 cases in nonsmokers, corresponding to an absolute risk increase of 24.3 (95% CI 15.4–26.7) cases per 100,000 person-years.

PAF Calculations

Using the average prevalence of smoking from included cohort studies and the summary estimates obtained from all studies combined, the PAF of VTE due to smoking were 8.7% (95% CI 4.8%–12.3%) for ever smoking, 5.8% (95% CI 3.6%–8.2%) for current smoking, and 2.7% (95% CI 0.8%–4.5%) for former smoking. If cardiovascular risk factor-adjusted risk estimates were used, then the proportions of VTE explained by three categories of smoking increased to 10.6% (95% CI 7.8%–12.8%), 7.7% (95% CI 6.1%–9.1%), and 2.8% (95% CI 1.1%–4.5%), respectively.

Discussion

The present meta-analysis, involving approximately 4 million participants and more than 35,000 patients with VTE from 32 observational studies, found a slightly increased risk of VTE for smokers compared with non-smokers. The risk was higher in studies adjusted for conventional cardiovascular risk factors, especially for BMI. The risk of developing VTE was greater for current smokers than for former smokers, and a dose-response relationship was found for daily smoking and pack-years smoked.

Recent studies have suggested that patients with obesity, hypertension, diabetes, or dyslipidemia were at risk of developing VTE, whereas conflicting results were reported for smoking [10],[11],[49][53]. This meta-analysis is the first to our knowledge to confirm smoking to be an independent risk factor for VTE. The risk magnitude appears to be less robust than those reported for well-established major risk factors such as cancer, surgery, pregnancy, use of estrogens, or mutation of factor V Leiden and prothrombin [54][57]. However, smoking is more common and its coexistence is associated with an additive causative effect. For example, there was a synergistic effect on VTE risk for smoking and oral contraceptive use. Pomp et al. reported the OR of developing VTE for oral contraceptive users was 3.90, but increased to 8.79 when current smoking was added [37]. One prospective cohort study also identified a hazard ratio of 3.75 for the association of the combination of current smoking and the prothrombin mutation with the risk of VTE, significantly higher than that of the prothrombin mutation only [58]. Thus, given the multi-factorial nature of VTE, it is highly likely that the concomitant action of smoking may be responsible for a proportion of VTE in the general population.

A causal relationship between VTE and smoking may be mediated by different mechanisms. Our results suggest that the association of smoking with VTE risk may be largely mediated by an acute mechanism, supported by a dose-response relationship for the amount of current smoking and the higher risk in current compared to former smokers. In addition, the association was not solely due to smoking-related secondary diseases, because we found a positive association between current smoking and both unprovoked and provoked VTE. Furthermore, there is biological plausibility for the relationship. A procoagulant state, reduced fibrinolysis, inflammation, and increased blood viscosity may underlie the association between smoking and VTE risk [59][61]. Smoking is associated with a higher level of plasma fibrinogen, hence the increase of factor VIII, which has been reported to be associated with VTE [62]. It has been shown that the fibrinogen concentration decreased quickly after cessation of smoking and the fibrinogen concentration was nearly equal in former smokers and never smokers [63],[64]. Yarnell et al. detected a positive relationship between the amount of current tobacco consumption and plasminogen activator inhibitor-1 concentration, which may also be related to VTE [65][67]. These findings might suggest an acute causal association and the dose-response relationship between VTE and smoking. However, a relatively weak association between former smoking and the risk of VTE was also observed. We suppose this association may be mediated by secondary smoking-related diseases. Former smoking is related to cardiovascular diseases, diabetes, and certain types of cancer [68][70], which may be associated with risk of VTE [54],[71]. It is also possible that chronic inhalation of tobacco smoke, causing progressive lung destruction, chronic obstructive pulmonary disease, and emphysema [72], may also result in a hypercoagulable state and thus contribute to an increased risk of VTE [73].

It is of note that lack of adjustment for BMI tends to deflate the pooled risk estimate, indicating that BMI is an important confounding factor when assessing the smoking-VTE association. Limiting studies to those adjusted for BMI identified no significant heterogeneity for ever and current smokers, suggesting that BMI may be a source of heterogeneity. Current smokers tend to be thinner than nonsmokers or former smokers [74][76], and several studies have shown that smokers' BMI is lower [77]. However, previous studies also identified obesity or weight gain to be an independent risk factor for developing VTE [11],[20],[78]. Thus, given that body leanness of some smokers might partly reduce the risk, the true magnitude of association between smoking and VTE may be greater. This may be an explanation for the non-significant association observed in previous observational studies and meta-analysis that did not control for BMI.

Strengths of this meta-analysis include the strict inclusion criteria, the large number of patients analyzed, the robustness of the findings in sensitivity analyses, the dose-response relationship, and the fact that all subgroup analyses were prespecified a priori. The absence of important publication bias supports the robustness of the study findings. A possible limitation of our study is the heterogeneity of the studies with regard to adjustment of the estimates for potential confounders. Although differences in levels of adjustment seems, at least in part, to explain this finding, heterogeneity still exists in former smokers, even after we confined the analysis to studies that adjusted for BMI. This suggests that apart from BMI, there are other factors that potentially may confound the risk estimates. Furthermore, baseline BMI was not significantly associated with the smoking-VTE risk, indicating that the relation between BMI and risk of VTE in smokers needs to be further elucidated. Inclusion of different types of studies into one meta-analysis may also introduce heterogeneity into the results. Despite this, the consistency of the finding of an increased risk of VTE among smokers in both case-control and cohort studies suggests that the association is valid. In addition, the results from a given study for the three comparisons (ever versus never, former versus never, current versus never) are not independent and there is a possibility of a type I error. However, the results continued to be statistically significant after adjusting for multiple comparisons by setting α = 0.05/3. Like all meta-analyses, our study has the limitation of being a retrospective analysis. Another limitation was the lack of individual participant data, which precluded determining the independent associations of individual variables with study outcomes. Instead, we used between-study meta-regressions, when possible.

In conclusion, the results from this meta-analysis suggest that smoking slightly increases the risk of VTE, independent of conventional cardiovascular risk factors. BMI may be a potential confounding factor in the risk estimates. The association between smoking and VTE has clinical relevance with respect to individual screening, risk factor modification, and the primary and secondary prevention of VTE. Future prospective studies are needed to elucidate the specific pathogenic mechanisms.

Supporting Information

Figure S1.

Forest plot for VTE incidence: risk estimates for current versus never smokers. The size of each square is proportional to the study's weight (inverse of variance).

https://doi.org/10.1371/journal.pmed.1001515.s001

(TIF)

Figure S2.

Forest plot for VTE incidence: risk estimates for former versus never smokers. The size of each square is proportional to the study's weight (inverse of variance).

https://doi.org/10.1371/journal.pmed.1001515.s002

(TIF)

Figure S3.

Pooled relative risks of VTE for ever smokers stratified by VTE validation. VTE case confirmation was based on medical record, radiology, or autopsy (validated) and questionnaire or patient registry (not validated).

https://doi.org/10.1371/journal.pmed.1001515.s003

(TIF)

Figure S4.

Pooled relative risks of VTE for current smokers stratified by VTE validation. VTE case confirmation was based on medical record, radiology, or autopsy (validated) and questionnaire or patient registry (not validated).

https://doi.org/10.1371/journal.pmed.1001515.s004

(TIF)

Figure S5.

Pooled relative risks of VTE for former smokers stratified by VTE validation. VTE case confirmation was based on medical record, radiology, or autopsy (validated) and questionnaire or patient registry (not validated).

https://doi.org/10.1371/journal.pmed.1001515.s005

(TIF)

Figure S6.

Relationship between baseline BMI and smoking-VTE risk for ever smokers. Regression analyses were stratified, where appropriate, by level of adjustment for BMI. Meta-regression p = 0.64 for BMI-unadjusted risk estimates, p = 0.92 for BMI-adjusted risk estimates.

https://doi.org/10.1371/journal.pmed.1001515.s006

(TIF)

Author Contributions

Conceived and designed the experiments: SHW YJC. Performed the experiments: YJC ZHL FJY WTZ DDZ YGD SHW. Analyzed the data: YJC ZHL SHW. Contributed reagents/materials/analysis tools: YJC ZHL FJY WTZ DDZ YGD SHW. Wrote the first draft of the manuscript: YJC ZHL SHW. Contributed to the writing of the manuscript: YJC SHW ZHL FJY WTZ DDZ YGD. ICMJE criteria for authorship read and met: SHW YJC ZHL FJY WTZ DDZ YGD. Agree with manuscript results and conclusions: SHW YJC ZHL FJY WTZ DDZ YGD.

References

  1. 1. Schultz H (1998) Tobacco or health: A global status report. Ann Saudi Med 18: 195.
  2. 2. WHO urges more countries to require large, graphic health warnings on tobacco packaging: the WHO report on the global tobacco epidemic, 2011 examines anti-tobacco mass-media campaigns. Cent Eur J Public Health 19: 133, 151.
  3. 3. Flinterman LE, van Hylckama VA, Cannegieter SC, Rosendaal FR (2012) Long-term survival in a large cohort of patients with venous thrombosis: incidence and predictors. PLoS Med 9: e1001155
  4. 4. Cushman M, Tsai AW, White RH, Heckbert SR, Rosamond WD, et al. (2004) Deep vein thrombosis and pulmonary embolism in two cohorts: the longitudinal investigation of thromboembolism etiology. Am J Med 117: 19–25.
  5. 5. Lindblad B, Sternby NH, Bergqvist D (1991) Incidence of venous thromboembolism verified by necropsy over 30 years. BMJ 302: 709–711.
  6. 6. Tavora F, Crowder C, Kutys R, Burke A (2008) Discrepancies in initial death certificate diagnoses in sudden unexpected out-of-hospital deaths: the role of cardiovascular autopsy. Cardiovasc Pathol 17: 178–182.
  7. 7. Holst AG, Jensen G, Prescott E (2010) Risk factors for venous thromboembolism: results from the Copenhagen City Heart Study. Circulation 121: 1896–1903.
  8. 8. Severinsen MT, Kristensen SR, Johnsen SP, Dethlefsen C, Tjonneland A, et al. (2009) Smoking and venous thromboembolism: a Danish follow-up study. J Thromb Haemost 7: 1297–1303.
  9. 9. Rosengren A, Freden M, Hansson PO, Wilhelmsen L, Wedel H, et al. (2008) Psychosocial factors and venous thromboembolism: a long-term follow-up study of Swedish men. J Thromb Haemost 6: 558–564.
  10. 10. Tsai AW, Cushman M, Rosamond WD, Heckbert SR, Polak JF, et al. (2002) Cardiovascular risk factors and venous thromboembolism incidence: the longitudinal investigation of thromboembolism etiology. Arch Intern Med 162: 1182–1189.
  11. 11. Ageno W, Becattini C, Brighton T, Selby R, Kamphuisen PW (2008) Cardiovascular risk factors and venous thromboembolism: a meta-analysis. Circulation 117: 93–102.
  12. 12. Greenland S (1987) Quantitative methods in the review of epidemiologic literature. Epidemiol Rev 9: 1–30.
  13. 13. Higgins JP, Thompson SG (2002) Quantifying heterogeneity in a meta-analysis. Stat Med 21: 1539–1558.
  14. 14. LEVIN ML (1953) The occurrence of lung cancer in man. Acta Unio Int Contra Cancrum 9: 531–541.
  15. 15. van Houwelingen HC, Arends LR, Stijnen T (2002) Advanced methods in meta-analysis: multivariate approach and meta-regression. Stat Med 21: 589–624.
  16. 16. Greenland S, Longnecker MP (1992) Methods for trend estimation from summarized dose-response data, with applications to meta-analysis. Am J Epidemiol 135: 1301–1309.
  17. 17. Miller JJ (1978) The inverse of the Freeman-Turkey double arcsine transformation. Am Stat 32: 138.
  18. 18. Egger M, Davey SG, Schneider M, Minder C (1997) Bias in meta-analysis detected by a simple, graphical test. BMJ 315: 629–634.
  19. 19. Goldhaber SZ, Grodstein F, Stampfer MJ, Manson JE, Colditz GA, et al. (1997) A prospective study of risk factors for pulmonary embolism in women. JAMA 277: 642–645.
  20. 20. Hansson PO, Eriksson H, Welin L, Svardsudd K, Wilhelmsen L (1999) Smoking and abdominal obesity: risk factors for venous thromboembolism among middle-aged men: “the study of men born in 1913”. Arch Intern Med 159: 1886–1890.
  21. 21. Klatsky AL, Armstrong MA, Poggi J (2000) Risk of pulmonary embolism and/or deep venous thrombosis in Asian-Americans. Am J Cardiol 85: 1334–1337.
  22. 22. Glynn RJ, Rosner B (2005) Comparison of risk factors for the competing risks of coronary heart disease, stroke, and venous thromboembolism. Am J Epidemiol 162: 975–982.
  23. 23. Lindqvist PG, Epstein E, Olsson H (2009) The relationship between lifestyle factors and venous thromboembolism among women: a report from the MISS study. Br J Haematol 144: 234–240.
  24. 24. Rosengren A, Freden M, Hansson PO, Wilhelmsen L, Wedel H, et al. (2008) Psychosocial factors and venous thromboembolism: a long-term follow-up study of Swedish men. J Thromb Haemost 6: 558–564.
  25. 25. Lutsey PL, Virnig BA, Durham SB, Steffen LM, Hirsch AT, et al. (2010) Correlates and consequences of venous thromboembolism: The Iowa Women's Health Study. Am J Public Health 100: 1506–1513.
  26. 26. Hippisley-Cox J, Coupland C (2011) Development and validation of risk prediction algorithm (QThrombosis) to estimate future risk of venous thromboembolism: prospective cohort study. BMJ 343: d4656.
  27. 27. Enga KF, Braekkan SK, Hansen-Krone IJ, le Cessie S, Rosendaal FR, et al. (2012) Cigarette smoking and the risk of venous thromboembolism: The Tromso Study. J Thromb Haemost 10: 2068–2074.
  28. 28. Wattanakit K, Lutsey PL, Bell EJ, Gornik H, Cushman M, et al. (2012) Association between cardiovascular disease risk factors and occurrence of venous thromboembolism. A time-dependent analysis. Thromb Haemost 108: 508–515.
  29. 29. Sweetland S, Parkin L, Balkwill A, Green J, Reeves G, et al. (2013) Smoking, surgery, and venous thromboembolism risk in women: United Kingdom cohort study. Circulation 127: 1276–1282.
  30. 30. Dreyer NA, Pizzo SV (1980) Blood coagulation and idiopathic thromboembolism among fertile women. Contraception 22: 123–135.
  31. 31. Lu Y, Hui R, Zhao Y (2001) [Insertion/deletion polymorphsim of the angiotensin I converting enzyme gene and pulmonary thromboembolism in Chinese population]. Zhonghua Jie He He Hu Xi Za Zhi 24: 265–268.
  32. 32. Ray JG, Langman LJ, Vermeulen MJ, Evrovski J, Yeo EL, et al. (2001) Genetics University of Toronto Thrombophilia Study in Women (GUTTSI): genetic and other risk factors for venous thromboembolism in women. Curr Control Trials Cardiovasc Med 2: 141–149.
  33. 33. Tosetto A, Frezzato M, Rodeghiero F (2003) Prevalence and risk factors of non-fatal venous thromboembolism in the active population of the VITA Project. J Thromb Haemost 1: 1724–1729.
  34. 34. Worralurt C, Taneepanichskul S (2005) Risk factors of venous thromboembolism (VTE) in Thai reproductive aged female: King Chulalongkorn Memorial Hospital experience. J Med Assoc Thai 88: 1502–1505.
  35. 35. Hirohashi T, Yoshinaga K, Sakurai T, Kanai M, Shimizu K, et al. (2006) [Study of the echocardiographic diagnosis of acute pulmonary thromboembolism and risk factors for venous thromboembolism]. J Cardiol 47: 63–71.
  36. 36. Sugimura K, Sakuma M, Shirato K (2006) Potential risk factors and incidence of pulmonary thromboembolism in Japan: results from an overview of mailed questionnaires and matched case-control study. Circ J 70: 542–547.
  37. 37. Pomp ER, Rosendaal FR, Doggen CJ (2008) Smoking increases the risk of venous thrombosis and acts synergistically with oral contraceptive use. Am J Hematol 83: 97–102.
  38. 38. Prandoni P, Bilora F, Marchiori A, Bernardi E, Petrobelli F, et al. (2003) An association between atherosclerosis and venous thrombosis. N Engl J Med 348: 1435–1441.
  39. 39. Jang MJ, Choi WI, Bang SM, Lee T, Kim YK, et al. (2009) Metabolic syndrome is associated with venous thromboembolism in the Korean population. Arterioscler Thromb Vasc Biol 29: 311–315.
  40. 40. Yamada N, Ota S, Liu Y, Crane MM, Chang CM, et al. (2010) Risk factors for nonfatal pulmonary embolism in a Japanese population: a hospital-based case-control study. Angiology 61: 269–274.
  41. 41. Bhoopat L, Rojnuckarin P, Hiransuthikul N, Intragumtornchai T (2010) Low vegetable intake is strongly associated with venous thromboembolism in Thai population. Blood Coagul Fibrinolysis 21: 758–763.
  42. 42. Quist-Paulsen P, Naess IA, Cannegieter SC, Romundstad PR, Christiansen SC, et al. (2010) Arterial cardiovascular risk factors and venous thrombosis: results from a population-based, prospective study (the HUNT 2). Haematologica 95: 119–125.
  43. 43. Zhu J, Zheng WJ, Kong FC, Zhang WJ, Wang HY, et al. (2010) CYP1A1, smoking and venous thromboembolism. Thromb Haemost 104: 702–708.
  44. 44. Cay N, Ipek A, Gumus M, Birkan Z, Ozmen E (2012) Platelet activity indices in patients with deep vein thrombosis. Clin Appl Thromb Hemost 18: 206–210.
  45. 45. Di Minno MN, Tufano A, Guida A, Di Capua M, De Gregorio AM, et al. (2011) Abnormally high prevalence of major components of the metabolic syndrome in subjects with early-onset idiopathic venous thromboembolism. Thromb Res 127: 193–197.
  46. 46. Abudureheman K, Mahemuti A, Xia YN, Hu XM (2012) [Association between gene polymorphisms of methylenetetrahydrofolate reductase and plasma homocysteine in Uygur patients with venous thromboembolism]. Zhonghua Xin Xue Guan Bing Za Zhi 40: 1030–1036.
  47. 47. Cil H, Yavuz C, Islamoglu Y, Tekbas EO, Demirtas S, et al. (2012) Platelet count and mean platelet volume in patients with in-hospital deep venous thrombosis. Clin Appl Thromb Hemost 18: 650–653.
  48. 48. Blondon M, Wiggins KL, McKnight B, Psaty BM, Rice KM, et al. (2013) The association of smoking with venous thrombosis in women. A population-based, case-control study. Thromb Haemost 109.
  49. 49. Parkin L, Sweetland S, Balkwill A, Green J, Reeves G, et al. (2012) Body mass index, surgery, and risk of venous thromboembolism in middle-aged women: a cohort study. Circulation 125: 1897–1904.
  50. 50. Schmidt M, Johannesdottir SA, Lemeshow S, Lash TL, Ulrichsen SP, et al. (2013) Obesity in young men, and individual and combined risks of type 2 diabetes, cardiovascular morbidity and death before 55 years of age: a Danish 33-year follow-up study. BMJ Open 3: e002698.
  51. 51. Venous thromboembolic disease and combined oral contraceptives: results of international multicentre case-control study. World Health Organization Collaborative Study of Cardiovascular Disease and Steroid Hormone Contraception. Lancet 346: 1575–1582.
  52. 52. Sultan AA, Tata LJ, West J, Fiaschi L, Fleming KM, et al. (2013) Risk factors for first venous thromboembolism around pregnancy: a population-based cohort study from the United Kingdom. Blood 121: 3953–3961.
  53. 53. Kawasaki T, Kambayashi J, Ariyoshi H, Sakon M, Suehisa E, et al. (1997) Hypercholesterolemia as a risk factor for deep-vein thrombosis. Thromb Res 88: 67–73.
  54. 54. Horsted F, West J, Grainge MJ (2012) Risk of venous thromboembolism in patients with cancer: a systematic review and meta-analysis. PLoS Med 9: e1001275
  55. 55. Januel JM, Chen G, Ruffieux C, Quan H, Douketis JD, et al. (2012) Symptomatic in-hospital deep vein thrombosis and pulmonary embolism following hip and knee arthroplasty among patients receiving recommended prophylaxis: a systematic review. JAMA 307: 294–303.
  56. 56. Scarabin PY, Oger E, Plu-Bureau G (2003) Differential association of oral and transdermal oestrogen-replacement therapy with venous thromboembolism risk. Lancet 362: 428–432.
  57. 57. Segal JB, Brotman DJ, Necochea AJ, Emadi A, Samal L, et al. (2009) Predictive value of factor V Leiden and prothrombin G20210A in adults with venous thromboembolism and in family members of those with a mutation: a systematic review. JAMA 301: 2472–2485.
  58. 58. Severinsen MT, Overvad K, Johnsen SP, Dethlefsen C, Madsen PH, et al. (2010) Genetic susceptibility, smoking, obesity and risk of venous thromboembolism. Br J Haematol 149: 273–279.
  59. 59. Lee KW, Lip GY (2003) Effects of lifestyle on hemostasis, fibrinolysis, and platelet reactivity: a systematic review. Arch Intern Med 163: 2368–2392.
  60. 60. Miller GJ, Bauer KA, Cooper JA, Rosenberg RD (1998) Activation of the coagulant pathway in cigarette smokers. Thromb Haemost 79: 549–553.
  61. 61. Yarnell JW, Sweetnam PM, Rumley A, Lowe GD (2000) Lifestyle and hemostatic risk factors for ischemic heart disease: the Caerphilly Study. Arterioscler Thromb Vasc Biol 20: 271–279.
  62. 62. Oger E, Lacut K, Van Dreden P, Bressollette L, Abgrall JF, et al. (2003) High plasma concentration of factor VIII coagulant is also a risk factor for venous thromboembolism in the elderly. Haematologica 88: 465–469.
  63. 63. Feher MD, Rampling MW, Brown J, Robinson R, Richmond W, et al. (1990) Acute changes in atherogenic and thrombogenic factors with cessation of smoking. J R Soc Med 83: 146–148.
  64. 64. Bakhru A, Erlinger TP (2005) Smoking cessation and cardiovascular disease risk factors: results from the Third National Health and Nutrition Examination Survey. PLoS Med 2: e160
  65. 65. Yarnell JW, Sweetnam PM, Rumley A, Lowe GD (2001) Lifestyle factors and coagulation activation markers: the Caerphilly Study. Blood Coagul Fibrinolysis 12: 721–728.
  66. 66. Yukizawa Y, Inaba Y, Watanabe S, Yajima S, Kobayashi N, et al. (2012) Association between venous thromboembolism and plasma levels of both soluble fibrin and plasminogen-activator inhibitor 1 in 170 patients undergoing total hip arthroplasty. Acta Orthop 83: 14–21.
  67. 67. Gary T, Hafner F, Froehlich H, Stojakovic T, Scharnagl H, et al. (2010) High factor VIII activity, high plasminogen activator inhibitor 1 antigen levels and low factor XII activity contribute to a thrombophilic tendency in elderly venous thromboembolism patients. Acta Haematol 124: 214–217.
  68. 68. Willi C, Bodenmann P, Ghali WA, Faris PD, Cornuz J (2007) Active smoking and the risk of type 2 diabetes: a systematic review and meta-analysis. JAMA 298: 2654–2664.
  69. 69. Yeh JM, Hur C, Schrag D, Kuntz KM, Ezzati M, et al. (2013) Contribution of H. pylori and smoking trends to US incidence of intestinal-type noncardia gastric adenocarcinoma: a microsimulation model. PLoS Med 10: e1001451
  70. 70. Khaw KT, Friesen MD, Riboli E, Luben R, Wareham N (2012) Plasma phospholipid fatty acid concentration and incident coronary heart disease in men and women: the EPIC-Norfolk prospective study. PLoS Med 9: e1001255.
  71. 71. Lutsey PL, Virnig BA, Durham SB, Steffen LM, Hirsch AT, et al. (2010) Correlates and consequences of venous thromboembolism: The Iowa Women's Health Study. Am J Public Health 100: 1506–1513.
  72. 72. Grumelli S, Corry DB, Song LZ, Song L, Green L, et al. (2004) An immune basis for lung parenchymal destruction in chronic obstructive pulmonary disease and emphysema. PLoS Med 1: e8.
  73. 73. Schneider C, Bothner U, Jick SS, Meier CR (2010) Chronic obstructive pulmonary disease and the risk of cardiovascular diseases. Eur J Epidemiol 25: 253–260.
  74. 74. Gordon T, Kannel WB, Dawber TR, McGee D (1975) Changes associated with quitting cigarette smoking: the Framingham Study. Am Heart J 90: 322–328.
  75. 75. Smoking wastes a good Parisienne. JAMA 262: 1185–1186.
  76. 76. Albanes D, Jones DY, Micozzi MS, Mattson ME (1987) Associations between smoking and body weight in the US population: analysis of NHANES II. Am J Public Health 77: 439–444.
  77. 77. Shimokata H, Muller DC, Andres R (1989) Studies in the distribution of body fat. III. Effects of cigarette smoking. JAMA 261: 1169–1173.
  78. 78. Cushman M, Kuller LH, Prentice R, Rodabough RJ, Psaty BM, et al. (2004) Estrogen plus progestin and risk of venous thrombosis. JAMA 292: 1573–1580.