Skip to main content
JACC: Advances logoLink to JACC: Advances
. 2025 Oct 21;4(11):102259. doi: 10.1016/j.jacadv.2025.102259

Statin Use in Patients With Cancer

Drug Interaction and Statin Usage

Emily Chou a,, Carlo S Legasto b, Alan K Chin b, Alan H Baik c, Brian C Schulte d
PMCID: PMC12589975  PMID: 41124987

Abstract

Background

Cardiovascular disease and cancer share common risk factors. Statins, which are widely prescribed for cardiovascular disease prevention, have potential for drug-drug interactions (DDIs) with oncology agents.

Objectives

This study quantifies and characterizes interactions between statins and recently approved oncology agents. Additionally, the study evaluates statin prescribing trends in patients with cancer.

Methods

Food and Drug Administration oncology drug approvals from June 2019 to June 2024 were screened for interactions with 5 commonly prescribed statins using UpToDate Lexidrug. Interaction severity was classified into 5 risk levels. Statin prescribing rates, stratified by cancer type, were characterized through a scoping review of 20 PubMed-indexed observational studies.

Results

Of 138 oncology agents, 33 (23.9%) exhibited DDIs with statins. Simvastatin demonstrated the highest interaction rate (22% [30/138] of oncology drugs), while pravastatin had the fewest (4% [5/138] oncology drugs). Most interactions (88%, 71/81) were classified as level C, requiring monitoring. Contraindicated interactions (level X) were identified for adagrasib, tucatinib, and asciminib. Across the 23,3774 patients in the included studies, statin use was most common in prostate (53%, 57,926/109,140 of patients) and lung cancer (32%, 1403/4344), and least common in liver (8%, 3100/39,722) and breast cancer (10%, 1519/15,078).

Conclusions

Statin-oncology agent DDIs are relatively common but rarely is statin use contraindicated. Frequency of statins use varies according to cancer type. These findings highlight the importance of individualized risk-benefit assessments that consider prognosis, cardiovascular risk, potential drug interactions, and patient preferences to guide statin use in cancer patients.

Key words: cancer survivorship, cardiotoxicity, comorbidities, drug interactions, hyperlipidemia, statin safety

Central Illustration

graphic file with name ga1.jpg


The relationship between cardiovascular disease (CVD) and cancer has been well characterized.1,2 CVD and cancer have been associated with some common risk factors such as age, obesity, hypertension, diabetes mellitus, and smoking.2, 3, 4, 5 Ideal cardiovascular health, as defined by the American Heart Association, is inversely associated with incident cancer.6 Exogenous cholesterol has been found to accumulate in cancer tissue, with increased dietary cholesterol associated with incident cancer.7, 8, 9, 10 Furthermore, cardiac remodeling may potentiate cancer progression, suggesting early diagnosis and treatment of cardiac disease may improve cancer treatment outcomes.11

Statins are commonly prescribed for primary and secondary prevention of CVD and major adverse cardiovascular events. The time to benefit for primary prevention of major adverse cardiovascular events per 100 adults aged 50 to 75 years old is over 2 years.12 Previous studies have assessed the risks and benefits of discontinuing statin therapy in the setting of life-limiting disease.13,14 A study in patients with life expectancy of less than 1 year and Karnofsky Performance Status of <80% found that patients who discontinued statin therapy had better quality of life and saved $716.46 per patient over 212.6 days.13 Nonetheless, statins are often prescribed and filled by patients with life-limiting illnesses.15,16 The recognition of a life-limiting illness does not necessarily influence reduction in statin use in the last 6 months of life15 and one study reported that 31% of patients with cancer filled a statin prescription within 30 days of death.16

In contrast, several cancer-directed treatments, such as androgen deprivation therapies, antiestrogen therapies, anthracyclines, and tyrosine kinase inhibitors, are associated with hyperlipidemia.1 Therefore, patients with longer life-expectancies would likely benefit from continued lipid monitoring and appropriate statin therapy to reduce cholesterol levels and mitigate CVD and cancer progression.

However, drug-drug interactions (DDIs) are a significant concern in oncology. Polypharmacy is prevalent among cancer patients, who commonly receive complex medication regimens including systemic cancer-directed therapy, supportive therapies, and medications for chronic conditions. For example, a study of Southwest Oncology Group clinical trials found that 28.7% of subjects had DDIs classified as moderate or higher.17 Some oncologic agents interact with critical pathways in statin metabolism (for example through CYP3A4 metabolism); however, the extent and distribution of these interactions remain incompletely characterized. Even modest reductions in first-pass statin metabolism or CYP3A4 inhibition can greatly increase systemic statin levels, elevating the risk of myopathy and life-threatening rhabdomyolysis.18 Conversely, interactions with statins may directly and indirectly impact efficacy and toxicity profiles of cancer-directed agents.19 These considerations are crucial given the narrow therapeutic indices of many oncologic therapies and the increased risk of adverse drug effects.19

Therefore, this study aims to quantify and characterize interactions between statins and Food and Drug Administration (FDA)-approved oncologic agents approved between 2019 and 2024 and to examine frequency of use of statin in cancer patients.

Methods

Statin and oncology drug interactions

FDA oncology drug approvals from June 27, 2019, to June 27, 2024, were indexed from the FDA's website on oncology and hematologic malignancies approval notifications.20 This time frame was selected at the time of project initiation to represent recently approved oncologic agents. Two hundred seven approvals for 138 unique oncology agents were identified. DDIs between oncology agents and 5 most prescribed statins—rosuvastatin, atorvastatin, simvastatin, pravastatin, and lovastatin—were evaluated using UpToDate Lexidrug,21,22 a widely used and comprehensive tool for identifying DDIs. DDI severities were recorded according to the risk ratings designated by Lexidrug (Table 1).23

Table 1.

Lexidrug Online Interaction Risk Rating Categories and Descriptions

Risk Rating Action Description
A No interaction Data have not demonstrated either pharmacodynamic or pharmacokinetic interactions between the specified agents
B No action needed Data demonstrate that the specified agents may interact with each other, but there is little to no evidence of clinical concern resulting from their concomitant use.
C Monitor therapy Data demonstrate that the specified agents may interact with each other in a clinically significant manner. The benefits of concomitant use of these 2 medications usually outweigh the risks. An appropriate monitoring plan should be implemented to identify potential negative effects. Dosage adjustments of one or both agents may be needed in a minority of patients.
D Modify regimen Data demonstrate that the 2 medications may interact with each other in a clinically significant manner. A patient-specific assessment must be conducted to determine whether the benefits of concomitant therapy outweigh the risks. Specific actions must be taken to realize the benefits and/or minimize the toxicity resulting from concomitant use of the agents. These actions may include aggressive monitoring, empiric dosage changes, choosing alternative agents.
X Avoid combination Data demonstrate that the specified agents may interact with each other in a clinically significant manner. The risks associated with concomitant use of these agents usually outweigh the benefits. These agents are generally considered contraindicated.

Scoping review of statin prescribing patterns across cancer types

A scoping review was performed to characterize statin prescribing rates across patients diagnosed with different cancer types. This scoping review is reported in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis Statement Extension for Scoping Reviews guidelines (Figure 1).24 Screening and data extraction were conducted. While predefined inclusion and exclusion criteria were used to minimize subjectivity, the absence of duplicate independent review may introduce selection and extraction bias. A single database search was considered adequate given the descriptive nature of this scoping review. PubMed was searched in September 2024 with the following criteria: “(statin OR rosuvastatin OR atorvastatin OR pravastatin OR fluvastatin OR lovastatin OR simvastatin OR HMG-CoA reductase inhibitor) AND (adenocarcinoma OR cancer OR carcinoma OR oncology OR sarcoma OR leukemia OR lymphoma OR malignancy OR tumor OR melanoma OR myeloma OR neoplasm OR neuroblastoma OR dysplasia).” Exclusion criteria included systematic reviews, clinical trials, review articles, case reports, nonhuman subjects, and unrelated articles. Inclusion criteria included observational studies pertaining to statin use in patients with cancer. The review was limited to English-language articles with no restriction on publication year, up to September 2024.

Figure 1.

Figure 1

Preferred Reporting Items for Systematic Review and Meta-Analysis Extension for Scoping Reviews Flow Diagram

The process of identifying, screening, and selecting studies for inclusion in the scoping review.

Studies meeting inclusion criteria were included in syntheses based on availability of relevant data. The extracted data included the type of cancer, number of patients prescribed statins, and total number of patients with the specific cancer diagnosis. Additional variables including age, cancer stage, statin type, treatment duration, DDIs, and statin indication were sought post hoc but were inconsistently reported. Data preparation involved aggregating patient counts by cancer type to calculate the proportion of statin users within each category; no additional data transformation was performed. Because no means or variance measures were reported, statistical pooling and meta-analysis were not performed, making SDs not applicable and thus not reported. Included studies were assessed for quality using the Newcastle-Ottawa Scale (Supplemental Table 2).25 Given the descriptive nature of this scoping review without meta-analysis, no formal assessment of reporting bias or certainty of evidence was performed. This scoping review was not prospectively registered, and no protocol was prepared prior to conducting the review.

Statistical analysis

All analyses were performed using Microsoft Excel. Summary data are presented as percentages. Chi-square tests were used to assess differences in in the frequency of oncology drug-statin interactions stratified by statin type, compare rates of statin use among different cancer types, and compare rates of statin use across cancer stages. Statistical significance was defined as a 2-sided P value <0.05.

Ethical approval

This study did not involve the use of human or animal subjects and therefore did not require review or approval by an Institutional Review Board or ethics committee.

Results

Statin and oncology agent interactions

Out of 138 approved oncology agents, 33 were found to have any degree of interaction with rosuvastatin, atorvastatin, simvastatin, pravastatin, and lovastatin (Figure 2). Agents approved for the treatment of non–small cell lung cancer (NSCLC) had the highest number of interactions at 9 out of 26 agents. Agents approved for the treatment of prostatic adenocarcinoma had the highest proportion of interactions, at 7 out of 10 agents. Other indications with notable interactions included lymphoma (5 out of 18), breast cancer (4 out of 17), leukemia (2 out of 15), multiple myeloma (3 out of 13), agnostic indications (3 out of 11), and colorectal cancer (CRC) (3 out of 10).

Figure 2.

Figure 2

FDA-Approved Oncology Drugs per Indication and Interactions With Commonly Prescribed Statins

207 FDA approvals for oncology drugs from June 27, 2019, to June 27, 2024, were recorded and categorized by their specific indications. DDIs between each oncology agent and the top 5 most prescribed statins—rosuvastatin, atorvastatin, simvastatin, pravastatin, and lovastatin—were evaluated using UpToDate Lexi-Drug. Labels indicate the number of approved drugs with any level of interaction with a statin vs the total number of approved drugs for each indication. DDIs = drug-drug interactions; FDA = Food and Drug Administration; GIST = gastrointestinal stromal tumor; NSCLC = non–small cell lung cancer; SCLC = small cell lung cancer.

Interactions with specific statins were quantified as well (Figure 3). There was a statistically significant difference in interaction rates among the 5 statins (P < 0.001). Simvastatin had the highest interaction rate, affecting 30 of 138 recently approved oncology agents (22%), while pravastatin had the fewest interactions, with 5 of 138 agents (4%).

Figure 3.

Figure 3

Interactions Between Statins and Recently Approved Oncology drugs

Interactions between the 5 most commonly prescribed statins and oncology drugs approved from 2019 to 2024 were quantified. Abbreviation as in Figure 2.

The nature of interactions was characterized using risk ratings designated by Lexidrug. Only pharmacokinetic or pharmacodynamic DDIs were evaluated. Of 81 interactions between statins and oncology drugs, 71 (87%) were level C interactions, 6 (7%) were level X, 2 (2%) were level B, and 2 (2%) were level D (Figure 4A). Although drugs approved for prostatic adenocarcinoma had the highest rates of interactions, there was 1 level B, 18 level C, and 1 level D interactions (Figure 4B). Drugs approved for lymphoma exhibited 8 level C interactions and drugs approved for agnostic indications had 7 level C interactions. Agents approved for NSCLC had 1 level B, 21 level C, 1 level D, and 2 level X interactions. Similarly, breast cancer agents exhibited 9 level C interactions and 2 level X interactions. CRC agents had 7 level C interactions and 4 level X interactions (Figure 4B).

Figure 4.

Figure 4

Figure 4

Risk Ratings of Interactions Between Statins and Approved Oncology Drugs

(A) Distribution of risk ratings for all interactions between statins and approved oncology drugs. (B) 6 indications were identified to have approved agents with the most interactions with statin drugs. Risk ratings were recorded based on Lexidrug categories as described in Table 1.

Level X interactions involved adagrasib approved for NSCLC and CRC, tucatinib for breast cancer and CRC, and asciminib for chronic myeloid leukemia (CML). Notably, only simvastatin and lovastatin interacted with the included oncology agents at level X severity, apart from asciminib which also interacts with rosuvastatin and atorvastatin with level X severity (Supplemental Table 1).

Rates of statin use in patients with cancer

A scoping review was performed to assess rates of statin use based on cancer type. Initial PubMed search identified 9800 studies, of which 24 were removed as duplicates or retracted studies. Eighty-eight abstracts were identified as observational studies and were eligible for title and abstract review. Twenty-five were selected for full-text review; 5 were excluded, leaving 20 studies included in this analysis (Figure 1).26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45 Study details are delineated in Table 2. Additional characteristics are reported in Supplemental Tables 2 to 4. The studies were drawn from diverse sources, including national health databases, cancer registries, observational cohorts, and single institutions across several countries. Across the 20 included studies, prostate cancer was the most commonly reported cancer type (10 studies), followed by colorectal (9), lung (6), and breast (5). Conversely, certain cancers such as thyroid, sarcoma, biliary, and endometrial cancers were underrepresented. Of the 22 cancer types included, rates of statin use differed significantly across cancer types (P < 0.001). Fifteen cancer diagnoses were associated with rates of statin use of 20% or higher. Statin use was highest in patients with prostate and lung cancer (53% and 32%, respectively) (Figure 5). These were followed by esophageal and kidney cancer (31% and 30%). Patients with hepatocellular carcinoma used statins at the lowest rate. Four studies reported specific statins prescribed (Supplemental Table 4). Atorvastatin (619 of 1855 total patients), simvastatin (568), and pravastatin (272) were the most frequently used. Lovastatin (195), fluvastatin (119), rosuvastatin (28), and pitavastatin (9) were less commonly reported.

Table 2.

Characteristics of Observational Studies With Rates of Statin Use in Cancer Patients

First Author, Year Data Source Cancer Type Statin Users Statin Nonusers % Statin Users
Lötsch et al,25 2014 Vienna Cancer and Thrombosis Study at the Medical University of Vienna, Austria, 2003-2011 Brain 20 162 11.0%
Breast 18 181 9.0%
Colorectal 14 132 9.6%
Gastric 6 51 10.5%
Kidney 7 33 17.5%
Lung 38 177 17.7%
Lymphoma 17 220 7.2%
Multiple myeloma 3 35 7.9%
Pancreas 8 77 9.4%
Prostate 27 121 18.2%
Chan et al,26 2015 Prostate cancer patients from the Health Professionals Follow-Up Study, 1992-2008 Prostate 685 3,264 17.3%
Shao et al,27 2015 National Health Insurance claims database and cancer registry databases of The Collaboration Center of Health Information Application, Taiwan. Hepatocellular carcinoma patients treated 2001-2010 and followed to 2012 Liver 1988 18,212 9.8%
Chen et al,28 2016 Brain cancer patients from the National Health Insurance Bureau in Taiwan, 2004-2011 Brain 32 213 13.1%
Wang et al,29 2016 Women’s Health Initiative Observational Study, recruited 1993-1998 Nonmelanoma skin cancer 1,529 10,026 13.2%
Wu et al,30 2016 Taiwan National Health Insurance Research Data linked to the Taiwan Cancer Registry, 2001-2012 Liver 934 17,958 4.9%
Mikkelsen et al,31 2017 Prostate cancer patients at 2 Danish Urological Departments, 2007-2013 Prostate 141 396 26.3%
Palumbo et al,32 2017 Prostate cancer patients at the University of Perugia, 2009-2014 Prostate 55 195 22.0%
Emilsson et al,33 2018 Surveillance, Epidemiology, and End Results (SEER)-Medicare database (2007-2009) Bladder 49 819 5.6%
Breast 289 5,547 5.0%
Colorectal 192 3,491 5.2%
Prostate 367 5,471 6.3%
Anderson-Carter et al,34 2019 Prostate Cancer patients in the national Veterans Affairs database, 2000-2008 and followed to 2016 Prostate 53,360 33,986 61.1%
Fransgaard et al,35 2019 National Clinical Registry of the Danish Colorectal Cancer Group, 2003-2015 Colorectal 386 1861 17.2%
Jiménez-Vacas et al,36 2020 Prostate cancer patients from the Reina Sofia University Hospital Prostate 30 45 40.0%
Majidi et al,37 2020 Ovarian Cancer Prognosis and Lifestyle study, Australian women diagnosed with ovarian cancer from 2012-2015 and followed for 5-8 years Ovarian 200 755 20.9%
Fernandez et al,38 2021 Head and neck squamous cell carcinoma patients from Walter Reed National Military Medical Center, the University of Rochester Medical Center, and an observational clinical study conducted by the National Institutes of Health in partnership with Johns Hopkins University Head and neck 113 164 40.8%
Okada et al,39 2021 Follow-up observational study of the Japan Primary Prevention of Atherosclerosis with Aspirin for Diabetes trial Breast 3 14 17.6%
Colorectal 14 44 24.1%
Gastric 9 37 19.6%
Liver 1 23 4.2%
Lung 2 3 40.0%
Pancreas 6 16 27.3%
Prostate 4 16 20.0%
Rossi et al,40 2021 Patients with metastatic non–small cell lung cancer at S. Andrea Hospital Sapienza Rome University, 2015-2020 Lung 88 74 54.3%
Chung et al,41 2022 Taiwan National Health Insurance Research Database, 2001-2008 Colorectal 362 956 27.5%
Pourlotfi et al, 2022 Swedish Colorectal Cancer Register, 2007-2016 Colorectal 5,896 13,222 30.84%
Okamoto et al,43 2023 Colorectal cancer patients at the University of Tokyo Hospital Colorectal 55 195 22.0%
Lin et al,44 2024 United Kingdom Biobank, recruited 2006-2010 and followed up to 2021 Biliary 75 218 25.6%
Bladder 370 791 31.9%
Brain 167 612 21.4%
Breast 1,209 7,817 13.4%
Colorectal 1,423 4,275 25.0%
Endometrial 293 1,234 19.2%
Esophageal 308 695 30.7%
Gastric 198 492 28.7%
Head and neck 245 722 25.3%
Kidney 467 1,069 30.4%
Leukemia 318 981 24.5%
Liver 177 429 29.2%
Lung 1,275 2,687 32.2%
Lymphoma 522 1828 22.2%
Melanoma 555 2,294 19.5%
Multiple myeloma 200 645 23.7%
Ovarian 145 821 15.0%
Pancreas 375 878 29.9%
Prostate 3,257 7,720 29.7%
Sarcoma 98 314 23.8%
Thyroid 87 348 20.0%

Figure 5.

Figure 5

Rates of Statin Use in Cancer Patients by Cancer Types

A scoping review was performed to identify observational studies with available data on statin use rates in cancer patients.

Okada et al explicitly assessed primary prevention, while others did not differentiate between primary and secondary atherosclerotic cardiovascular disease (ASCVD) prevention. Duration of statin use was also largely not specified. Chan et al reported 214 patients with prostatic adenocarcinoma used statins for 1 to <6 years and 217 for >6 years, while Wang et al reported 417 patients with skin cancer, used statins for <1 year, 558 for 1 to <3 years, 291 for 3 to <5 years, and 263 for >5 years. DDIs were not reported in any of the included studies.

Discussion

Managing statin-oncology drug interactions

We demonstrate the rate of interactions between statins and cancer-directed agents approved between 2019 and 2024 is 23.9% (33 of 138) (Figure 2). These interactions are not homogenous across statin types. Pravastatin exhibited the lowest interaction rates, while simvastatin demonstrated the highest rate of interaction (Figure 3). Statin use was prominent in prostate and lung cancer patients and least in liver and breast cancer patients (Figure 5). These findings underscore the importance of individualizing statin therapy based on prognosis, cardiovascular risk, and DDIs.

Contraindicated combinations between statins and oncology drugs are rare, with 7% of interactions identified as contraindicated combinations (Figure 4A). Eighty-eight percent of statin-oncology DDIs were assigned risk ratings of C, meaning the benefits of concomitant therapy outweigh the risks and patients should be monitored for clinically significant interactions (Figure 4A).

In instances where statin efficacy may be compromised due to interactions, switching to an alternative agent or attempting dose reduction is reasonable rather than discontinuing therapy altogether. Lorlatinib was approved for the treatment of anaplastic lymphoma kinase- or c-ros oncogene 1-positive NSCLC based on the CROWN Trial (NCT03052608), but 72% of patients developed hypercholesterolemia with the majority necessitating statin treatment.46 Lorlatinib, a moderate cytochrome P450 3A4 (CYP3A4) inducer, may ameliorate the efficacy of statins metabolized by CYP3A4 pathways. Conversely, adagrasib and tucatinib, strong CYP3A4 inhibitors, may elevate serum concentrations of statins, increasing risk of serious adverse events.47 In these cases, an alternative statin can be used that avoids CYP3A4 metabolism such as fluvastatin, rosuvastatin, pitavastatin, or pravastatin. Nonstatin lipid-lowering medications such as ezetimibe and proprotein convertase subtilisin/kexin type 9 inhibitors can be considered, as ezetimibe is metabolized through glucuronidation and proprotein convertase subtilisin/kexin type 9 inhibitors are eliminated through lysosomal degradation, thus having a lower likelihood for DDIs.48,49

Though not validated in patients with cancer specifically, the 10-year ASCVD risk calculator could be used to assist decision-making on statin use for primary prevention.50 Statins carry a risk of rhabdomyolysis, which is also associated with multiple anticancer agents, necessitating cautious statin selection and monitoring.51,52 Conversely, several anticancer drugs, including anthracyclines and immunotherapies, are associated with cardiotoxicity, where statin therapy may offer protective benefits.53, 54, 55

For patients with CVD risk factors and malignancies with an indolent course, statin continuation may be of significant benefit. Survival in patients with CML is particularly lengthy with tyrosine kinase inhibitors such as imatinib. Long-term treatment with imatinib resulted in a 10-year median overall survival (mOS) of 83.3%.56 Patients receiving treatment for CML are expected to lose <3 life-years.57 Asciminib, approved for Philadelphia chromosome-positive CML, is contraindicated in combination with rosuvastatin and atorvastatin and demonstrates level C interaction with other statins (Supplemental Table 1).58 However, considering the life expectancy of CML patients, alternate statins should be continued based on estimated CVD risk.1

Rates of statin use across cancer types

Patients with prostate cancer exhibit the highest rate of statin use at 53%. This is likely in part due to overlapping prevalence of prostate cancer and dyslipidemia in older males.59 Furthermore, androgen deprivation therapies may elevate low density lipoprotein and triglyceride levels, increase visceral and subcutaneous fat, and promote insulin resistance, contributing to heightened ASCVD risk.60, 61, 62 Current management of these adverse effects includes high-intensity statin therapy and aggressive management of CVD risk factors.63

Agents prescribed for prostate cancer demonstrate the highest rates of interaction with statins (70%) (Figure 2). However, all interactions were of grade C or lower, barring the grade D interaction between darolutamide and rosuvastatin. Darolutamide is approved for castration-resistant prostatic adenocarcinoma, with mOS exceeding 3 years.59,64 Given the mOS and risk of CVD in this population, the benefits of continuing statin therapy frequently exceed the risks of potential DDIs, particularly in those with preexisting CVD.

Statin use is also prevalent in lung cancer patients, at 32%. Smoking increases the risk of both lung cancer and CVD mortality, and in one longitudinal study, patients with lung cancer were found to have 2 to 4 times increased risk of CVD.65,66 In contrast, patients with hepatocellular carcinoma exhibited the lowest rate of statin use at 8%, possibly attributed to concerns of hepatotoxicity with statins, and the relatively poor prognosis compared to other malignancies.67

Limitations and future directions

Despite the valuable insights provided by this study, several limitations should be acknowledged. The scoping review was conducted solely using PubMed, which may have limited the breadth of literature retrieved compared to a multidatabase search. DDIs were assessed using only UpToDate Lexi-Drug, which may not capture all potential interactions or emerging evidence from other sources. Additionally, this review was not prospectively registered, reflecting its exploratory scope. The review was conducted by a single reviewer, which may introduce bias in study selection and data extraction. This study is based on observational data, which may introduce selection bias and potentially overestimate statin use among cancer patients. The focus on FDA-approved oncology drugs from the past 5 years was intended to capture current therapies but may have excluded older treatments with relevant interactions. Finally, some findings are derived from pooled data with relatively small sample sizes, which may affect the precision and generalizability of the results. Only 1 study explicitly assessed statin use for primary prevention of ASCVD; the remaining studies did not distinguish between use for primary and secondary prevention.40 Five papers reported statin use stratified by cancer stage (Supplemental Table 3). Additionally, heterogeneous age reporting across studies limits comparison of age effects on statin use (Supplemental Table 1). These limitations highlight the need for cautious interpretation and further research.

Conclusions

In this study, we demonstrate that statin-oncology agent DDIs are relatively common; however, statin use is rarely contraindicated. Frequency of statin use differs in patients with various types of cancers. These findings highlight the importance of individualized risk-benefit assessments when deciding upon statin use in cancer patients (Central Illustration).

Perspectives.

COMPETENCY IN MEDICAL KNOWLEDGE: In patients with cancer on statin medications, only a small subset of DDIs are considered strict contraindications and most can be managed with alternative statin selection or dose modifications.

TRANSLATIONAL OUTLOOK: Further research is needed to better understand the indications of statin therapy in patients with cancer and the absolute reduction in cardiovascular events and disease progression.

Central Illustration.

Central Illustration

Statins in Oncology Patients

Analysis of DDIs between statins and recently approved oncology agents, assessing severity and distribution across cancer types. Of 138 FDA-approved oncology agents, 33 (24%) exhibited DDIs with statins, with most interactions requiring monitoring. Simvastatin showed the highest interaction rate. Statin prescribing was highest in prostate and lung cancer, and lowest in liver and breast cancer. Findings suggest that DDIs between statins and oncology drugs are uncommon and rarely contraindicated, supporting the continuation of statins in cancer patients when clinically appropriate. CVD= cardiovascular disease; other abbreviations as in Figure 2.

Funding support and author disclosures

Dr Baik has received consulting and advising fees from Kiniksa Pharmaceuticals. All other authors have reported that they have no relationships relevant to the contents of this paper to disclose.

Footnotes

The authors attest they are in compliance with human studies committees and animal welfare regulations of the authors’ institutions and Food and Drug Administration guidelines, including patient consent where appropriate. For more information, visit the Author Center.

Appendix

For a supplemental table, please see the online version of this paper.

Supplementary material

Supplemental Material
mmc1.docx (52.9KB, docx)

References

  • 1.de Jesus M., Mohammed T., Singh M., Tiu J.G., Kim A.S. Etiology and management of dyslipidemia in patients with cancer. Front Cardiovasc Med. 2022;9 doi: 10.3389/fcvm.2022.892335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Aparecida Silveira E., Vaseghi G., de Carvalho Santos A.S., et al. Visceral obesity and its shared role in cancer and cardiovascular disease: a scoping review of the pathophysiology and pharmacological treatments. Int J Mol Sci. 2020;21(23):9042. doi: 10.3390/ijms21239042. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Koene R.J., Prizment A.E., Blaes A., Konety S.H. Shared risk factors in cardiovascular disease and cancer. Circulation. 2016;133(11):1104–1114. doi: 10.1161/CIRCULATIONAHA.115.020406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Wolin K.Y., Carson K., Colditz G.A. Obesity and cancer. Oncologist. 2010;15(6):556–565. doi: 10.1634/theoncologist.2009-0285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Islami F., Moreira D.M., Boffetta P., Freedland S.J. A systematic review and meta-analysis of tobacco use and prostate cancer mortality and incidence in prospective cohort studies. Eur Urol. 2014;66(6):1054–1064. doi: 10.1016/j.eururo.2014.08.059. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Rasmussen-Torvik L.J., Shay C.M., Abramson J.G., et al. Ideal cardiovascular health is inversely associated with incident cancer: the atherosclerosis risk in communities study. Circulation. 2013;127(12):1270–1275. doi: 10.1161/CIRCULATIONAHA.112.001183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Murai T. Cholesterol lowering: role in cancer prevention and treatment. Biol Chem. 2015;396(1):1–11. doi: 10.1515/hsz-2014-0194. [DOI] [PubMed] [Google Scholar]
  • 8.Jowett M. The phosphatide and cholesterol contents of normal and malignant human tissues. Biochem J. 1931;25(6):1991–1998. doi: 10.1042/bj0251991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Hu J., La Vecchia C., de Groh M., Negri E., Morrison H., Mery L. Dietary cholesterol intake and cancer. Ann Oncol. 2012;23(2):491–500. doi: 10.1093/annonc/mdr155. [DOI] [PubMed] [Google Scholar]
  • 10.Riscal R., Bull C.J., Mesaros C., et al. Cholesterol auxotrophy as a targetable vulnerability in clear cell renal cell carcinoma. Cancer Discov. 2021;11(12):3106–3125. doi: 10.1158/2159-8290.CD-21-0211. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Avraham S., Abu-Sharki S., Shofti R., et al. Early cardiac remodeling promotes tumor growth and metastasis. Circulation. 2020;142(7):670–683. doi: 10.1161/CIRCULATIONAHA.120.046471. [DOI] [PubMed] [Google Scholar]
  • 12.Yourman L.C., Cenzer I.S., Boscardin W.J., et al. Evaluation of time to benefit of statins for the primary prevention of cardiovascular events in adults aged 50 to 75 years: a meta-analysis. JAMA Intern Med. 2021;181(2):179–185. doi: 10.1001/jamainternmed.2020.6084. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kutner J.S., Blatchford P.J., Taylor D.H.J., et al. Safety and benefit of discontinuing Statin therapy in the setting of advanced, life-limiting illness: a randomized clinical trial. JAMA Intern Med. 2015;175(5):691–700. doi: 10.1001/jamainternmed.2015.0289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Rea F., Biffi A., Ronco R., et al. Cardiovascular outcomes and mortality associated with discontinuing statins in older patients receiving polypharmacy. JAMA Netw Open. 2021;4(6) doi: 10.1001/jamanetworkopen.2021.13186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Stavrou E.P., Buckley N., Olivier J., Pearson S.A. Discontinuation of statin therapy in older people: does a cancer diagnosis make a difference? An observational cohort study using data linkage. BMJ Open. 2012;2(3) doi: 10.1136/bmjopen-2012-000880. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Silveira M.J., Kazanis A.S., Shevrin M.P. Statins in the last six months of life: a recognizable, life-limiting condition does not decrease their use. J Palliat Med. 2008;11(5):685–693. doi: 10.1089/jpm.2007.0215. [DOI] [PubMed] [Google Scholar]
  • 17.van Leeuwen R.W.F., Jansman F.G.A., van den Bemt P.M.L.A., et al. Drug-drug interactions in patients treated for cancer: a prospective study on clinical interventions. Ann Oncol. 2015;26(5):992–997. doi: 10.1093/annonc/mdv029. [DOI] [PubMed] [Google Scholar]
  • 18.Wiggins B.S., Saseen J.J., Page R.L., et al. Recommendations for management of clinically significant drug-drug interactions with statins and select agents used in patients with cardiovascular disease: a scientific statement from the American heart association. Circulation. 2016;134(21):e468–e495. doi: 10.1161/CIR.0000000000000456. [DOI] [PubMed] [Google Scholar]
  • 19.Del Re M., Roncato R., Argentiero A., et al. Clinical relevance and methodological approach for the assessment of drug-drug interactions in cancer patients: a position statement from the Italian association of medical oncology (AIOM) and the Italian society of pharmacology (SIF) ESMO Open. 2025;10(6) doi: 10.1016/j.esmoop.2025.105119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.U.S. Food and Drug Administration Oncology (Cancer)/Hematologic malignancies approval notifications. https://www.fda.gov/drugs/resources-information-approved-drugs/oncology-cancerhematologic-malignancies-approval-notifications
  • 21.Matyori A., Brown C.P., Ali A., Sherbeny F. Statins utilization trends and expenditures in the U.S. before and after the implementation of the 2013 ACC/AHA guidelines. Saudi Pharm J. 2023;31(6):795–800. doi: 10.1016/j.jsps.2023.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lexi-Interact Drug Interactions [Internet] Hudson (OH): UpToDate, Inc. 2025. https://www.uptodate.com/drug-interactions
  • 23.Lexi-interact data fields. Lexicomp. http://webstore.lexi.com/Information/Product-Information/Lexi-Interact-Fields#:∼:text=Severity%20indicators%20include:%20Minor%20(effects,may%20be%20included%20as%20well
  • 24.Page M.J., McKenzie J.E., Bossuyt P.M., et al. The PRISMA 2020 statement: an updated guideline for reporting systematic reviews. BMJ. 2021;372 doi: 10.1136/bmj.n71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Wells G., Shea B., O’Connel . The Newcastle-Ottawa Scale (NOS) for Assessing the Quality of Nonrandomised Studies in Meta-Analyses. University of Ottawa; 2000. https://www.ohri.ca/programs/clinical_epidemiology/oxford.asp [Google Scholar]
  • 26.Lötsch F., Königsbrügge O., Posch F., Zielinski C., Pabinger I., Ay C. Statins are associated with low risk of venous thromboembolism in patients with cancer: a prospective and observational cohort study. Thromb Res. 2014;134(5):1008–1013. doi: 10.1016/j.thromres.2014.09.001. [DOI] [PubMed] [Google Scholar]
  • 27.Chan J.M., Kenfield S.A., Paciorek A., Platz E.A., Giovannucci E.L., Stampfer M.J. Postdiagnostic statin use and the risk of lethal prostate cancer in the health professionals Follow-up study. Cancer Epidemiol Biomarkers Prev. 2015;24(10):1638–1640. doi: 10.1158/1055-9965.EPI-15-0671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Shao J.Y.H., Lee F.P., Chang C.L., Wu S.Y. Statin-based palliative therapy for hepatocellular carcinoma. Medicine (Baltimore) 2015;94(42) doi: 10.1097/MD.0000000000001801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Chen B.K., Chiu H.F., Yang C.Y. Statins are associated with a reduced risk of brain cancer: a population-based case-control study. Medicine (Baltimore) 2016;95(17) doi: 10.1097/MD.0000000000003392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Wang A., Stefanick M.L., Kapphahn K., et al. Relation of statin use with non-melanoma skin cancer: prospective results from the Women’s health initiative. Br J Cancer. 2016;114(3):314–320. doi: 10.1038/bjc.2015.376. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Wu L.L., Hsieh M.C., Chow J.M., Liu S.H., Chang C.L., Wu S.Y. Statins improve outcomes of nonsurgical curative treatments in hepatocellular carcinoma patients. Medicine (Baltimore) 2016;95(36) doi: 10.1097/MD.0000000000004639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Mikkelsen M.K., Thomsen F.B., Berg K.D., et al. Associations between statin use and progression in men with prostate cancer treated with primary androgen deprivation therapy. Scand J Urol. 2017;51(6):464–469. doi: 10.1080/21681805.2017.1362032. [DOI] [PubMed] [Google Scholar]
  • 33.Palumbo I., Matrone F., Montesi G., et al. Statins protect against acute RT-related rectal toxicity in patients with prostate cancer: an observational prospective study. Anticancer Res. 2017;37(3):1453–1457. doi: 10.21873/anticanres.11469. [DOI] [PubMed] [Google Scholar]
  • 34.Emilsson L., García-Albéniz X., Logan R.W., Caniglia E.C., Kalager M., Hernán M.A. Examining bias in studies of statin treatment and survival in patients with cancer. JAMA Oncol. 2018;4(1):63–70. doi: 10.1001/jamaoncol.2017.2752. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Anderson-Carter I., Posielski N., Liou J.I., et al. The impact of statins in combination with androgen deprivation therapyin patients with advanced prostate cancer: a large observational study. Urol Oncol. 2019;37(2):130–137. doi: 10.1016/j.urolonc.2018.11.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Fransgaard T., Hallas J., Thygesen L.C., Gögenur I. Association of statin use and oncological outcomes after neoadjuvant radiotherapy in patients with rectal cancer. Anticancer Res. 2019;39(4):2177–2182. doi: 10.21873/anticanres.13332. [DOI] [PubMed] [Google Scholar]
  • 37.Jiménez-Vacas J.M., Herrero-Aguayo V., Montero-Hidalgo A.J., et al. Clinical, cellular, and molecular evidence of the additive antitumor effects of biguanides and statins in prostate cancer. J Clin Endocrinol Metab. 2021;106(2):e696–e710. doi: 10.1210/clinem/dgaa877. [DOI] [PubMed] [Google Scholar]
  • 38.Majidi A., Na R., Jordan S.J., De Fazio A., Webb P.M. Statin use and survival following a diagnosis of ovarian cancer: a prospective observational study. Int J Cancer. 2021;148(7):1608–1615. doi: 10.1002/ijc.33333. [DOI] [PubMed] [Google Scholar]
  • 39.Fernandez K.A., Allen P., Campbell M., et al. Atorvastatin is associated with reduced cisplatin-induced hearing loss. J Clin Invest. 2021;131(1) doi: 10.1172/JCI142616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Okada S., Morimoto T., Ogawa H., et al. Association between statins and cancer incidence in diabetes: a cohort study of Japanese patients with type 2 diabetes. J Gen Intern Med. 2021;36(3):632–639. doi: 10.1007/s11606-020-06167-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Rossi A., Filetti M., Taurelli S.B., et al. Statins and immunotherapy: togetherness makes strength the potential effect of statins on immunotherapy for NSCLC. Cancer Rep (Hoboken) 2021;4(4) doi: 10.1002/cnr2.1368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Chung K.C., Juang S.E., Chen H.H., et al. Association between metabolic syndrome and colorectal cancer incidence and all-cause mortality: a hospital-based observational study. BMC Gastroenterol. 2022;22(1):453. doi: 10.1186/s12876-022-02505-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Pourlotfi A., Ahl Hulme R., Forssten M.P., et al. Statin therapy and its association with long-term survival after Colon cancer surgery. Surgery. 2022;171(4):890–896. doi: 10.1016/j.surg.2021.08.002. [DOI] [PubMed] [Google Scholar]
  • 44.Okamoto K., Nozawa H., Emoto S., Murono K., Sasaki K., Ishihara S. Does Statin suppress oxaliplatin-induced peripheral neuropathy in patients with colorectal cancer? A single-center observational study. Support Care Cancer. 2023;31(12):660. doi: 10.1007/s00520-023-08134-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Lin J.R., Han D.D., Wei W., et al. Regular use of aspirin and statins reduces the risk of cancer in individuals with systemic inflammatory diseases. Cancer Res. 2024;84(11):1889–1897. doi: 10.1158/0008-5472.CAN-23-2941. [DOI] [PubMed] [Google Scholar]
  • 46.Solomon B.J., Bauer T.M., Mok T.S.K., et al. Efficacy and safety of first-line lorlatinib versus crizotinib in patients with advanced, ALK-positive non-small-cell lung cancer: updated analysis of data from the phase 3, randomised, open-label CROWN study. Lancet Respir Med. 2023;11(4):354–366. doi: 10.1016/S2213-2600(22)00437-4. [DOI] [PubMed] [Google Scholar]
  • 47.Zhang J., Johnson M., Barve M., et al. Practical guidance for the management of adverse events in patients with KRASG12C-Mutated non-small cell lung cancer receiving adagrasib. Oncologist. 2023;28(4):287–296. doi: 10.1093/oncolo/oyad051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Kosoglou T., Statkevich P., Johnson-Levonas A.O., Paolini J.F., Bergman A.J., Alton K.B. Ezetimibe: a review of its metabolism, pharmacokinetics and drug interactions. Clin Pharmacokinet. 2005;44(5):467–494. doi: 10.2165/00003088-200544050-00002. [DOI] [PubMed] [Google Scholar]
  • 49.Horton J.D., Cohen J.C., Hobbs H.H. Molecular biology of PCSK9: its role in LDL metabolism. Trends Biochem Sci. 2007;32(2):71–77. doi: 10.1016/j.tibs.2006.12.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Grundy S.M., Stone N.J., Bailey A.L., et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA guideline on the management of blood cholesterol: executive summary: a report of the American college of cardiology/American heart association task force on clinical practice guidelines. J Am Coll Cardiol. 2019;73(24):3168–3209. doi: 10.1016/j.jacc.2018.11.002. [DOI] [PubMed] [Google Scholar]
  • 51.Mendes P., Robles P.G., Mathur S. Statin-induced rhabdomyolysis: a comprehensive review of case reports. Physiother Can. 2014;66(2):124–132. doi: 10.3138/ptc.2012-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kurnik M., Kolar Kus P., Krepek M., Vlaović J., Podbregar M. Fatal rhabdomyolysis and fulminant myocarditis with malignant arrhythmias after one dose of ipilimumab and nivolumab. Immunotherapy. Published online November 16, 2024;16:1203–1210. doi: 10.1080/1750743X.2024.2427563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Neilan T.G., Quinaglia T., Onoue T., et al. Atorvastatin for anthracycline-associated cardiac dysfunction: the STOP-CA randomized clinical trial. JAMA. 2023;330(6):528–536. doi: 10.1001/jama.2023.11887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Dadson K., Thavendiranathan P., Hauck L., et al. Statins protect against early stages of doxorubicin-induced cardiotoxicity through the regulation of Akt signaling and SERCA2. CJC Open. 2022;4(12):1043–1052. doi: 10.1016/j.cjco.2022.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Drobni Z.D., Gongora C., Taron J., et al. Impact of immune checkpoint inhibitors on atherosclerosis progression in patients with lung cancer. J Immunother Cancer. 2023;11(7) doi: 10.1136/jitc-2023-007307. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Hochhaus A., Larson R.A., Guilhot F., et al. Long-term outcomes of imatinib treatment for chronic myeloid leukemia. N Engl J Med. 2017;376(10):917–927. doi: 10.1056/NEJMoa1609324. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Bower H., Björkholm M., Dickman P.W., Höglund M., Lambert P.C., Andersson T.M.L. Life expectancy of patients with chronic myeloid leukemia approaches the life expectancy of the general population. J Clin Oncol. 2016;34(24):2851–2857. doi: 10.1200/JCO.2015.66.2866. [DOI] [PubMed] [Google Scholar]
  • 58.Réa D., Mauro M.J., Boquimpani C., et al. A phase 3, open-label, randomized study of asciminib, a STAMP inhibitor, vs bosutinib in CML after 2 or more prior TKIs. Blood. 2021;138(21):2031–2041. doi: 10.1182/blood.2020009984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Fizazi K., Shore N., Tammela T.L., et al. Darolutamide in nonmetastatic, castration-resistant prostate cancer. N Engl J Med. 2019;380(13):1235–1246. doi: 10.1056/NEJMoa1815671. [DOI] [PubMed] [Google Scholar]
  • 60.Hamilton E.J., Gianatti E., Strauss B.J., et al. Increase in visceral and subcutaneous abdominal fat in men with prostate cancer treated with androgen deprivation therapy. Clin Endocrinol (Oxf) 2011;74(3):377–383. doi: 10.1111/j.1365-2265.2010.03942.x. [DOI] [PubMed] [Google Scholar]
  • 61.Braga-Basaria M., Muller D.C., Carducci M.A., Dobs A.S., Basaria S. Lipoprotein profile in men with prostate cancer undergoing androgen deprivation therapy. Int J Impot Res. 2006;18(5):494–498. doi: 10.1038/sj.ijir.3901471. [DOI] [PubMed] [Google Scholar]
  • 62.Basaria S., Muller D.C., Carducci M.A., Egan J., Dobs A.S. Hyperglycemia and insulin resistance in men with prostate carcinoma who receive androgen-deprivation therapy. Cancer. 2006;106(3):581–588. doi: 10.1002/cncr.21642. [DOI] [PubMed] [Google Scholar]
  • 63.Bhatia N., Santos M., Jones L.W., et al. Cardiovascular effects of androgen deprivation therapy for the treatment of prostate cancer: ABCDE steps to reduce cardiovascular disease in patients with prostate cancer. Circulation. 2016;133(5):537–541. doi: 10.1161/CIRCULATIONAHA.115.012519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Smith M.R., Hussain M., Saad F., et al. Darolutamide and survival in metastatic, hormone-sensitive prostate cancer. N Engl J Med. 2022;386(12):1132–1142. doi: 10.1056/NEJMoa2119115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zhang S., Liu L., Shi S., et al. Bidirectional association between cardiovascular disease and lung cancer in a prospective cohort study. J Thorac Oncol. 2024;19(1):80–93. doi: 10.1016/j.jtho.2023.09.004. [DOI] [PubMed] [Google Scholar]
  • 66.Pope C.A., 3rd, Burnett R.T., Turner M.C., et al. Lung cancer and cardiovascular disease mortality associated with ambient air pollution and cigarette smoke: shape of the exposure-response relationships. Environ Health Perspect. 2011;119(11):1616–1621. doi: 10.1289/ehp.1103639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dal Maso L., Panato C., Tavilla A., et al. Cancer cure for 32 cancer types: results from the EUROCARE-5 study. Int J Epidemiol. 2020;49(5):1517–1525. doi: 10.1093/ije/dyaa128. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Material
mmc1.docx (52.9KB, docx)

Articles from JACC: Advances are provided here courtesy of Elsevier

RESOURCES