Next Article in Journal
Predictive Biomarkers of Pathological Response to Neoadjuvant Chemoradiotherapy for Locally Advanced Soft Tissue Sarcomas
Previous Article in Journal
Novel Investigational Agents and Pathways That May Influence the Future Management of Acute Myeloid Leukemia
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

The Association between Statins and Liver Cancer Risk in Patients with Heart Failure: A Nationwide Population-Based Cohort Study

1
Division of Gastroenterology, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 11490, Taiwan
2
Division of Cardiology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
3
Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
4
Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
5
Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
6
Division of Cardiology, Department of Internal Medicine, Taitung MacKay Memorial Hospital, Taitung 95054, Taiwan
7
Division of Cardiovascular Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
8
Department of Emergency, University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA
9
Division of Gastroenterology and Hepatology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
10
TMU Research Center for Digestive Medicine, Taipei Medical University, Taipei 110, Taiwan
11
Division of Gastroenterology and Hepatology, Department of Internal Medicine, Shuang Ho Hospital, New Taipei City 23561, Taiwan
12
Graduate Institute of Data Science, College of Management, Taipei Medical University, Taipei 11031, Taiwan
13
Department of Neurosurgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan
14
Division of Cardiology, Department of Internal Medicine and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Cancers 2023, 15(11), 2959; https://doi.org/10.3390/cancers15112959
Submission received: 21 April 2023 / Revised: 25 May 2023 / Accepted: 26 May 2023 / Published: 29 May 2023
(This article belongs to the Section Cancer Epidemiology and Prevention)

Abstract

:

Simple Summary

Heart failure is a major public health challenge with similar risk factors to those of cancer. HMG-CoA reductase inhibitors, also known as statins, are widely prescribed lipid-lowering agents. Chemoprevention has been reported as a pleiotropic effect of statins. We aimed to evaluate the chemoprotective effect of statins on liver cancer in patients with heart failure and to further identify the differences in effectiveness among statin doses and types. The results demonstrated that statins potentially decreased the risk of liver cancer in patients with heart failure in the entire cohort as well as in sex-, age-, and dose-stratified subgroup analyses as compared with the control group. Moreover, both hydrophilic and lipophilic statins showed significant risk reductions. The findings of the present study demonstrate a potential benefit in terms of liver cancer risk for patients with heart failure using statins.

Abstract

Heart failure (HF) and cancer have similar risk factors. HMG-CoA reductase inhibitors, also known as statins, are chemoprotective agents against carcinogenesis. We aimed to evaluate the chemoprotective effects of statins against liver cancer in patients with HF. This cohort study enrolled patients with HF aged ≥20 years between 1 January 2001 and 31 December 2012 from the National Health Insurance Research Database in Taiwan. Each patient was followed to assess liver cancer risk. A total of 25,853 patients with HF were followed for a 12-year period; 7364 patients used statins and 18,489 did not. The liver cancer risk decreased in statin users versus non-users (adjusted hazard ratio (aHR) = 0.26, 95% confidence interval (CI): 0.20–0.33) in the entire cohort in the multivariate regression analysis. In addition, both lipophilic and hydrophilic statins reduced the liver cancer risk in patients with HF (aHR 0.34, 95% CI: 0.26–0.44 and aHR 0.42, 95% CI: 0.28–0.54, respectively). In the sensitivity analysis, statin users in all dose-stratified subgroups had a reduced liver cancer risk regardless of age, sex, comorbidity, or other concomitant drug use. In conclusion, statins may decrease liver cancer risk in patients with HF.

1. Introduction

Heart failure (HF) is a major public health challenge and global epidemic [1,2] caused by aging demographics and the increasing prevalence of comorbidities, such as hypertension, diabetes, coronary artery disease, obesity, and atrial fibrillation [3]. Improvements in HF treatment have further extended life expectancy and increased the prevalence of non-cardiac morbidity in patients with HF. Some epidemiological studies have demonstrated that cancer is the major cause of non-cardiac death in patients with HF [4,5,6]. Predisposing conditions such as neurohormonal activation, systemic inflammation, and oxidative stress have been suggested to contribute to both HF and malignancy [7,8].
HMG-CoA reductase inhibitors (statins) are the most widely prescribed lipid-lowering agents and inhibit the mevalonate pathway. Moreover, studies have demonstrated that statins exert pleiotropic effects, such as anti-inflammatory, antioxidant, and immunomodulatory effects [9,10,11,12]. Various experimental and clinical studies have reported that statins are chemoprotective against carcinogenesis because of their potential pleiotropic mechanisms [13,14,15]. However, although statin use reportedly decreases the risk of many cancers [16], the chemoprotective effects of hydrophilic and lipophilic statins on liver cancer have been inconsistent [17,18]
Primary liver cancer is the fourth leading cause of cancer-related deaths [19], and a nationwide study of statin use in patients with HF reported a lower risk of cancer incidence and cancer-related mortality [20]. In the present study, we aimed to investigate the liver cancer risk in patients with HF with or without statin treatment as well as the differences among statin doses and types.

2. Materials and Methods

Taiwan’s National Health Insurance program has been mandatory for all citizens since 1995. The program provides universal health insurance coverage to Taiwanese residents [21]. Data obtained from the National Health Insurance Research Database are similar to those of the general population in regard to age, gender, and health-care costs. Data are anonymized before being released to researchers.
Patients who received a diagnosis of HF (with the International Classification of Diseases, Ninth Revision, Clinical Modification code 428.X; n = 45,153) who had at least two outpatient department visits or one hospital admission between 1 January 2001 and 31 December 2012 were identified from the database (n = 35,043). Patients who were younger than 40 years (n = 1509), who had a history of any cancer before the enrollment date (n = 4155) or who had received a statin prescription within 6 months before the enrollment date (n = 3526) were excluded. A total of 25,853 patients with HF were included in the study cohort and were followed up for 12 years. Of the 25,853 patients with HF, 7364 used statins and 18,489 did not (Figure 1).
The primary outcome was the incidence of primary liver cancer (with the International Classification of Diseases, Ninth Revision, Clinical Modification code 155.X) during the follow-up period. Follow-up ended on 31 December 2012 or upon receipt of a new diagnosis of liver cancer, withdrawal from the National Health Insurance program, loss to follow-up, or death, whichever occurred first. Demographic characteristics (age and sex), comorbidities (diabetes, hypertension, and dyslipidemia), Charlson comorbidity index (CCI) scores, urbanization level, monthly income, and use of non-statin lipid-lowering drugs (metformin, aspirin, and angiotensin-converting enzyme inhibitors (ACEIs)/angiotensin II receptor blockers (ARBs)) were collected.
We aimed to evaluate the preventive effects of statins in patients with HF who have a higher risk of liver cancer. The end point was primary liver cancer. To measure statin exposure, we used the defined daily dose (DDD) as a measurement tool as defined by the World Health Organization to assume the average maintenance dose per day of a drug consumed for its main indication in adults [22]. A prescription refill lasts for 3 months and can be filled up to three times; therefore, we categorized the DDDs (for the entire observation period for each patient) of statins into four groups in each cohort (<28, 28–90, 91–365, and >365 cumulative DDDs (cDDDs)) to examine the dose–response relationship. Patients with <28 cDDDs who received statins were defined as statin non-users [23]. Furthermore, to compare the effect of the solubility difference of each statin, we categorized statin use into individual statin groups in each cohort to evaluate the preventive effects of different statins.
To estimate the effect of statins, a propensity score (PS) was derived using a logistic regression model accounting for covariates predicting statin exposure. All potential confounders were included in the regressor list. This method has been used in observational studies to reduce selection bias [24]. The covariates in the main model were adjusted for the PSs for age (40–54, 55–64, and ≥75 years), sex, CCI score, diabetes, hypertension, dyslipidemia, urbanization level, and monthly income level (0, 1–21,000, 21,000–33,300, and ≥33,301 New Taiwan dollars) [25]. The Kaplan–Meier method was used for investigating the liver-cancer-free survival rate in patients with HF who were stratified according to statin use status and statin use dose status.
A Cox proportional hazards model was used to calculate the hazard ratios (HRs) for liver cancer among the statin users and non-users. In the multivariate analysis, the HRs were adjusted for age, sex, CCI score, diabetes, hypertension, dyslipidemia, urbanization level, and monthly income. A stratified analysis was conducted to evaluate the effect of statin use on age and sex. All analyses were conducted using SAS software (version 9.3; SAS, Cary, NC, USA); two-tailed p < 0.05 was considered significant. In epidemiological studies, through external adjustments, sensitivity analyses can be used to clarify the effects of drugs and other covariates [26].
Therefore, in the present study, sensitivity analyses with adjustments were used to determine the associations of age and sex; diabetes, dyslipidemia, hypertension, and CCI score; and the use of non-statin lipid-lowering drugs (metformin, aspirin, and ACEIs/ARBs) with the incidence of liver cancer. In addition to the covariates in the main model, the models were stratified by the use of different drugs as additional covariates.

3. Results

The demographic characteristics, medical conditions, medication use, level of urbanization, and monthly income level of the entire cohort and for patients with HF with and without statin use are listed in Table 1. In total, 25,853 patients with HF were enrolled in the study cohort: 7364 (28.5%) patients with HF used statins and 18,489 (71.5%) did not. The prevalence of pre-existing medical comorbidities, namely hypertension (76.58% versus 72.13%, p < 0.001), diabetes (39.42% versus 31.50%, p < 0.001), and dyslipidemia (44.91% versus 28.06%, p < 0.001) was higher among statin users versus non-users. The percentage of patients with hepatitis B or hepatitis C was lower among statin users than non-users (91.12% versus 88.59%, p < 0.001). In addition, several significant differences were observed in the distribution of age, sex, CCI score, monthly income level, urbanization level, and non-statin lipid-lowering drug use, such as metformin, a renin–angiotensin-aldosterone system inhibitor (RAASI), or aspirin, between statin users and non-users.
The distribution of statin use in patients with HF is shown in Table 2. In total, 17,973 (69.52%) patients never used statins, and 516 (2.00%) patients used statins for less than 28 days; 7364 (28.48%) patients used statins for more than 28 days. Among the participants who had ever used statins, 3495 (44.35%) patients used more than one type of statin. The most prescribed statin was atorvastatin (54.11%), followed by rosuvastatin (28.22%) and simvastatin (28.05%).
The liver cancer risk among statin users and non-users in the study cohort is described in Table 3 and Figure 2. The total follow-up durations were 51,080.5 and 85,867.4 person–years for statin users and non-users, respectively. After adjusting for age, sex, CCI score, diabetes, hypertension, dyslipidemia, urbanization level, and monthly income level using PS matching, the risk of liver cancer was analyzed. Compared with patients with HF without statin use, the adjusted HRs (aHRs) for liver cancer risk were decreased in patients with HF who used statins (aHR 0.26, 95% CI [0.20, 0.33]). Further stratified analyses revealed that the aHRs remained significantly decreased in patients with HF who used statins, regardless of age or sex. Compared with the patients with HF without statin use, the aHRs for liver cancer decreased among statin users aged 40–64, 65–74, and ≥75 years (aHR 0.23, 95% CI [0.15,0.34]; aHR 0.30, 95% CI [0.20,0.44]; and 0.25 [0.13, 0.48], respectively). In the sex-stratified analysis, the aHRs for liver cancer were lower in patients with HF versus those without statin use (women: aHR 0.25, 95% CI [0.17, 0.36]; men: aHR 0.27, 95% CI [0.19, 0.38]).
The incidence rates and aHRs for liver cancer associated with different statin doses during the follow-up period are listed in Table 4 and Figure 3. Compared with the non-users, statin users had a lower risk of liver cancer (aHR 0.26, 95% CI [0.20, 0.33]). Subgroup analysis of different statin doses, namely 28–90, 91–365, and >365 cDDDs, were associated with a lower liver cancer risk compared with non-users (aHR 0.42, 95% CI [0.27, 0.67]; aHR 0.32,95% CI [0.21, 0.48]; and aHR 0.18, 95% CI [0.12, 0.26], respectively). Statins can be classified as lipophilic or hydrophilic based on their solubility in water. Lipophilic statins include simvastatin, lovastatin, atorvastatin, and fluvastatin; hydrophilic statins include pravastatin and rosuvastatin. Both lipophilic and hydrophilic statins reduced the liver cancer risk in patients with HF (aHR 0.34, 95% CI [0.26,0.44] and aHR 0.42, 95% CI [0.28,0.64], respectively). All subgroups at each statin dose showed a decreased risk of liver cancer, except for the group with 28–90 cDDDs among hydrophilic statin users. Subgroup analyses of individual statins revealed a significantly lower risk of liver cancer in patients treated with simvastatin, atorvastatin, and rosuvastatin alone (aHRs: 0.51, 0.39, and 0.34, respectively).
Table 5 presents the sensitivity analyses results examining the association between statin treatment and reduced liver cancer risk as measured by aHRs. To estimate the effect of statin dose on liver cancer risk reduction, the entire cohort was treated with <28, 28–90, 91–365, or >365 cDDDs. Additional covariate adjustments were made to the main model, including non-statin medication, metformin, RAASI, and aspirin use, to assess the association of statin use with the risk of liver cancer for the four potential chemoprotective drugs separately. The results showed a significantly lower risk of liver cancer compared with the nonusers in the main model with the additional covariates, with a tendency towards dose-dependent risk reduction. In addition, subgroup analyses by age, sex, CCI score, diabetes, dyslipidemia, hypertension, hepatitis B or C, non-statin lipid-lowering drugs, metformin, RAASI, and aspirin were conducted. All aHRs indicated that the statins in each dose-stratified group significantly reduced the liver cancer risk in all subgroups, regardless of age, sex, comorbidities, or concomitant drug use, as compared with that in the statin group with <28 cDDDs.

4. Discussion

To the best of our knowledge, this is the first study to assess the effect of statins on the reduction of liver cancer risk in patients with HF. The effects of different statin types and doses were also investigated. Our results demonstrated that statin users had a significantly lower risk of liver cancer than nonusers among patients with HF in the entire cohort and in the sex- and age-stratified subgroup analyses. A significant risk reduction was also noted in the dose-stratified subgroups of 28–90, 91–365, and > 365 cDDDs. A potential dose-dependent effect of statins on liver disease risk reduction was observed. In addition, both hydrophilic and lipophilic statin use significantly decreased the liver cancer risk compared with that in non-users among patients with HF.
HF and cancer share common pathophysiological mechanisms [27,28,29], and several studies have indicated that patients with HF are prone to carcinogenesis [7,30,31,32] derived from a number of predisposing conditions of malignancy, including neurohormonal activation to tumorigenesis, systemic inflammation, and oxidative stress [7,8]. Meanwhile, HMG-CoA reductase inhibitors (statins) and lipid-lowering agents [33] also exert anti-inflammatory [34], antioxidative [35], and anti-cancer effects [14,36]. Statin use may confer chemoprotective effects through various mechanisms, including the inhibition of downstream products in the mevalonate pathway [9,10,11,37,38], triggering tumor-specific apoptosis [39], arresting the growth 1 phase of the cell cycle by inhibiting the proteasome pathway [40], reversing the likelihood of malignancy, and reducing the invasiveness of carcinoma in situ [38]. The dominant anti-inflammatory effects of statins in patients with HF were reported in a previous study [34], which implied the potential of diminishing cancer progression in this patient group. A retrospective cohort study revealed that statins were associated with a lower liver cancer risk in patients with HF in a subgroup analysis [20]. Our study is the first to demonstrate that statin use is associated with a decreased risk of liver cancer in patients with HF compared with patients without statin use based on the analysis of a nationwide database, independent of sex and age.
Our study assessed the effect of statin dose on liver cancer risk reduction and revealed that statin use had a tendency towards dose-dependent risk reduction among dose-stratified groups in the entire cohort and by statin cohorts. Nevertheless, in the subgroup analysis of metformin, among the subgroup of metformin use of less than 28 cDDDs, all statin use > 28 cDDDs decreased the liver cancer risk, whereas statin use of more than 365 cDDDs was associated with a significantly lower risk of liver cancer than that of statin use of 28–90 cDDDs, revealing a dose-dependent effect (aHR 0.50, CI [0.30–0.84], aHR 0.15, [0.08–0.27] for 28–90 and >365 cDDDs, respectively). Meanwhile, among the subgroup of metformin use of > 365 cDDDs, statin use of 28–90 cDDDs did not significantly reduce the risk of liver cancer (aHR 0.27, CI [0.07–1.11]). However, after a longer period of statin usage, a significantly lower risk was observed among patients with metformin use > 365 cDDDs (aHR 0.27, CI [0.11–0.67] and aHR 0.22, CI [0.12–0.41] for statin use 91–365 cDDDs and >365 cDDDs respectively). Metformin is one of the standard treatments for patients with diabetes. It has been reported before that diabetes is associated with an increased risk of cancer occurrence [41]. Therefore, patients with long-duration metformin use may need longer statin use to exhibit potential chemoprotective effects. Similarly, a case–control study from a nationwide population-based database evaluated the effect of 15 exposure combinations comprising four common drugs (statins, aspirin, metformin, and ACEIs/ARBs) on chemoprotective effects in comparison with a non-exposure group. The results demonstrated that individual or concomitant use of statins, aspirin, and ACEIs/ARBs could reduce the risk of liver cancer compared with the non-exposure group, although metformin use or concomitant use with metformin may increase the risk of liver cancer [42].
Consensus on the chemoprotective effects of hydrophilic and lipophilic statins in reducing the risk of liver cancer remains controversial [17,18]. Lipophilic statins enter cells predominantly via passive diffusion and are widely distributed in tissues, whereas the uptake of hydrophilic statins involves a liver-specific carrier-mediated mechanism [43]. For hepatitis virus carriers, interruption of the mevalonate pathway prevents viral replication by potentiating antiviral therapy and stimulating anti-tumor immunity [44,45,46,47,48]. Most nationwide studies and meta-analyses have shown that lipophilic statins reduce the risk of liver cancer; however, the results for hydrophilic statins have been inconsistent [15,18,49]. In our study, two hydrophilic statins were evaluated, with rosuvastatin being the most commonly used hydrophilic statin in our cohort (28.22%), followed by pravastatin (12.91%). Our findings showed that hydrophilic statins reduced the risk of liver cancer; however, in our single-drug analysis, only rosuvastatin significantly reduced the risk of liver cancer. Similar results were reported in two other meta-analysis studies [18,50]. Rosuvastatin has a more potent affinity for the active site of HMG-CoA reductase than other statins, and the hepatic uptake of rosuvastatin is reportedly more selective and efficient compared with other drugs [51,52,53,54]. Hence, these features potentially make rosuvastatin different from other hydrophilic statins and exhibit a strong chemoprotective effect in reducing the risk of liver cancer. However, as a high-potency statin [55], rosuvastatin may be more commonly indicated for patients with higher cholesterol levels and more severe heart failure, introducing bias by indication. Future studies are warranted to validate the findings of the present study.
Despite efforts to balance confounding factors, the present study has several limitations. First, this study was conducted using data from a health insurance claims database that lacks information on certain liver cancer risk factors, such as alcohol consumption, smoking habits, aflatoxin exposure, body mass index, and atherosclerosis [56,57]. Hence, we were unable to control for these potential confounding factors. However, we used PS matching to match patients by age, sex, CCI score, diabetes, hypertension, dyslipidemia, urbanization level, and monthly income. Urbanization level and monthly income were alternative factors for lifestyle and environmental factors [25,58]. Moreover, CCI scores include myocardial infarction, peripheral vascular disease, and cerebrovascular disease, which have a strong relationship with atherosclerosis. Second, we had no access to laboratory data or details of medical treatments for patients with hyperlipidemia. However, we conducted a sensitivity analysis of the adjusted HRs by dose-exposure-stratified subgroup analysis, which revealed persistent effectiveness trends in the main mode with additional covariates. Further investigations of the relationship between the effectiveness and dosage of statins may be considered to assist clinical novelty in real-world practice. Third, this was not a prospective, randomized control study. Although our study demonstrated significant results, further research is warranted to investigate the precise cause–effect relationship between statin use and liver cancer in patients with HF. Fourth, the HF patients were enrolled through the codes of International Classification of Diseases, Ninth Revision, which could not reveal the etiology of heart failure, such as cardiogenic or non-cardiogenic HF. Finally, the main factors underlying liver cancer in Taiwan differ from those in Western countries. Thus, the transferability of our findings to other healthcare systems may not be feasible.

5. Conclusions

This is the first nationwide population-based cohort study to investigate the effects of statins on liver cancer risk in patients with HF. Our study revealed that statin use is associated with decreased liver cancer risk in patients with HF in the entire cohort, as well as in sex- and age-stratified subgroup analyses, compared with participants without statin use. A reduction in liver cancer risk was also observed in each dose-stratified subgroup. In addition, both hydrophilic and lipophilic statins showed significant risk reduction.

Author Contributions

M.-C.L. and M.-Y.L.: conception and design of the study, analysis and interpretation of the data, and drafting and critical revision of the manuscript for important intellectual content. C.-C.C. (Chun-Chih Chiu), T.-Y.Y., Y.-A.F., W.J., M.-Y.C., M.-H.H., Y.-H.L. and T.-L.Y.: data collection and critical revision of the manuscript for important intellectual content. C.-C.C. (Chun-Chao Chen), W.-R.H., K.-J.L. and J.-C.L.: final approval of the draft for publication. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the Higher Education Sprout Project of the Ministry of Education (MOE) in Taiwan, grant number 112FRP-01-1; Taipei Medical University-Shuang Ho Hospital, Ministry of Health and Welfare; and Taipei Medical University–National Taiwan University of Science and Technology Joint Research Program, grant number TMU-NTUST-105-08.

Institutional Review Board Statement

The present study was conducted in accordance with all relevant guidelines and regulations. This study was approved by the Joint Institutional Review Board of Taipei Medical University (approval No. N201804043).

Informed Consent Statement

The Joint Institutional Review Board of Taipei Medical University waived the need for informed consents from participants. This waiver does not affect the rights and welfare of the participants.

Data Availability Statement

The data supporting the findings of the present research were sourced from the NHIRD in Taiwan. Due to legal restrictions imposed by the government of Taiwan related to the Personal Information Protection Act, the database cannot be made publicly available. However, upon reasonable request to the authors and with permission from the NHIRD, the relevant data are available.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Roger, V.L. Epidemiology of heart failure. Circ. Res. 2013, 113, 646–659. [Google Scholar] [CrossRef]
  2. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. Circulation 2022, 145, e895–e1032. [Google Scholar] [CrossRef]
  3. Jones, N.R.; Roalfe, A.K.; Adoki, I.; Hobbs, F.R.; Taylor, C.J. Survival of patients with chronic heart failure in the community: A systematic review and meta-analysis. Eur. J. Heart Fail 2019, 21, 1306–1325. [Google Scholar] [CrossRef] [PubMed]
  4. Hasin, T.; Gerber, Y.; McNallan, S.M.; Weston, S.A.; Kushwaha, S.S.; Nelson, T.J.; Cerhan, J.R.; Roger, V.L. Patients with heart failure have an increased risk of incident cancer. J. Am. Coll. Cardiol. 2013, 62, 881–886. [Google Scholar] [CrossRef] [PubMed]
  5. Banke, A.; Schou, M.; Videbaek, L.; Møller, J.E.; Torp-Pedersen, C.; Gustafsson, F.; Dahl, J.S.; Køber, L.; Hildebrandt, P.R.; Gislason, G.H.; et al. Incidence of cancer in patients with chronic heart failure: A long-term follow-up study. Eur. J. Heart Fail 2016, 18, 260–266. [Google Scholar] [CrossRef] [PubMed]
  6. Hasin, T.; Gerber, Y.; Weston, S.A.; Jiang, R.; Killian, J.M.; Manemann, S.M.; Cerhan, J.R.; Roger, V.L. Heart Failure After Myocardial Infarction Is Associated With Increased Risk of Cancer. J. Am. Coll. Cardiol. 2016, 68, 265–271. [Google Scholar] [CrossRef]
  7. Bertero, E.; Canepa, M.; Maack, C.; Ameri, P. Linking Heart Failure to Cancer: Background Evidence and Research Perspectives. Circulation 2018, 138, 735–742. [Google Scholar] [CrossRef]
  8. de Boer, R.A.; Hulot, J.S.; Tocchetti, C.G.; Aboumsallem, J.P.; Ameri, P.; Anker, S.D.; Bauersachs, J.; Bertero, E.; Coats, A.J.; Čelutkienė, J.; et al. Common mechanistic pathways in cancer and heart failure. A scientific roadmap on behalf of the Translational Research Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur. J. Heart Fail 2020, 22, 2272–2289. [Google Scholar] [CrossRef]
  9. Joharatnam-Hogan, N.; Alexandre, L.; Yarmolinsky, J.; Lake, B.; Capps, N.; Martin, R.M.; Ring, A.; Cafferty, F.; E Langley, R. Statins as Potential Chemoprevention or Therapeutic Agents in Cancer: A Model for Evaluating Repurposed Drugs. Curr. Oncol. Rep. 2021, 23, 29. [Google Scholar] [CrossRef]
  10. Chan, K.K.W.; Oza, A.M.; Siu, L.L. The statins as anticancer agents. Clin. Cancer Res. 2003, 9, 10–19. [Google Scholar]
  11. Danesh, F.R.; Sadeghi, M.M.; Amro, N.; Philips, C.; Zeng, L.; Lin, S.; Sahai, A.; Kanwar, Y.S. 3-Hydroxy-3-methylglutaryl CoA reductase inhibitors prevent high glucose-induced proliferation of mesangial cells via modulation of Rho GTPase/ p21 signaling pathway: Implications for diabetic nephropathy. Proc. Natl. Acad. Sci. USA 2002, 99, 8301–8305. [Google Scholar] [CrossRef] [PubMed]
  12. Blanco-Colio, L.M.; Muñoz-García, B.; Martín-Ventura, J.L.; Lorz, C.; Díaz, C.; Hernández, G.; Egido, J. 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors decrease Fas ligand expression and cytotoxicity in activated human T lymphocytes. Circulation 2003, 108, 1506–1513. [Google Scholar] [CrossRef] [PubMed]
  13. Juneja, M.; Kobelt, D.; Walther, W.; Voss, C.; Smith, J.; Specker, E.; Neuenschwander, M.; Gohlke, B.-O.; Dahlmann, M.; Radetzki, S.; et al. Statin and rottlerin small-molecule inhibitors restrict colon cancer progression and metastasis via MACC1. PLoS Biol. 2017, 15, e2000784. [Google Scholar] [CrossRef] [PubMed]
  14. Nielsen, S.F.; Nordestgaard, B.G.; Bojesen, S.E. Statin use and reduced cancer-related mortality. N. Engl. J. Med. 2012, 367, 1792–1802. [Google Scholar] [CrossRef]
  15. Simon, T.G.; Duberg, A.-S.; Aleman, S.; Hagstrom, H.; Nguyen, L.H.; Khalili, H.; Chung, R.T.; Ludvigsson, J.F. Lipophilic Statins and Risk for Hepatocellular Carcinoma and Death in Patients with Chronic Viral Hepatitis: Results From a Nationwide Swedish Population. Ann. Intern. Med. 2019, 171, 318–327. [Google Scholar] [CrossRef]
  16. Jiang, W.; Hu, J.-W.; He, X.-R.; Jin, W.-L.; He, X.-Y. Statins: A repurposed drug to fight cancer. J. Exp. Clin. Cancer Res. 2021, 40, 241. [Google Scholar] [CrossRef]
  17. Shi, M.; Zheng, H.; Nie, B.; Gong, W.; Cui, X. Statin use and risk of liver cancer: An update meta-analysis. BMJ Open 2014, 4, e005399. [Google Scholar] [CrossRef]
  18. Wang, Y.; Wang, W.; Wang, M.; Shi, J.; Jia, X.; Dang, S. A Meta-Analysis of Statin Use and Risk of Hepatocellular Carcinoma. Can. J. Gastroenterol. Hepatol. 2022, 2022, 5389044. [Google Scholar] [CrossRef]
  19. Villanueva, A. Hepatocellular Carcinoma. N. Engl. J. Med. 2019, 380, 1450–1462. [Google Scholar] [CrossRef]
  20. Ren, Q.-W.; Yu, S.-Y.; Teng, T.-H.K.; Li, X.; Cheung, K.-S.; Wu, M.-Z.; Li, H.-L.; Wong, P.-F.; Tse, H.-F.; Lam, C.S.P.; et al. Statin associated lower cancer risk and related mortality in patients with heart failure. Eur. Heart J. 2021, 42, 3049–3059. [Google Scholar] [CrossRef]
  21. Cheng, T.M. Reflections on the 20th anniversary of Taiwan’s single-payer National Health Insurance System. Health Aff. 2015, 34, 502–510. [Google Scholar] [CrossRef] [PubMed]
  22. Jeong, S.-M.; Shin, D.W.; Yoo, T.G.; Cho, M.H.; Jang, W.; Lee, J.; Kim, S. Association between statin use and Alzheimer’s disease with dose response relationship. Sci. Rep. 2021, 11, 15280. [Google Scholar] [CrossRef]
  23. Singh, S.; Singh, P.P. Statins for prevention of hepatocellular cancer: One step closer? Hepatology 2014, 59, 724–726. [Google Scholar] [CrossRef] [PubMed]
  24. D’Agostino, R.B., Jr. Propensity score methods for bias reduction in the comparison of a treatment to a non-randomized control group. Stat. Med. 1998, 17, 2265–2281. [Google Scholar] [CrossRef]
  25. Xing, Q.-Q.; Li, J.-M.; Dong, X.; Zeng, D.-Y.; Chen, Z.-J.; Lin, X.-Y.; Pan, J.-S. Socioeconomics and attributable etiology of primary liver cancer, 1990–2019. World J. Gastroenterol. 2022, 28, 2361–2382. [Google Scholar] [CrossRef]
  26. Schneeweiss, S. Sensitivity analysis and external adjustment for unmeasured confounders in epidemiologic database studies of therapeutics. Pharmacoepidemiol. Drug Saf. 2006, 15, 291–303. [Google Scholar] [CrossRef]
  27. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
  28. Kalogeropoulos, A.; Georgiopoulou, V.; Psaty, B.M.; Rodondi, N.; Smith, A.L.; Harrison, D.G.; Liu, Y.; Hoffmann, U.; Bauer, D.C.; Newman, A.B.; et al. Inflammatory markers and incident heart failure risk in older adults: The Health ABC (Health, Aging, and Body Composition) study. J. Am. Coll Cardiol. 2010, 55, 2129–2137. [Google Scholar] [CrossRef]
  29. Tsutamoto, T.; Hisanaga, T.; Wada, A.; Maeda, K.; Ohnishi, M.; Fukai, D.; Mabuchi, N.; Sawaki, M.; Kinoshita, M. Interleukin-6 spillover in the peripheral circulation increases with the severity of heart failure, and the high plasma level of interleukin-6 is an important prognostic predictor in patients with congestive heart failure. J. Am. Coll. Cardiol. 1998, 31, 391–398. [Google Scholar] [CrossRef]
  30. Meijers, W.C.; Maglione, M.; Bakker, S.J.L.; Oberhuber, R.; Kieneker, L.M.; de Jong, S.; Haubner, B.J.; Nagengast, W.B.; Lyon, A.R.; van der Vegt, B.; et al. Heart Failure Stimulates Tumor Growth by Circulating Factors. Circulation 2018, 138, 678–691. [Google Scholar] [CrossRef]
  31. Bertero, E.; Ameri, P.; Maack, C. Bidirectional Relationship Between Cancer and Heart Failure: Old and New Issues in Cardio-oncology. Card Fail Rev. 2019, 5, 106–111. [Google Scholar] [CrossRef] [PubMed]
  32. Ausoni, S.; Azzarello, G. Development of Cancer in Patients With Heart Failure: How Systemic Inflammation Can Lay the Groundwork. Front. Cardiovasc. Med. 2020, 7, 598384. [Google Scholar] [CrossRef]
  33. Gbelcová, H.; Švéda, M.; Laubertová, L.; Varga, I.; Vítek, L.; Kolář, M.; Strnad, H.; Zelenka, J.; Böhmer, D.; Ruml, T. The effect of simvastatin on lipid droplets accumulation in human embryonic kidney cells and pancreatic cancer cells. Lipids Health Dis. 2013, 12, 126. [Google Scholar] [CrossRef] [PubMed]
  34. Pinchuk, T.V.; Fedulaev, Y.N.; Khairetdinova, G.A.; Denisova, N.N.; Chura, O.V.; Logunova, I.Y. Anti-inflammatory effects of simvastatin in patients with chronic heart failure. Bull. Exp. Biol. Med. 2014, 157, 552–554. [Google Scholar] [CrossRef] [PubMed]
  35. Cao, T.-T.; Chen, H.-H.; Dong, Z.; Xu, Y.-W.; Zhao, P.; Guo, W.; Wei, H.-C.; Zhang, C.; Lu, R. Stachydrine Protects Against Pressure Overload-Induced Cardiac Hypertrophy by Suppressing Autophagy. Cell Physiol. Biochem. 2017, 42, 103–114. [Google Scholar] [CrossRef]
  36. Sleijfer, S.; van der Gaast, A.; Planting, A.S.; Stoter, G.; Verweij, J. The potential of statins as part of anti-cancer treatment. Eur. J. Cancer 2005, 41, 516–522. [Google Scholar] [CrossRef]
  37. Takemoto, M.; Liao, J.K. Pleiotropic effects of 3-hydroxy-3-methylglutaryl coenzyme a reductase inhibitors. Arterioscler Thromb Vasc Biol. 2001, 21, 1712–1719. [Google Scholar] [CrossRef]
  38. Freed-Pastor, W.A.; Mizuno, H.; Zhao, X.; Langerød, A.; Moon, S.-H.; Rodriguez-Barrueco, R.; Barsotti, A.; Chicas, A.; Li, W.; Polotskaia, A.; et al. Mutant p53 disrupts mammary tissue architecture via the mevalonate pathway. Cell 2012, 148, 244–258. [Google Scholar] [CrossRef]
  39. Wong, W.W.; Dimitroulakos, J.; Minden, M.D.; Penn, L.Z. HMG-CoA reductase inhibitors and the malignant cell: The statin family of drugs as triggers of tumor-specific apoptosis. Leukemia 2002, 16, 508–519. [Google Scholar] [CrossRef]
  40. Rao, S.; Porter, D.C.; Chen, X.; Herliczek, T.; Lowe, M.; Keyomarsi, K. Lovastatin-mediated G1 arrest is through inhibition of the proteasome, independent of hydroxymethyl glutaryl-CoA reductase. Proc. Natl. Acad. Sci. USA 1999, 96, 7797–7802. [Google Scholar] [CrossRef]
  41. Zhu, B.; Qu, S. The Relationship Between Diabetes Mellitus and Cancers and Its Underlying Mechanisms. Front. Endocrinol. 2022, 13, 800995. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, C.-H.; Huang, C.-W.; Nguyen, P.A.; Lin, M.-C.; Yeh, C.-Y.; Islam, M.; Rahmanti, A.R.; Yang, H.-C. Chemopreventive Effects of Concomitant or Individual Use of Statins, Aspirin, Metformin, and Angiotensin Drugs: A Study Using Claims Data of 23 Million Individuals. Cancers 2022, 14, 1211. [Google Scholar] [CrossRef]
  43. Egom, E.E.A.; Hafeez, H. Biochemistry of Statins. Adv. Clin. Chem. 2016, 73, 127–168. [Google Scholar]
  44. Kapadia, S.B.; Chisari, F.V. Hepatitis C virus RNA replication is regulated by host geranylgeranylation and fatty acids. Proc. Natl. Acad. Sci. USA 2005, 102, 2561–2566. [Google Scholar] [CrossRef] [PubMed]
  45. Syed, G.H.; Amako, Y.; Siddiqui, A. Hepatitis C virus hijacks host lipid metabolism. Trends Endocrinol. Metab. 2010, 21, 33–40. [Google Scholar] [CrossRef] [PubMed]
  46. Dorobantu, C.; Macovei, A.; Lazar, C.; Dwek, R.A.; Zitzmann, N.; Branza-Nichita, N. Cholesterol depletion of hepatoma cells impairs hepatitis B virus envelopment by altering the topology of the large envelope protein. J. Virol. 2011, 85, 13373–13383. [Google Scholar] [CrossRef] [PubMed]
  47. Xia, Y.; Xie, Y.; Yu, Z.; Xiao, H.; Jiang, G.; Zhou, X.; Yang, Y.; Li, X.; Zhao, M.; Li, L.; et al. The Mevalonate Pathway Is a Druggable Target for Vaccine Adjuvant Discovery. Cell 2018, 175, 1059–1073 e21. [Google Scholar] [CrossRef]
  48. Bader, T.; Korba, B. Simvastatin potentiates the anti-hepatitis B virus activity of FDA-approved nucleoside analogue inhibitors in vitro. Antivir. Res. 2010, 86, 241–245. [Google Scholar] [CrossRef]
  49. Zou, B.; Odden, M.C.; Nguyen, M.H. Statin Use and Reduced Hepatocellular Carcinoma Risk in Patients with Nonalcoholic Fatty Liver Disease. Clin. Gastroenterol. Hepatol. 2023, 21, 435–444.e6. [Google Scholar] [CrossRef]
  50. Zhong, G.-C.; Liu, Y.; Ye, Y.-Y.; Hao, F.-B.; Wang, K.; Gong, J.-P. Meta-analysis of studies using statins as a reducer for primary liver cancer risk. Sci. Rep. 2016, 6, 26256. [Google Scholar] [CrossRef]
  51. Nezasa, K.-I.; Higaki, K.; Matsumura, T.; Inazawa, K.; Hasegawa, H.; Nakano, M.; Koike, M. Liver-specific distribution of rosuvastatin in rats: Comparison with pravastatin and simvastatin. Drug Metab. Dispos. 2002, 30, 1158–1163. [Google Scholar] [CrossRef] [PubMed]
  52. Efthimiadis, A. Rosuvastatin and cardiovascular disease: Did the strongest statin hold the initial promises? Angiology 2008, 59 (Suppl 2), 62S–64S. [Google Scholar] [CrossRef] [PubMed]
  53. Jones, P.H.; Davidson, M.H.; A Stein, E.; E Bays, H.; McKenney, J.M.; Miller, E.; A Cain, V.; Blasetto, J.W. Comparison of the efficacy and safety of rosuvastatin versus atorvastatin, simvastatin, and pravastatin across doses (STELLAR* Trial). Am. J. Cardiol. 2003, 92, 152–160. [Google Scholar] [CrossRef] [PubMed]
  54. Paoletti, R.; Fahmy, M.; Mahla, G.; Mizan, J.; Southworth, H. Rosuvastatin demonstrates greater reduction of low-density lipoprotein cholesterol compared with pravastatin and simvastatin in hypercholesterolaemic patients: A randomized, double-blind study. J. Cardiovasc Risk 2001, 8, 383–390. [Google Scholar] [CrossRef]
  55. Grundy, S.M.; Stone, N.J.; Bailey, A.L.; Beam, C.; Birtcher, K.K.; Blumenthal, R.S.; Braun, L.T.; De Ferranti, S.; Faiella-Tommasino, J.; Forman, D.E.; et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 139, e1082–e1143. [Google Scholar]
  56. de Silva, R.; Nikitin, N.P.; Bhandari, S.; Nicholson, A.; Clark, A.L.; Cleland, J.G. Atherosclerotic renovascular disease in chronic heart failure: Should we intervene? Eur. Heart J. 2005, 26, 1596–1605. [Google Scholar] [CrossRef]
  57. Li, M.; Cima, M.J.; Milner, D.A., Jr. If It’s Not One Thing, It’s Another: An Inverse Relationship of Malignancy and Atherosclerotic Disease. PLoS ONE 2015, 10, e0126855. [Google Scholar] [CrossRef]
  58. Chen, W.; Zheng, R.; Zhang, S.; Zeng, H.; Zuo, T.; Xia, C.; Yang, Z.; He, J. Cancer incidence and mortality in China in 2013: An analysis based on urbanization level. Chin. J. Cancer Res. 2017, 29, 1–10. [Google Scholar] [CrossRef]
Figure 1. Data selection flowchart. During 2001 to 2012, there were 45,153 patients with HF diagnoses enrolled in this study. Patients diagnosed with HF without subsequent outpatient or inpatient visits, aged less than 40 years, or with pre-existing cancer or a history of statins prescription within 6 months before the date of enrollment were excluded. A total of 25,853 patients were enrolled in the study cohort.
Figure 1. Data selection flowchart. During 2001 to 2012, there were 45,153 patients with HF diagnoses enrolled in this study. Patients diagnosed with HF without subsequent outpatient or inpatient visits, aged less than 40 years, or with pre-existing cancer or a history of statins prescription within 6 months before the date of enrollment were excluded. A total of 25,853 patients were enrolled in the study cohort.
Cancers 15 02959 g001
Figure 2. Free of liver cancer survival rate in HF patients in Taiwan (n = 25,853) from 1 January 2001 to 31 December 2012 stratified according to statin use status (log–rank test, χ2 = 123.135; df = 1; p < 0.001).
Figure 2. Free of liver cancer survival rate in HF patients in Taiwan (n = 25,853) from 1 January 2001 to 31 December 2012 stratified according to statin use status (log–rank test, χ2 = 123.135; df = 1; p < 0.001).
Cancers 15 02959 g002
Figure 3. Free of liver cancer survival rate in HF patients in Taiwan (n = 25,853) from 1 January 2001 to 31 December 2012 stratified according to statin use dose status (log–rank test, χ2 = 126.632; df = 3; p < 0.001).
Figure 3. Free of liver cancer survival rate in HF patients in Taiwan (n = 25,853) from 1 January 2001 to 31 December 2012 stratified according to statin use dose status (log–rank test, χ2 = 126.632; df = 3; p < 0.001).
Cancers 15 02959 g003
Table 1. Characteristic of the sample population.
Table 1. Characteristic of the sample population.
Whole Cohort
(n = 25,853)
Patients with Statin Use (≥28 Days; n = 7364)Patients without Statin Use (<28 Days; n = 18,489)p a
n%n%n%
Age, years (Mean ± SD)69.71 (13.18)65.86 (11.15)71.25 (12.58)<0.001
  40–54389215.05142819.39246413.33<0.001
  55–64454517.58177624.12276914.98
  65–74766529.65250534.02516027.91
  ≥75975137.72165522.47809643.79
Gender
  Female13,40951.87412656.03928350.21<0.001
  Male12,44448.13323843.97920649.79
CCI +
  0556221.51182524.78373720.21<0.001
  1649925.14199727.12450224.35
  2541120.93152320.68388821.03
  ≥3838132.42201927.42636234.41
Diabetes
  No17,12666.24446160.5812,66568.50<0.001
  Yes872733.76290339.42582431.50
Hypertension
  No687826.60172523.42515327.87<0.001
  Yes18,97573.40563976.5813,33672.13
Dyslipidemia
  No17,35867.14405755.0913,30171.94<0.001
  Yes849532.86330744.91518828.06
Non-statin lipid-lowering drugs
  <28 days22,90988.61542773.7017,48294.55<0.001
  28–365 days20277.84127417.307534.07
  >365 days9173.556639.002541.37
Metformin
  <28 days20,06177.60450261.1415,55984.15<0.001
  28–365 days22038.5282311.1813807.46
  >365 days358913.88203927.6915508.38
RAA
  <28 days725728.0795713.00630034.07<0.001
  28–365 days690626.71152120.65538529.13
  >365 days11,69045.22488666.35680436.80
Aspirin
  <28 days10,70641.41170123.10900548.70<0.001
  28–365 days673726.06187325.43486426.31
  >365 days841032.53379051.47462024.99
Level of Urbanization
  Urban16,52163.90494367.1211,57862.62<0.001
  Suburban593522.96154220.94439323.76
  Rural339713.1487911.94251813.62
Monthly income (TWD)
  0327512.6779110.74248413.44<0.001
  1–21,000637924.67156721.28481226.03
  21,000–33,30010,33739.98288839.22744940.29
  ≥33,301586222.67211828.76374420.25
Hepatitis B/C
  No23,09089.31671091.1216,38088.59<0.001
  Yes276310.696548.88210911.41
a Comparison between non-statin and statin users; CCI +: Charlson comorbidity index.
Table 2. Distribution of statin use.
Table 2. Distribution of statin use.
Whole Cohort
(n = 25,853)
n%
Total cohort
  Never used17,97369.52
  ≤28 days of statin use5162.00
  >28 days of statin use736428.48
Ever statin users
  Single statin users438555.65
  Sequential multiple statins users349544.35
Statin type ever prescribed a
  Simvastatin221028.05
  Lovastatin120015.23
  Atorvastatin426454.11
  Fluvastatin119715.19
  Pravastatin101712.91
  Rosuvastatin222428.22
a The sum is more than 100% due to sequential multiple statin prescriptions in 44.35% of the ever-used subjects.
Table 3. Risk of liver cancer among statin and non-statin users in the study cohort.
Table 3. Risk of liver cancer among statin and non-statin users in the study cohort.
All Group
(n = 25,853)
Patients without Statin Use
(Total Follow-Up 85,867.4 Person–Years)
Patients with Statin Use
(Total Follow-Up 51,080.5 Person–Years)
Adjusted HR
(95% CI)
No. of
Patients
with Liver Cancer
Incidence Rate
(per 105 Person–Years)
(95% CI)
No. of
Patients
with Liver Cancer
Incidence Rate
(per 105 Person–Years)
(95% CI)
Whole cohort
  Liver Cancer441513.6(465.6, 561.5)71139.0(106.7, 171.3)0.26(0.20, 0.33) ***
Age, 40–64 a
  Liver Cancer153537.0(451.9, 622.1)30126.9(81.5, 172.3)0.23(0.15, 0.34) ***
Age, 65–74 b
  Liver Cancer160590.7(499.2, 682.2)31171.6(111.2, 232.0)0.30(0.20, 0.44) ***
Age, ≥75 c
  Liver Cancer128422.6(349.4, 495.8)10106.6(40.5, 172.7)0.25(0.13, 0.48) ***
Female d
  Liver Cancer202447.2(385.5, 508.8)33110.0(72.5, 147.5)0.25(0.17, 0.36) ***
Male e
  Liver Cancer239587.3(512.9, 661.8)38180.3(123.0, 237.6)0.27(0.19, 0.38) ***
***: p < 0.001.a Total follow-up 28,492.4 and 23,637.0 person–year for patients without statin use and patients with statin use. b Total follow-up 27,086.6 and 18,062.3 person–year for patients without statin use and patients with statin use. c Total follow-up 30,288.4 and 9381.2 person–year for patients without statin use and patients with statin use. d Total follow-up 45,173.6 and 30,001.8 person–year for patients without statin use and patients with statin use. e Total follow-up 40,693.7 and 21,078.7 person–year for patients without statin use and patients with statin use. C.I.: confidence interval. HR: hazard ratio. The main model is adjusted for age, sex, Charlson comorbidity index score, diabetes, hypertension, dyslipidemia, level of urbanization, and monthly income in propensity score.
Table 4. Incidence rate and adjusted HRs of liver cancer associated with statin use during the follow-up period in HF patients.
Table 4. Incidence rate and adjusted HRs of liver cancer associated with statin use during the follow-up period in HF patients.
VariableNo. of
Patients
No. of Person-YearsNo. of Patients with
Liver Cancer
Incidence Rate
(per 105 Person–Years)
(95% CI)
Adjusted HR (95% CI)p-Value for
Trend
Total statin use
  Non-user (<28 cDDDs)18,48985,867.4441513.6(465.6, 561.5)1.00<0.001
  User (≥28 cDDDs)736451,080.571139.0(106.7, 171.3)0.26(0.20, 0.33) ***
  28–90 cDDDs14308530.119222.7(122.6, 322.9)0.42(0.27, 0.67) ***
  91–365 cDDDs229214,038.924171.0(102.6, 239.3)0.32(0.21, 0.48) ***
  >365 cDDDs364228,511.52898.2(61.8, 134.6)0.18(0.12, 0.26) ***
Lipophilia statin use
  Non-user (<28 cDDDs)19,55891,882.1448487.6(442.4, 532.7)1.00<0.001
  User (≥28 cDDDs)629545,065.864142.0(107.2, 176.8)0.34(0.26, 0.44) ***
  28–90 cDDDs13768485.919223.9(123.2, 324.6)0.49(0.31, 0.78) **
  91–365 cDDDs215014,083.920142.0(79.8, 204.2)0.33(0.21, 0.52) ***
  >365 cDDDs276922,495.925111.1(67.6, 154.7)0.27(0.18, 0.41) ***
Hydrophilia statin use
  Non-user (<28 cDDDs)18,48985,867.4488568.3(517.9, 618.7)1.00<0.001
  User (≥28 cDDDs)298621,378.324112.3(67.3, 157.2)0.42(0.28, 0.64) ***
  28–90 cDDDs7795168.57135.4(35.1, 235.8)0.47(0.22, 1.00)
  91–365 cDDDs10867493.39120.1(41.6, 198.6)0.46(0.24, 0.90) *
  >365 cDDDs11218716.5891.8(28.2, 155.4)0.35(0.17, 0.71) **
Individual statin use
(≥28 cDDDs)
  Simvastatin221017,280.422127.3 (74.1, 180.5)0.51(0.33, 0.79) **
  Lovastatin120010,016.918179.7 (96.7, 262.7)0.70(0.43, 1.13)
  Atorvastatin426430,556.439127.6 (87.6, 167.7)0.39(0.28, 0.55) ***
  Fluvastatin11979276.513140.1 (64.0, 216.3)0.63(0.36, 1.10)
  Pravastatin10177827.813166.1 (75.8, 256.4)0.73(0.41, 1.27)
  Rosuvastatin222415,714.81489.1 (42.4, 135.8)0.34(0.20, 0.58) ***
* p < 0.05, ** p < 0.01, *** p < 0.001.The covariates in the main model were adjusted for the propensity scores for age, sex, Charlson comorbidity index score, diabetes, hypertension, dyslipidemia, level of urbanization, and monthly income. Lipophilia statins include simvastatin, lovastatin, atorvastatin, and fluvastatin. Hydrophilia statins include pravastatin and rosuvastatin. The HRs of individual statin users (≥28 cDDDs) were compared with the total statin use of non-users (<28 cDDDs).
Table 5. Sensitivity analysis of adjusted HRs of statins in the reduction of liver cancer risk.
Table 5. Sensitivity analysis of adjusted HRs of statins in the reduction of liver cancer risk.
Statin Usep for Trend
<28 cDDDs28–90 cDDDs91–365 cDDDs>365 cDDDs
Adjusted HR
(95% CI)
Adjusted HR
(95% CI)
Adjusted HR
(95% CI)
Adjusted HR
(95% CI)
Main model 1.000.42 (0.27, 0.67) ***0.32 (0.21, 0.48) ***0.18 (0.12, 0.26) ***<0.001
Additional covariates
  Main model + Non-statin 1.000.43 (0.27, 0.69) ***0.34 (0.22, 0.51) ***0.19 (0.13, 0.29) ***<0.001
  Main model + Metformin1.000.41 (0.26, 0.65) ***0.31 (0.20, 0.47) ***0.18 (0.12, 0.27) ***<0.001
  Main model + RAA1.000.44 (0.28, 0.69) ***0.37 (0.24, 0.56) ***0.23 (0.15, 0.34) ***<0.001
  Main model + Aspirin1.000.45 (0.29, 0.72) ***0.37 (0.24, 0.56) ***0.22 (0.15, 0.33) ***<0.001
Subgroup effects
  Age, years
    40–641.000.21 (0.08, 0.57) **0.23 (0.11, 0.47) ***0.23 (0.14, 0.38) ***<0.001
    65–741.000.49 (0.25, 0.97) **0.46 (0.26, 0.81) ***0.15 (0.08, 0.30) ***<0.001
    ≥751.000.72 (0.32, 1.63)0.24 (0.08, 0.75) *0.05 (0.01, 0.37) **<0.001
  Sex
    Female1.000.48 (0.26, 0.89) *0.24 (0.12, 0.48) ***0.17 (0.10, 0.30) ***<0.001
    Male1.000.36 (0.18, 0.73) **0.39 (0.23, 0.66) ***0.18 (0.11, 0.31) ***<0.001
  CCI +
    01.000.30 (0.07, 1.23)0.41 (0.16, 1.02)0.32 (0.15, 0.65) **<0.001
    11.000.14 (0.04, 0.58) **0.22 (0.09, 0.54) ***0.13 (0.06, 0.29) ***<0.001
    21.000.46 (0.19, 1.13)0.35 (0.16, 0.76) **0.13 (0.05, 0.32) ***<0.001
    ≥31.000.63 (0.33, 1.21)0.30 (0.14, 0.63) **0.17 (0.08, 0.36) ***<0.001
  Diabetes
    No1.000.53 (0.31, 0.90) *0.37 (0.22, 0.63) ***0.17 (0.10, 0.30) ***<0.001
    Yes1.000.25 (0.10, 0.62) **0.23 (0.12, 0.46) ***0.16 (0.09, 0.29) ***<0.001
  Dyslipidemia
    No1.000.35 (0.19, 0.66) **0.28 (0.16, 0.50) ***0.18 (0.11, 0.30) ***<0.001
    Yes1.000.54 (0.27, 1.06)0.36 (0.20, 0.66) ***0.17 (0.10, 0.31) ***<0.001
  Hypertension
    No1.000.42 (0.17, 1.04)0.39 (0.17, 0.89) *0.31 (0.16, 0.62) ***<0.001
    Yes1.000.42 (0.25, 0.72) **0.30 (0.18, 0.48) ***0.14 (0.09, 0.23) ***<0.001
  Hepatitis B/C
    No1.000.55 (0.29, 1.04)0.42 (0.24, 0.74) **0.20 (0.11, 0.35) ***<0.001
    Yes1.000.38 (0.19, 0.73) **0.29 (0.16, 0.53) ***0.28 (0.17, 0.48) ***<0.001
  Non-Statin
    <28 days1.000.39 (0.23, 0.66) ***0.33 (0.21, 0.53) ***0.17 (0.11, 0.27) ***<0.001
    28–365 days 1.000.86 (0.32, 2.33)0.46 (0.17, 1.23)0.28 (0.12, 0.66) **0.002
    >365 days1.00 0.22 (0.03, 1.97)0.29 (0.07, 1.33) 0.123
  Metformin
    <28 days1.000.50 (0.30, 0.84) **0.37 (0.23, 0.62) ***0.15 (0.08, 0.27) ***<0.001
    28–365 days 1.000.22 (0.05, 0.90) *0.19 (0.06, 0.63) **0.16 (0.05, 0.51) **<0.001
    >365 days1.000.27 (0.07, 1.11)0.27 (0.11, 0.67) **0.22 (0.12, 0.41) ***<0.001
  RAA
    <28 days1.000.68 (0.33, 1.39)0.53 (0.25, 1.13)0.17 (0.06, 0.55) **<0.001
    28–365 days 1.000.27 (0.10, 0.73) **0.26 (0.10, 0.63) **0.27 (0.12, 0.61) **<0.001
    >365 days1.000.45 (0.21, 0.97) *0.38 (0.21, 0.69) **0.23 (0.14, 0.38) ***<0.001
  Aspirin
    <28 days1.000.47 (0.22, 0.99) *0.32 (0.14, 0.71) **0.20 (0.09, 0.46) ***<0.001
    28–365 days 1.000.52 (0.25, 1.06)0.42 (0.21, 0.84) *0.24 (0.11, 0.53) ***<0.001
    >365 days1.000.36 (0.13, 0.99) *0.38 (0.19, 0.76) **0.23 (0.13, 0.40) ***<0.001
*: p < 0.05, **: p < 0.01, ***: p < 0.001. HR: hazard ratio. + CCI: Charlson Comorbidity Index. The main model is adjusted for age, sex, Charlson comorbidity index, diabetes, hypertension, dyslipidemia, level of urbanization, and monthly income in propensity score. The models were adjusted for covariates in the main model as well as each additional listed covariate.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lu, M.-C.; Chen, C.-C.; Lu, M.-Y.; Lin, K.-J.; Chiu, C.-C.; Yang, T.-Y.; Fang, Y.-A.; Jian, W.; Chen, M.-Y.; Hsu, M.-H.; et al. The Association between Statins and Liver Cancer Risk in Patients with Heart Failure: A Nationwide Population-Based Cohort Study. Cancers 2023, 15, 2959. https://doi.org/10.3390/cancers15112959

AMA Style

Lu M-C, Chen C-C, Lu M-Y, Lin K-J, Chiu C-C, Yang T-Y, Fang Y-A, Jian W, Chen M-Y, Hsu M-H, et al. The Association between Statins and Liver Cancer Risk in Patients with Heart Failure: A Nationwide Population-Based Cohort Study. Cancers. 2023; 15(11):2959. https://doi.org/10.3390/cancers15112959

Chicago/Turabian Style

Lu, Meng-Chuan, Chun-Chao Chen, Meng-Ying Lu, Kuan-Jie Lin, Chun-Chih Chiu, Tsung-Yeh Yang, Yu-Ann Fang, William Jian, Ming-Yao Chen, Min-Huei Hsu, and et al. 2023. "The Association between Statins and Liver Cancer Risk in Patients with Heart Failure: A Nationwide Population-Based Cohort Study" Cancers 15, no. 11: 2959. https://doi.org/10.3390/cancers15112959

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop