Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
December-2025 Volume 56 Issue 6

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Review Open Access

Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review)

  • Authors:
    • Zeyin He
    • Lili Zhang
    • Shiyi Gong
    • Xudan Yang
    • Guixuan Xu
  • View Affiliations / Copyright

    Affiliations: Department of Pathology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, P.R. China, Clinical Nutrition Section, Department of Laboratory Medicine, The Third People's Hospital of Chengdu, Chengdu, Sichuan 610014, P.R. China, Department of Nutrition and Food Hygiene, School of Public Health, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
    Copyright: © He et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 226
    |
    Published online on: October 17, 2025
       https://doi.org/10.3892/ijmm.2025.5667
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

Cholesterol and its metabolites exert multifaceted and profound effects on cancer initiation, progression and therapeutic response as well as patient prognosis. The present review systematically summarizes the oncogenic role of cholesterol metabolism in malignancies. Cancer cells extensively remodel cholesterol homeostasis through enhanced synthesis, increased uptake and impaired efflux, thereby sustaining proliferative signaling, suppressing ferroptotic cell death, promoting autophagic survival and facilitating epithelial‑mesenchymal transition, collectively fueling tumor invasion and metastasis. Within the tumor immune microenvironment, cholesterol exhibits dual immunoregulatory roles; it potentiates T‑cell antitumor function while its oxidized derivatives contribute to T‑cell exhaustion. Therapeutic targeting of cholesterol metabolism represents a promising strategy to trigger ferroptosis, reverse chemoresistance and reinvigorate antitumor immunity. Nevertheless, epidemiological evidence regarding the correlation between cholesterol levels and cancer risk remains contentious, underscoring the context‑dependent and complex nature of cholesterol in oncology. Targeting cholesterol metabolism may thus offer a novel integrative approach for cancer therapy, meriting further mechanistic and clinical investigation.

Introduction

Cholesterol is an essential component of mammalian cell membranes and is indispensable for maintaining normal cellular functions (1). The synthesis of cholesterol is maintained through a dynamic homeostatic process, with intracellular cholesterol levels being precisely regulated by a sophisticated feedback system involving synthesis, uptake, efflux, transport, esterification and enzymatic conversion (2). Cancer cells frequently exhibit an elevated demand for cholesterol to facilitate their growth (3). Specifically, they augment the uptake of exogenous cholesterol and lipoproteins and reprogram cholesterol metabolism by regulating genes related to cholesterol synthesis, efflux and intake to increase cholesterol influx and decrease efflux (4,5). Additionally, tumors can elevate cholesterol levels within the tumor microenvironment (TME) by stimulating cholesterol efflux from monocytes and macrophages via intercellular signaling (6). Furthermore, cholesterol serves as a precursor for biologically active metabolites, such as oxysterols (7), steroid hormones (8) and lipid rafts (LRs) (9), which collectively promote cancer cell proliferation, angiogenesis, metastasis and other pro-tumorigenic processes (10).

A previous study has shown that the crosstalk between cholesterol metabolism and the TME contributes to tumorigenesis and progression (11). Moreover, oncogenic signaling pathways (12), ferroptosis (13), autophagy (14), epithelial-mesenchymal transition (EMT) (15) and the immune response (16) are modulated through cholesterol metabolism. Current evidence indicates that targeting cholesterol metabolism, either alone or in combination with other strategies, to enhance cancer treatment efficacy has proven to be a viable antitumor approach. Notably, interventions targeting cholesterol metabolism enzymes [such as HMG-CoA reductase (HMGCR) (17), sterol regulatory element binding proteins (SREBPs) (18), squalene epoxidase (SQLE) (19) and acetyl-CoA acetyltransferase 1 (ACAT1) (20)] or transporters [ATP-binding cassette transporter A1 (ABCA1) (21), ATP-binding cassette transporter G1 (ABCG1) (22) and low-density lipoprotein receptor (LDLR) (23)] have shown considerable anticancer potential.

However, the association between cholesterol and cancer progression is currently controversial. Several epidemiological studies have shown that hypercholesterolemia and high cholesterol diets are associated with an increased risk of developing hepatocellular carcinoma (HCC) (24), colorectal cancer (CRC) (25), prostate cancer (PC) (26) and other cancer types (27). Consistently, statins and proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, which effectively lower serum cholesterol levels by targeting cholesterol metabolism genes, have been found to suppress cancer progression in certain settings (28). By contrast, other large cohort studies report that serum total cholesterol (TC) levels are either inversely correlated with cancer risk or show no significant association (29,30). For instance, a large-scale prospective Korean cohort study demonstrated that higher TC levels were negatively associated with mortality rates in HCC and gastric cancer (GC) (31). The role of dietary cholesterol in carcinogenesis is similarly debated. While high-cholesterol diets have been shown to accelerate HCC and CRC development (32), a recent study showed a significant positive association between higher daily dietary cholesterol intake and ovarian cancer (OV) risk, whereas abnormal lipid levels were not associated with the risk of OV (33). These conflicting epidemiological data underscore the complexity of the role of cholesterol in cancer and highlight the need for further mechanistic and population-level studies to clarify the relationship between serum cholesterol levels and cancer risk.

In the present review, the latest progress in the interaction between cholesterol and cancer progression is summarized, focusing on the functional roles of cholesterol and its derivatives within cancer cells and their reciprocal regulation within the TME. The current therapeutic strategies targeting molecules such as HMGCR, SREBPs and SQLE are also discussed, offering new perspectives for anticancer drug development.

Cholesterol metabolism

Cholesterol is an essential lipid molecule and a critical component for maintaining normal functions in the human body (1,34). Cholesterol fulfills multiple vital roles, including maintaining membrane integrity, facilitating cell signaling cell membrane structure, mediating cell communication, enhancing immunity and serving as a precursor for synthesizing steroids, sex hormones, vitamin D, bile salts and oxysterols (35-37). Cholesterol metabolites also hold significant value. In the liver, cholesterol is converted into bile acids, which are essential for the emulsification and absorption of dietary lipids and represent the primary route for eliminating excess cholesterol from the body (38,39). Moreover, cholesterol serves as the precursor for all steroid hormones (such as cortisol, aldosterone, estrogen and testosterone). In the skin, under ultraviolet radiation, cholesterol is transformed into a vitamin D precursor, which is subsequently activated to regulate calcium and phosphate homeostasis (40-42). Thus, in healthy individuals, the homeostasis of cholesterol metabolism is indispensable for cellular activity, digestion function, endocrine regulation and skeletal health.

The body acquires cholesterol through two primary pathways: Endogenous synthesis (70-80%) and exogenous dietary intake (20-30%) (43). Dietary cholesterol is absorbed by Niemann-Pick-C1 like-1 protein (NPC1L1) on the intestinal epithelial cell membrane and subsequently esterified by ACAT1/2, enabling its uptake by the liver in the form of chylomicrons (44). The liver esterifies cholesterol synthesized internally and absorbed from food, assembling it with apolipoproteins and other components into very low-density lipoproteins (VLDLs), which are subsequently secreted into the bloodstream. These VLDLs are metabolized into low-density lipoproteins (LDLs), which are taken up by peripheral tissues via LDLRs. Excess cholesterol in peripheral tissues is delivered back to the liver via high-density lipoprotein cholesterol (HDL-C)-mediated reverse cholesterol transport, where it can be recycled or converted to bile acids for excretion (1,45). Intracellular cholesterol levels are regulated through receptor-mediated endocytosis of LDL-C and HDL-C, as well as through de novo synthesis in the endoplasmic reticulum (ER) via the mevalonate (MVA) pathway, which includes squalene biosynthesis and modification procedures (34). Key regulatory enzymes in this pathway are HMGCR and SQLE, which catalyze the conversion of HMG-CoA to MVA and squalene to 2,3-epoxysqualene, respectively. Subsequent steps involve enzymes such as farnesyl diphosphate synthase (FDPS) and geranylgeranyl pyrophosphate synthase (GGPPs), which are crucial for the biosynthesis of farnesyl pyrophosphate (FPP) and geranylgeranyl pyrophosphate (GGPP). Under the action of squalene synthase and squalene epoxidase, FPP is converted to squalene and eventually to cholesterol (46).

Cholesterol homeostasis is transcriptionally regulated by SREBPs. When intracellular cholesterol levels are high, SREBPs are retained in the ER through interaction with the SREBP cleavage-activating protein/insulin-induced gene-1 (INSIG-1) complex. When the cholesterol level of the endoplasmic membrane decreases, SREBPs are transported to the Golgi apparatus. Following proteolytic processing, their transcriptionally active N-terminal domains are released (47). Excess intracellular cholesterol is either esterified by ACAT1 and stored in lipid droplets or refluxed from cells via transporters such as ABCA1 and ABCG1 (48). The dysregulation of cholesterol metabolic enzymes is implicated in various pathologies, including familial hypercholesterolemia (49), atherosclerosis (35) and Alzheimer's disease (50). In summary, cholesterol levels in normal cells are tightly controlled through a balance of synthesis, uptake, efflux, transport and esterification. A thorough understanding of these regulatory mechanisms is essential for elucidating the pathophysiology of cholesterol-related disorders (Fig. 1).

Cholesterol metabolism and cancer.
The major pathways involved in intracellular cholesterol metabolism
include biosynthesis, uptake, efflux, esterification and
conversion. Intracellular cholesterol levels are precisely
regulated through these pathways and cholesterol transport. (A)
De novo biosynthesis converts acetyl-CoA to cholesterol
through ~30 enzymatic steps, with HMGCR and SQLE serving as key
rate-limiting enzymes. HMGCR, a glycoprotein localized to the ER,
reduces HMG-CoA to MVA. SQLE, another rate-limiting enzyme
downstream of HMGCR in cholesterol biosynthesis, converts the
non-sterol intermediate squalene into 2,3-oxysqualene. (B) Besides
synthesis, cells acquire cholesterol via LDLR-mediated endocytosis
of LDL particles. LDL-derived cholesterol is transported via
endosomes and lysosomes, with the assistance of NPC1/2 and sterol
transfer proteins, to the plasma membrane and ER. Cholesterol can
be converted into CE by ACAT1 and stored in lipid droplets; it can
also be transformed into hydroxycholesterol, epoxycholesterol,
vitamin D and steroid hormones. Excess cholesterol in the cell is
secreted extracellularly via ABCA1/ABCG1. HMG-CoA,
3-hydroxy-3-methylglutaryl-CoA; HMGCS, HMG-CoA synthase; HMGCR,
3-hydroxy-3-methylglutaryl-CoA reductase; SQLE, squalene epoxidase;
MVA, mevalonate; IPP, isoprene unit isopentenyl diphosphate; FDPS,
farnesyl diphosphate synthase; FPP, farnesyl pyrophosphate; FDFT1,
farnesyl-diphosphate farnesyltransferase 1; LSS, lanosterol
synthase; LDL, low-density lipoprotein; LDLR, LDL receptor; SCAP,
sterol regulatory element binding protein cleavage-activating
protein; SR-B2, scavenger receptor class B protein; CE, cholesterol
ester; EE, early late endosome; LE, late endosome; Ly, lysosome;
ER, endoplasmic reticulum; ACAT1/2, acetyl-CoA acetyltransferase
1/2; NPC1/2, Niemann-Pick C1-like protein 1/2; ABCA1, ATP binding
cassette transporter A1; ABCG1, ATP-binding cassette transporter
G1.

Figure 1

Cholesterol metabolism and cancer. The major pathways involved in intracellular cholesterol metabolism include biosynthesis, uptake, efflux, esterification and conversion. Intracellular cholesterol levels are precisely regulated through these pathways and cholesterol transport. (A) De novo biosynthesis converts acetyl-CoA to cholesterol through ~30 enzymatic steps, with HMGCR and SQLE serving as key rate-limiting enzymes. HMGCR, a glycoprotein localized to the ER, reduces HMG-CoA to MVA. SQLE, another rate-limiting enzyme downstream of HMGCR in cholesterol biosynthesis, converts the non-sterol intermediate squalene into 2,3-oxysqualene. (B) Besides synthesis, cells acquire cholesterol via LDLR-mediated endocytosis of LDL particles. LDL-derived cholesterol is transported via endosomes and lysosomes, with the assistance of NPC1/2 and sterol transfer proteins, to the plasma membrane and ER. Cholesterol can be converted into CE by ACAT1 and stored in lipid droplets; it can also be transformed into hydroxycholesterol, epoxycholesterol, vitamin D and steroid hormones. Excess cholesterol in the cell is secreted extracellularly via ABCA1/ABCG1. HMG-CoA, 3-hydroxy-3-methylglutaryl-CoA; HMGCS, HMG-CoA synthase; HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase; SQLE, squalene epoxidase; MVA, mevalonate; IPP, isoprene unit isopentenyl diphosphate; FDPS, farnesyl diphosphate synthase; FPP, farnesyl pyrophosphate; FDFT1, farnesyl-diphosphate farnesyltransferase 1; LSS, lanosterol synthase; LDL, low-density lipoprotein; LDLR, LDL receptor; SCAP, sterol regulatory element binding protein cleavage-activating protein; SR-B2, scavenger receptor class B protein; CE, cholesterol ester; EE, early late endosome; LE, late endosome; Ly, lysosome; ER, endoplasmic reticulum; ACAT1/2, acetyl-CoA acetyltransferase 1/2; NPC1/2, Niemann-Pick C1-like protein 1/2; ABCA1, ATP binding cassette transporter A1; ABCG1, ATP-binding cassette transporter G1.

Cholesterol and cancer progression

Cholesterol metabolism in cancer cells

During growth and invasion, cancer cells require a continuous supply of cholesterol to sustain biosynthetic processes and cellular functions. Consequently, abnormal activation of cholesterol metabolism genes is commonly observed across multiple types of cancer (51). Cholesterol metabolism-related genes are upregulated, cholesterol influx is increased and cholesterol efflux is decreased. HMGCR is the primary rate-limiting enzyme in cholesterol biosynthesis, and its high expression in malignant tumors promotes tumor progression. For instance, HMGCR induces immunosuppression in OV by activating X-box binding protein 1 and programmed death-ligand 1 (PD-L1), correlating with poor prognosis (52). In PC, stromal HMGCR upregulation induced by coculture with malignant cells promotes tumor growth (53). HCC is an aggressive human cancer with increasing incidence worldwide (54). Research has revealed that HMGCR promotes tumor growth in a mouse model of primary liver cancer by enhancing cholesterol synthesis and activating the PDZ-binding motif/TEA domain transcription factors 2/anillin/kinesin family member 23 pathway (55). Additionally, HMGCR overexpression augments the growth and migration of GC cells (56), breast cancer (BC) cells (57) and glioblastoma (GBM) cells (58).

SREBPs, as key regulators of cholesterol metabolism (59), contribute to establishing energy and lipid reserves in tumor cells, playing a crucial role in tumor proliferation, stemness and drug resistance (60). In GBM cells, SREBP1 promotes lipid droplet autophagy by regulating key autophagy genes (such as autophagy-related protein 9B, autophagy-associated gene 4A and light chain 3B), leading to cholesterol ester hydrolysis and the release of cholesterol into lysosomes, ultimately promoting tumor growth (61). Patients with HCC have a poor prognosis and are prone to drug resistance (62). The liver is the primary organ for cholesterol metabolism, and abnormal cholesterol levels are closely associated with the progression of HCC. A previous study has revealed that SREBP1 enhances monounsaturated fatty acid synthesis by upregulating stearoyl-CoA desaturase 1, thereby suppressing ferroptosis in HCC cells and promoting xenograft tumor invasion and sorafenib resistance in mice (63). Notably, Su and Koeberle (64) conducted a systematic review on the role of SREBP1 in HCC progression, metastasis and drug resistance. The review mainly summarized that SREBP1 is an independent prognostic indicator for overall survival (OS) and disease-free survival in patients with HCC. SREBP1 can also regulate signaling pathways in tumor cells, including the PI3K-AKT-mTOR axis, protein kinase A (PKA), c-Myc and Janus kinase (JAK). Moreover, SREBPs have been shown to be upregulated in the tissues of patients with PC (65), BC (66) and OV (67).

SQLE, as the second rate-limiting enzyme downstream of HMGCR, is considered an oncogene that promotes carcinogenic signaling (68). It has been reported that SQLE could enhance the PI3K/AKT signaling pathway to promote distant metastasis in head and neck squamous cell carcinoma (69). In CRC, SQLE induces EMT by triggering the Wnt/β-catenin pathway, thereby promoting tumor metastasis (70). Moreover, SQLE, which is abnormally expressed in liver cancer models, induces immune suppression by promoting cholesterol accumulation in the TME and inhibiting CD8+ T cell function (71). Research has also shown that SQLE contributes to the resistance of BC cells to apoptosis by enhancing glycolysis (72). SQLE expression is specifically elevated in HCC and is strongly associated with poor clinical outcomes (73). SQLE significantly augments HCC growth in both in vitro and in vivo models, linked to the activation of serine-threonine kinase receptor associated protein-dependent transforming growth factor-β (TGF-β)/SMAD signaling pathways (74).

Farnesyl-diphosphate farnesyltransferase (FDFT1), another key enzyme in the cholesterol pathway, is dysregulated in various tumor types and represents a potential biomarker and therapeutic target (75). FDFT1 is highly expressed in HCC tissues and is correlated with poor patient prognosis (76,77). In HCC, FDFT1 suppressed fructose-1,6-bisphosphate aldolase B expression, thereby releasing its inhibitory control over the AKT signaling pathway and activating the PI3K/AKT pathway, ultimately promoting tumor growth and metastasis (76). Prognosis analysis showed that FDFT1, as a potential prognostic marker, was associated with shorter survival in patients with CRC and may partially contribute to the formation of a suppressive immune microenvironment (78). This indicates that the tumor-promoting effect of FDFT1 is dependent on the TME (79). Notably, another study has revealed a unique role of FDFT1 in CRC. This study found that under fasting conditions, SREBP2 upregulates FDFT1 expression, while elevated FDFT1 expression negatively regulates the AKT/mTOR/hypoxia-inducible factor-1α (HIF-1α) signaling pathway, thereby inhibiting glycolysis and proliferation in CRC cells (80). This suggests that FDFT1 may exert tumor-suppressive effects under specific metabolic conditions, such as fasting, indicating the complexity of its functional role.

ACAT1 is a multifunctional metabolic enzyme that plays a complex and sometimes seemingly contradictory role in various cancer types; it can suppress tumors under specific conditions while simultaneously promoting tumor progression through different mechanisms (81). In HCC, ACAT1-mediated acetylation of glycerophosphate O-acyltransferase (GNPAT) can stabilize GNPAT protein levels by antagonizing tripartite motif containing 21-catalyzed ubiquitination, ultimately promoting xenograft tumor growth (82). In bladder cancer (BLCA), ACAT1 promotes BLCA cell proliferation and invasion by activating the AKT/GSK3β/c-Myc signaling pathway (83). Additionally, a recent study has identified ACAT1 as a key factor in reducing the sensitivity of GBM to ferroptosis, with its mechanism of action dependent on the regulation of the iron efflux protein, solute carrier family 40 member 1 (84). However, under the influence of exogenous proinflammatory factors interleukin (IL)-12 and IL-18 proteins, ACAT1 is phosphorylated at the S60 site and translocates from mitochondria to the nucleus. Within the nucleus, it exerts its acetyltransferase activity, specifically acetylated the K146 site of the NF-κB family protein p50, thereby releasing its transcriptional repression on multiple immunochemical genes and natural killer (NK) cell activation ligands, thereby powerfully recruiting and activating NK cells. This enhances their tumor-killing capacity and inhibits tumor growth (85).

NPC1L1 is the primary protein responsible for the absorption of exogenous cholesterol. NPC1L1 is highly expressed in CRC and serves as an independent prognostic factor that is significantly correlated with pathological staging (86,87). An in vitro study revealed that targeting NPC1L1 with ezetimibe blocks the AKT/mTOR signaling pathway in HCC and suppresses cancer cell activity (88).

Collectively, these findings underscore that cholesterol metabolism is notably reprogrammed in cancer cells, with multiple enzymes in the biosynthetic pathway actively contributing to tumor development and progression.

Cholesterol and molecular regulation

Numerous studies have demonstrated that aberrantly activated genes or signaling pathways can promote cancer cell proliferation and suppress apoptosis through the regulation of cholesterol metabolism-related genes (89-93). For instance, the tumor suppressor p53 can inhibit the MVA pathway, and its deficiency or mutation releases multiple constraints on tumor growth (94). The p53 protein directly suppresses the transcription of SQLE and SREBP genes by binding to their promoters, thereby downregulating the MVA pathway in CRC, HCC, OV and BC (95). By contrast, upregulated or unmutated p53 transcriptionally induces ABCA1 gene expression to block SREBP2 activation and inhibit the MVA pathway (96,97). In recent years, the function of the tumor suppressor phosphatase and tensin homolog protein (PTEN) in metabolic regulation has attracted significant attention (98). As a phosphatase, PTEN dephosphorylates phosphatidylinositol-3,4,5-trisphosphate, thereby directly inhibiting the oncogenic PI3K/AKT/mTOR signaling pathway (99). PTEN suppression in endocrine therapy-resistant BC cells leads to increased SQLE expression and a corresponding sensitization to the inhibition of cholesterol synthesis (100). In pancreatic ductal adenocarcinoma (PDAC), PTEN deficiency enhances the protein stability of SQLE by activating the PI3K/AKT/GSK3β-mediated proteasome pathway (101). Activated AKT phosphorylates the cytoplasmic phosphorylating rate-limiting enzyme phosphoenolpyruvate carboxy kinase 1, which is translocated to the ER and acts as a protein kinase that phosphorylates the INSIG protein and disrupts the interaction between the INSIG protein and the SREBP shear-activating proteins, which in turn activates the transcription of SREBPs as well as downstream genes related to lipid synthesis and uptake and promotes the progression of HCC (102). AKT regulates SREBP activation by activating mTOR1; it promotes the protein expression of nuclear SREBP2 through a dual mechanism: First, by phosphorylating and inhibiting its nuclear translocation inhibitor, lipin1; second, by regulating cholesterol transport from lysosomes to the ER (103). Furthermore, suppression of ciliary function activates the Wnt/β-catenin pathway, which synergistically promotes transcription of MVA pathway genes by directly interacting with SREBP2 (104).

Liver X receptors (LXRs) are nuclear receptors and members of the nuclear receptor family that regulate intracellular lipid homeostasis. Activated by high intracellular cholesterol, LXRs function as cholesterol sensors (105). c-Fos promotes alterations in cholesterol metabolism by suppressing the transcriptional activity of LXRα, leading to the accumulation of toxic sterols and bile acids, thereby promoting hepatocellular carcinogenesis (106). High expression of anoctamin-1 contributes to in vivo metastasis of primary esophageal squamous cell carcinoma by inhibiting LXR signaling, leading to cholesterol accumulation and decreased cholesterol hydroxylation via downregulation of the expression of the cholesterol hydroxylase cytochrome P450 family 27 subfamily A member 1 (107).

LDLR is the key molecule that maintains cholesterol balance in the body, releasing cholesterol into cells for utilization while simultaneously lowering cholesterol levels in the blood (108). LDLR-mediated cholesterol uptake plays a contributory role in cancer (109). A recent study has demonstrated that leukocyte immunoglobulin-like receptor B1 directly binds to the LDLR protein to form a complex, thereby enhancing LDLR-mediated cholesterol uptake and conferring resistance to ferroptosis in multiple myeloma (MM) cells (110).

Apolipoprotein B (APOB) is an essential structural component of VLDL and LDL; its C-terminal region contains an LDLR-binding domain that specifically recognizes LDLRs on hepatocyte surfaces, mediating LDL uptake and clearance to maintain plasma cholesterol homeostasis (111). APOB is classified into two primary subtypes: i) ApoB-100, which is synthesized in the liver and serves as the major structural protein of VLDL, intermediate-density lipoprotein (IDL), and LDL; and ii) ApoB-48, which is synthesized in the small intestine, constitutes the core component of chylomicrons and is primarily responsible for transporting dietary lipids (112). APOB is closely associated with metastasis in CRC and liver cancer. Compared with the control group, silencing of APOB in HCC cells has been shown to increase the relative rate of cell proliferation (113). A recent nested case-control study revealed that genetically predicted APOB levels are associated with a 31% reduction in HCC risk [95% confidence interval (CI), 19-42%] and each 0.1 g/l increase in circulating APOB levels was linked to an 11% decrease in HCC risk (95% CI, 8-14%) (114). Another prospective cohort study from Sweden also found that elevated circulating levels of APOB increased the risk of developing CRC (115).

PCSK9 is a key regulator of LDLR protein levels, binding to LDLRs on the cell surface and directing them to lysosomes for degradation (116). In the TME, tumor cell-derived PCSK9 disrupts T-cell receptor signaling by binding to LDLR on the membrane of CD8+ T cells and directing LDLR to lysosomal degradation. This impaired CD8+ T cell activation, proliferation and effector function, ultimately leads to lymphoma metastasis in mice (117). In lung adenocarcinoma (LUAD), the long non-coding RNA EMX2OS competitively binds to microRNA-1185-5p to regulate the LDLR, thereby promoting lung cancer cell proliferation and invasion (118). In addition, MEK/ERK signaling increases intracellular cholesterol uptake by upregulating LDLR expression, leading to HCC metastasis (119).

Similarly, dysregulation of cholesterol metabolism can activate gene expression. Poly (ADP-ribose) polymerase 1 (PARP1) is a multifunctional human ADP-ribosyl transferase (120). In addition to maintaining genomic integrity, PARP1 participates in transcriptional regulation (121). A recent study revealed that long-term high cholesterol promotes OV progression by activating focal adhesion kinase (FAK)/collagen type V alpha 1 chain (COL5A1) signaling through the upregulation of PARP1 expression (122). Several cholesterol derivatives and metabolites, including 7-ketocholesterol (123), 15α-hydroxycholesterol (124), 25-hydroxycholesterol (25-HC) (125), 17-β-estradiol (126) and vitamin D (127), have been shown to induce PARP1 expression.

Cholesterol is a key structural component of LRs. In PC cells, elevated serum cholesterol levels can increase cholesterol content within tumor cell liposomes, thereby activating the LR-mediated Src/PI3K/AKT signaling pathway (128,129). Mitochondrial cholesterol also exhibits physiological activity and promotes tumor cell proliferation and metastasis. For instance, a recent study revealed that the interaction between cytotoxin-associated protein A and cytochrome P450scc (CYP11A1) promotes mitochondrial cholesterol accumulation. This accumulated cholesterol then activates autophagy, thereby enhancing GC cell proliferation and suppressing apoptosis (130). The c-Myc proto-oncogene encodes a family of transcription factors and is one of the most commonly activated oncogenes in human tumors (131). Yang et al (132) found that c-Myc protein directly promotes SQLE transcription, thereby increasing cholesterol production and promoting tumor cell growth. Similarly, activated c-Myc protein has also been observed to enhance HMGCR transcriptional expression in T-cell acute lymphoblastic leukemia (133). Notably, upregulated SREBP1 protein directly interacts with SREBP1 binding elements in the c-Myc promoter region in PC to induce c-Myc activation to drive tumor stemness and metastasis (134). These studies illustrate a reciprocal regulatory relationship between cholesterol metabolism and genes, highlighting their critical roles in tumor cell proliferation and invasion (Fig. 2).

Genes and cholesterol metabolism.
Schematic representation of the mechanism by which genes regulate
cholesterol biosynthesis, uptake, efflux and esterification. PTEN,
mutations in the phosphatase and tensin homolog; PI3K,
phosphatidylinositol 3-kinase; SREBP, sterol regulatory binding
protein; mTOR, mechanistic target of rapamycin; MVA, mevalonate;
PCSK9, proprotein convertase subtilisin kexin/type 9; ATF3,
activating transcription factor 3; ERK,
extracellular-signal-regulated kinase; LDLR, low-density
lipoprotein receptor; LXR, liver X receptor; ABCA1, ATP binding
cassette transporter A1; ABCG1, ATP-binding cassette transporter
G1; CE, cholesterol ester; ACAT1, acetyl-CoA acetyltransferase
1.

Figure 2

Genes and cholesterol metabolism. Schematic representation of the mechanism by which genes regulate cholesterol biosynthesis, uptake, efflux and esterification. PTEN, mutations in the phosphatase and tensin homolog; PI3K, phosphatidylinositol 3-kinase; SREBP, sterol regulatory binding protein; mTOR, mechanistic target of rapamycin; MVA, mevalonate; PCSK9, proprotein convertase subtilisin kexin/type 9; ATF3, activating transcription factor 3; ERK, extracellular-signal-regulated kinase; LDLR, low-density lipoprotein receptor; LXR, liver X receptor; ABCA1, ATP binding cassette transporter A1; ABCG1, ATP-binding cassette transporter G1; CE, cholesterol ester; ACAT1, acetyl-CoA acetyltransferase 1.

Targeting cholesterol metabolism in cancer cells

A large body of preclinical evidence has underscored the critical and multifaceted roles of cholesterol metabolism in cancer progression (135-138). Targeting key nodes of this pathway, such as inhibition of the MVA pathway, has emerged as a promising antitumor strategy (1,139). At present, statins, which act as inhibitors of HMGCR, and PCSK9 inhibitors have emerged as the predominant agents targeting cholesterol metabolism, extensively employed in both basic and clinical research (140,141). The anticancer function of statins has attracted significant attention. Existing evidence indicates that statins can promote tumor cell proliferation, differentiation and apoptosis (76,142,143). A clinical study demonstrated that initiating statin therapy within 1 year of diagnosis results in a 58% relative improvement in BC-specific survival and a 30% relative improvement in OS (144). Similarly, simvastatin (SIM) blocks the isoprenylation of Rab5 GTPase and its mediated endosomal maturation in antigen-presenting cells by depleting GGPP through inhibition of the MVA pathway, ultimately enhancing antitumor immunity by boosting antigen presentation and T cell activation (145). Bisphosphonates (BPs) are another widely studied inhibitor of the MVA pathway, which inhibits FDPS activity and prevents the conversion of isopentenyl pyrophosphate (IPP) to FPP (47,146,147). Bisphosphonate therapy has been reported to improve the prognosis of patients with bona fide MM, PC and BC (148). Moreover, BPs can reduce bone and visceral metastases in women with BC (149,150). Previous studies suggest that SQLE upregulation is closely associated with tumor progression. For instance, upregulated SQLE promotes HCC cell invasion by activating ERK (151) or AKT/mTOR signaling (152). However, targeting SQLE with terbinafine can delay tumor progression in endometrial cancer (ECa) (153), PC (154), CRC (155), HCC (156) and BC (157). Ezetimibe is an FDA-approved drug that acts on NPC1L1 to inhibit the progression of CRC (86), BC (158) and PC (159) by inhibiting intestinal cholesterol absorption. Intracellular cholesterol accumulation triggers LXRs and excess cholesterol is excreted via ABCA1 or ABCG1 (160). The LXR agonists RGX-104 (161), LXR 623 (162) and T0901317 (163) increase ABCA1 expression in a variety of tumor cells, decrease intracellular cholesterol levels and effectively inhibit the growth of mouse xenograft tumors. Additionally, T0901317 can reduce myeloid-derived suppressor cell infiltration through the LXR/APOE pathway, thereby lifting immune suppression, enhancing T cell function and ultimately improving overall immune therapy response (164).

Notably, statins also enhance the efficacy of conventional antitumor drugs (165). Retrospective studies have shown that statin therapy prolongs survival in patients with BC (166), HCC (167), CRC (168) and metastatic PC (169) treated with a combination of first-line chemotherapeutic agents. Results from a phase II clinical trial demonstrated that combining SIM with the epidermal growth factor receptor (EGFR) inhibitor, gefitinib, yielded superior antitumor effects compared with gefitinib alone in non-small cell lung cancer (NSCLC).

The NPC1 protein, which regulates endolysosomal cholesterol transport, has recently emerged as a therapeutic target. NPC1 represents a convergence point for cholesterol derived from LDL, HDL and VLDL, allowing simultaneous targeting of multiple cholesterol sources (170). A previous study has shown that targeted inhibition of NPC1 downregulates mTORC1 signaling and reduces autophagy and tumor growth (171). Moreover, lowering NPC1 expression enhances the therapeutic efficacy of cisplatin or trastuzumab in NSCLC (172), GC (173) and PC (174), leading to improved patient prognosis. These findings highlight the clinical potential of combining cholesterol metabolism inhibitors with existing anticancer agents to overcome therapy resistance. Cancer therapies targeting cholesterol metabolism are summarized in Table I.

Table I

Anticancer therapies that target cholesterol metabolism.

Table I

Anticancer therapies that target cholesterol metabolism.

ReagentTargetMechanism Phenotype/effectCancer type
StatinsHMGCRDecreases serum total cholesterol levelDecreases cancer mortality and prolongs survivalBC (276), HNSCC (277), CRC (278) and PC (279)
StatinsNPC1Suppresses NPC1 signalingInhibits tumor growthTNBC (280)
StatinsERαDecreases ERα expression and activity and promotes apoptosisReduces the size and weight of xenograft tumorsACC (281)
TerbinafineSQLEDecreases cholesterol levels and increases chemotherapy sensitivityImproves patient prognosisHCC (156) and CRC (70)
TerbinafineSQLESuppresses AKT/mTOR signalingInhibits tumor growthHCC (151)
RGX-104LXRActivates the LXR/ApoE signaling axis and increases radiosensitivityMyeloid-derived suppressor cell depletion and increased T-cell activationNSCLC (161,282)
LXR623/GW3965LXRActivates ATF4-dependent pro-apoptotic protein Noxa expressionPromotes tumor cell apoptosisCRC (283) and GBM (284)
FatostatinSREBPsInhibits AKT/mTORC1/GPX4 signalingPromotes tumor cell apoptosisGBM (285) and ECa (286)
AvasimibeACAT1Inhibits Wnt/β-catenin signalingBlocks tumor metastasisPC (287)
ItraconazoleNPC1Activates AMPK signalingInhibits tumor angiogenesisCRC (288) and NSCLC (289)

[i] HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase; HNSCC, head and neck cutaneous squamous cell carcinoma; BC, breast cancer; CRC, colorectal cancer; PC, prostate cancer; TNBC, triple negative BC; ACC, adrenocortical carcinoma; ERα, estrogen receptor α; NPC1, Niemann-Pick disease Type C; HCC, hepatocellular carcinoma; NSCLC, non-small cell lung cancer; GBM, glioblastoma; ECa, endometrial cancer; LXR, liver X receptor; ACAT1, acyltransferase/sterol O-acyltransferase 1; SREBPs, sterol regulator element binding proteins.

Cholesterol and ferroptosis

Ferroptosis, a regulated form of cell death driven by iron-dependent lipid peroxidation, represents a promising therapeutic avenue in oncology due to its involvement in tumor progression and treatment resistance (175). Glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 are central to regulating ferroptosis (176). Elevated cholesterol levels in cancer cells have been shown to suppress ferroptosis and impair immune responses (177). Recent studies have shown that 7-dehydrocholesterol (7-DHC) and B-ring sterols act as free-radical-trapping antioxidants to protect mitochondrial membranes from phospholipid autoxidation, thereby resisting ferroptosis (178,179). While most cancer cells succumb under stress, surviving populations often accumulate high cholesterol levels to evade ferroptosis death (84,180,181). Chronic exposure to 27-hydroxycholesterol (27-HC), an abundant metabolite of circulating cholesterol, causes sustained expression of GPX4 and significantly increases the tumorigenic and metastatic capacity of BC cells (13). These findings highlight that aberrant cholesterol metabolism can influence cancer pathogenesis by activating ferroptosis. In cancer cells, increased uptake via the upregulation of lipoprotein receptors and increased synthesis rates through the hyperactive MVA pathway results in cholesterol accumulation, fostering cellular proliferation, tumor metastasis and immune suppression (182-184). Studies have revealed that GPX4 relies on the output of the MVA pathway to hinder lipid peroxidation production and ferroptosis sensitivity in HCC cells (185,186). Exogenous cholesterol supplementation activates the B7H3-mediated AKT/SREBP2 signaling pathway to promote ferroptosis resistance in CRC cells as well as xenograft tumor formation in mice (187).

Elevated cholesterol levels reduce membrane fluidity and promote LR formation, thereby inhibiting the diffusion of lipid peroxidation substrates and suppressing ferroptosis (188-190). A novel nanozyme composed of an iron metal-organic framework (Fe-MOF) and nanoparticles loaded with cholesterol oxidase and PEGylation (CP) for integrated ferroptosis and immunotherapy, depletes cholesterol and disrupts LR integrity, downregulates GPX4 and ferroptosis suppressor protein 1 (FSP1) and further promotes ferroptosis. Concurrently, Fe-MOF/CP augments immunogenic cell death, reduces programmed death-ligand 1 expression and revitalizes exhausted CD8+ T cells (191). Similarly, in cholesterol-addicted lymphoma cells, targeting SCARB1 via HDL nanoparticles to reduce cholesterol uptake not only eliminates GPX4 expression but also triggers a compensatory response that enhances cholesterol biosynthesis, ultimately leading to ferroptosis (189). GPX4, a core regulator of ferroptosis, is modulated by IPP, a product of the MVA pathway and an intermediate in cholesterol synthesis. Thus, regulating the upstream synthesis pathways of IPP using inhibitors (such as FIN56, RSL3, statins, ML162 and ML210) induces ferroptosis by suppressing GPX4 (a protein responsible for preventing lipid peroxide formation); whereas FIN56 and withaferin can trigger GPX4 degradation (192). Squalene and HMGCR are thought to exert anti-ferroptosis effects on cancer cells (180,193). For example, SIM can inhibit the expression of HMGCR to downregulate the MVA pathway, GPX4 and FSP1, thereby inducing triple-negative BC cell ferroptosis (194,195). Conversely, SQLE exerts anti-ferroptosis effects, whereas exogenous cholesterol hydroperoxide induces dose-dependent cell death (196,197). These findings highlight the therapeutic potential of targeting cholesterol biosynthesis to sensitize cancer cells to ferroptosis (Fig. 3).

Regulation of ferroptosis by the
cholesterol biosynthesis pathway. 7-DHC, 27-HC and B-ring sterols
activate the ferroptosis process by upregulating GPX4, FSP1, PTGS2,
FT1 and FTH expression. Inhibition of key enzymes of cholesterol
metabolism such as HMGCR and IPP are expected as promising
approaches to induce ferroptosis in cancer cells. HMGCR,
3-hydroxy-3-methylglutaryl-CoA reductase; HDL, high-density
lipoprotein; 27-HC, 27-hydroxycholesterol; GPX4, glutathione
peroxidase 4; SIM, simvastatin; RSL3, ras-selective lethal 3; IPP,
isoprene unit isopentenyl diphosphate; Fe-MOF/CP, iron
metal-organic framework/cholesterol oxidase and PEGylation; FSP1,
Ferroptosis suppressor protein 1; PD-L1, programmed death ligand 1;
SREBP2, sterol regulatory binding protein 2; 7-DHC,
7-dehydrocholesterol.

Figure 3

Regulation of ferroptosis by the cholesterol biosynthesis pathway. 7-DHC, 27-HC and B-ring sterols activate the ferroptosis process by upregulating GPX4, FSP1, PTGS2, FT1 and FTH expression. Inhibition of key enzymes of cholesterol metabolism such as HMGCR and IPP are expected as promising approaches to induce ferroptosis in cancer cells. HMGCR, 3-hydroxy-3-methylglutaryl-CoA reductase; HDL, high-density lipoprotein; 27-HC, 27-hydroxycholesterol; GPX4, glutathione peroxidase 4; SIM, simvastatin; RSL3, ras-selective lethal 3; IPP, isoprene unit isopentenyl diphosphate; Fe-MOF/CP, iron metal-organic framework/cholesterol oxidase and PEGylation; FSP1, Ferroptosis suppressor protein 1; PD-L1, programmed death ligand 1; SREBP2, sterol regulatory binding protein 2; 7-DHC, 7-dehydrocholesterol.

Cholesterol and autophagy

In cancer, autophagy exhibits a context-dependent dual role, capable of both suppressing and promoting tumorigenesis (198). During the early stages, autophagy prevents tumor initiation by eliminating oncogenic proteins, toxic misfolded aggregates and damaged organelles (199). However, in the later stages of tumorigenesis, autophagy functions as a dynamic degradation and recycling system that can enhance cancer invasiveness by promoting metastasis, thereby sustaining tumor survival and growth (200). Autophagy involves a variety of signaling pathways, such as the mitogen-activated protein kinase (MAPK), mTOR, AKT, HIF-1α and p53 pathways (201). Lysosomal cholesterol activates mTORC1 through the SLC38A9/NPC1 signaling axis, inducing autophagy that promotes tumor metastasis and invasion (202). Similarly, another study revealed that cholesterol also induces autophagy in HCC cells by activating mTORC1 via the SNHG6/FAF2/mTOR axis, increasing the incidence of HCC driven by a high cholesterol diet (203). In MM, researchers have reported that cholesterol accumulation mediates autophagy activation via the PI3K/AKT pathway (204). Conversely, phosphorylated AKT levels as well as rapamycin signaling are markedly suppressed after the use of statins by lowering intracellular cholesterol levels (205). The mTOR, AKT and p53 pathways also modulate SREBP activity, which in turn regulates cholesterol synthesis (96,206). Among these, the AKT and mTOR pathways are the most frequently studied de novo cholesterol synthesis pathways in cancer cells (207). mTOR serves as the core molecule mediating the regulatory function of AKT over SREBPs. mTOR operates through a dual mechanism: on the one hand, it is activated by AKT (206); on the other, it drives cholesterol biosynthesis by enhancing SREBP2 activity and inhibiting its degradation (208). Whether cholesterol-induced autophagy initiates positive feedback regulation of these pathways warrants further investigation.

Emerging evidence indicates that cholesterol enrichment within organelles facilitates the recruitment of autophagy-initiation proteins and enhances autophagosome formation, contributing to chemoresistance (209,210). The cholesterol transporter protein GRAM domain containing 1B can coordinate cellular processes by mediating cholesterol distribution between organelles, thereby inhibiting autophagosome formation and reducing mitochondrial bioenergetic metabolism (211). Furthermore, cholesterol-rich membrane microstructure domain (CEMM)-mediated sequestration of the vesicle-associated membrane protein 3/syntaxin-6 complex inhibits autophagosome fusion. Conversely, CEMM deficiency promotes autophagosome formation and confers doxorubicin resistance in BC (212). Similarly, cholesterol inhibits the autophagic degradation of receptor tyrosine kinase in a Golgi membrane protein 1-dependent manner to promote HCC metastasis (213). Cholesterol accumulation in lysosomes induces autophagy initiation and enhances carboplatin resistance in OV cells by activating the NOTCH/DNA-binding protein recombination signal binding protein-Jκ signaling pathway (214). These results emphasize that cholesterol is an important regulator of signaling pathways in cancer.

Cholesterol metabolites also play a key role in activating autophagy. High 25-HC levels promote Kras-driven PDAC progression. Mechanistically, 25-HC promotes autophagy, leading to the downregulation of MHC-I and a reduction in CD8+ T-cell infiltration into tumors (215). BC is one of the most common female cancer types in the world, with estrogen receptor-positive BC being the most common subtype (216). The accumulation of cholesterol-5,6-epoxide (5,6-EC) metabolites enhances resistance to tamoxifen through the activation of autophagy to potentiate antiestrogen binding site activity (217,218). Unraveling the molecular interactions within the regulatory networks of cholesterol metabolism and autophagy provides core guidance for identifying novel anti-cancer targets and formulating targeted strategies (Fig. 4).

Cholesterol-induced activation of
autophagy. Exogenous cholesterol supplementation and elevated
endogenous cholesterol activate mTORC1-dependent autophagy through
multiple pathways, including those involving p53, AKT and NPC1.
Additionally, cholesterol derivatives such as 25-HC and 5,6-EC also
stimulate autophagy in tumor cells. CEMM, cholesterol-enriched
membrane micro-domains; 25-HC, 25-hydroxycholesterol; RBP-JK,
DNA-binding protein recombination signal binding protein-Jκ; VAMP3,
vesicle-associated membrane protein; GOLM1, Golgi membrane protein
1; RTK, receptor tyrosine kinase; PI3K, phosphatidylinositol
3-kinase; SREBP, sterol regulatory binding protein; mTOR,
mechanistic target of rapamycin; SNHG6, small nucleolar RNA host
gene 6; FAF2, Fas-associated factor family member 2; SLC38A9,
solute carrier family 38 member 9; NPC1, Niemann-Pick C1.

Figure 4

Cholesterol-induced activation of autophagy. Exogenous cholesterol supplementation and elevated endogenous cholesterol activate mTORC1-dependent autophagy through multiple pathways, including those involving p53, AKT and NPC1. Additionally, cholesterol derivatives such as 25-HC and 5,6-EC also stimulate autophagy in tumor cells. CEMM, cholesterol-enriched membrane micro-domains; 25-HC, 25-hydroxycholesterol; RBP-JK, DNA-binding protein recombination signal binding protein-Jκ; VAMP3, vesicle-associated membrane protein; GOLM1, Golgi membrane protein 1; RTK, receptor tyrosine kinase; PI3K, phosphatidylinositol 3-kinase; SREBP, sterol regulatory binding protein; mTOR, mechanistic target of rapamycin; SNHG6, small nucleolar RNA host gene 6; FAF2, Fas-associated factor family member 2; SLC38A9, solute carrier family 38 member 9; NPC1, Niemann-Pick C1.

Cholesterol and EMT

EMT is a key driver of tumor growth and metastasis, promoting a malignant phenotype by conferring enhanced metastatic capacity and resistance to therapeutic interventions. Tumor EMT states have also been found to exhibit high plasticity during transitions between epithelial and mesenchymal phenotypes (219). The progression of EMT is governed by the precise coordination of a core set of transcription factors, such as Snail1/2, zinc finger enhancer-binding protein 1 and Twist1, whose expression levels directly determine the degree of EMT activation and the transformation of cellular phenotypes (220). Cholesterol increases the risk of EMT in cancer. A high-cholesterol diet promotes lung metastasis of HCC in mice by activating sterol O-acyltransferase 1 (SOAT1)-mediated EMT. Mechanistically, SOAT1 increases cholesterol accumulation and esterification, thereby driving EMT and HCC progression (93). Treatment with 20 μmol/l cholesterol was found to promote in vivo graft tumor growth and distal metastasis in CRC by inducing EMT (19). Cholesterol has been demonstrated to regulate several classical signaling pathways that activate EMT. Cholesterol (10 μmol/l) induces adipocyte membrane-associated protein binding to EGFR substrate 15-associated protein, activates ERK1/2 and facilitates the EMT process in PC cells (221). The integrity of LRs is essential for the formation of the TGF-β receptor (TβR) I/TβRII/TGF-β1 complex, and LR disruption impedes canonical TGF-β signaling. Cholesterol within LRs promotes EMT, migration and invasion in BC cells via TGF-β-mediated MAPK activation (222). High cholesterol also promotes proliferation and migration in BC and PDAC cells through activation of Wnt/β-catenin signaling (223,224), and similarly induces EMT in melanoma via the MAPK pathway (225). Notably, our latest study revealed that long-term treatment of OV cells with high cholesterol concentrations (>20 μmol/l) significantly induces the expression of cellular EMT transcription factors and mesenchymal cell marker proteins. Mechanistically, cholesterol promotes invasion of orthotopic ovarian tumors into the liver and kidney in mice through activation of the PARP1/COL5A1/FAK/EMT axis (122). These studies collectively indicated that cholesterol plays a key role in promoting EMT.

Cholesterol-related genes also have key roles in EMT-induced metastasis and drug resistance. Upregulation of ABCA1 in CRC facilitates EMT induction and enhances the migratory and invasive abilities by modulating caveolin-1 stability (226). High ABCG1 expression enhances cisplatin resistance in LUAD by activating Slug-mediated EMT (227). Dysregulation of cholesterol metabolism not only promotes the development of HCC but also drives tumors to exhibit highly invasive characteristics, including intrahepatic spread and early extrahepatic metastasis (76,228,229). A recent study has shown that high expression of 7-DHC reductase is correlated with poor prognosis in patients with BLCA and promotes metastasis in mice via PI3K/AKT/mTOR-mediated EMT activation (230). 27-HC, the most abundant oxysterol, increases the risk of BC progression by promoting EMT and oncogenic transformation of BC stem cells through LXR activation (15). Accumulation of apolipoprotein E leads to cholesterol enrichment in HCC cells, stimulating PI3K/AKT signaling, inducing EMT and increasing sorafenib resistance and invasiveness (231). Aberrant HMGCR expression enhances statin resistance in therapy-resistant PC cells and augments TGF-β-induced EMT in lung and ovarian epithelial carcinomas (232-234). Additionally, HMGCR augments TGF/β-induced EMT progression in epithelial cell carcinomas (lung and ovarian) (235). Collectively, this scientific evidence highlights the important role played by cholesterol in promoting EMT, providing a new means of targeting cholesterol metabolism to combat tumor progression (Fig. 5). Cholesterol-activated EMT markers and transcription factors in different cancer types are summarized in Table II.

Cholesterol promotes EMT progression.
High cholesterol levels facilitate EMT by activating signaling
pathways including TGF-β/MAPK, Wnt/β-catenin, PI3K/AKT/mTOR,
PARP1/COL5A1/FAK and APMAP/EPS15R/EGFR. Conversely, LXR-induced
upregulation of ABCA1 and ABCG1 suppresses EMT in tumor cells. EMT,
epithelial-mesenchymal transition; TGF-β, transforming growth
factor β; MAPK, mitogen-activated protein kinase; PI3K,
phosphatidylinositol 3-kinase; mTOR, mechanistic target of
rapamycin; PARP1, Poly (ADP-ribose) polymerase 1; COL5A1, collagen
type V α1 chain; FAK, focal adhesion kinase; APMAP, adipocyte
plasma membrane associated protein; EGFR, epidermal growth factor
receptor; EPS15R, EGFR-substrate 15-related protein; LXR, liver X
receptor; ABCA1, ATP binding cassette transporter A1; ABCG1,
ATP-binding cassette transporter G1; HCD, high cholesterol diet;
SOAT1, sterol O-acyltransferases 1; ERK1/2, extracellular
signal-related kinases 1 and 2.

Figure 5

Cholesterol promotes EMT progression. High cholesterol levels facilitate EMT by activating signaling pathways including TGF-β/MAPK, Wnt/β-catenin, PI3K/AKT/mTOR, PARP1/COL5A1/FAK and APMAP/EPS15R/EGFR. Conversely, LXR-induced upregulation of ABCA1 and ABCG1 suppresses EMT in tumor cells. EMT, epithelial-mesenchymal transition; TGF-β, transforming growth factor β; MAPK, mitogen-activated protein kinase; PI3K, phosphatidylinositol 3-kinase; mTOR, mechanistic target of rapamycin; PARP1, Poly (ADP-ribose) polymerase 1; COL5A1, collagen type V α1 chain; FAK, focal adhesion kinase; APMAP, adipocyte plasma membrane associated protein; EGFR, epidermal growth factor receptor; EPS15R, EGFR-substrate 15-related protein; LXR, liver X receptor; ABCA1, ATP binding cassette transporter A1; ABCG1, ATP-binding cassette transporter G1; HCD, high cholesterol diet; SOAT1, sterol O-acyltransferases 1; ERK1/2, extracellular signal-related kinases 1 and 2.

Table II

EMT markers or TFs activated by cholesterol in different cancer types.

Table II

EMT markers or TFs activated by cholesterol in different cancer types.

Cancer typesEMT markers or TFs Phenotype/effectLimitations of the mechanism validation
HCC (93,231,290)E-cadherin, Vimentin, Twist1, N-cadherin, Snail1 and SlugProliferation and invasionAbsence of transcriptional regulation validation
GBM (285)N-cadherin, Snail1 and VimentinMigration and invasionAbsence of gene interference validation
TNBC (291,292)E-cadherin, N-cadherin Vimentin, ZEB1 and Snail2Migration and invasionAbsence of in vivo experiments
PC (221)N-cadherin and VimentinLiver metastasisUnvalidated in multiple tumor cell lines
OV (122)E-cadherin, N-cadherin, Snail, Twist1 and VimentinTumor invasionAbsence of transcriptional regulation validation
BLCA (230)E-cadherin, N-cadherin, MMP-9 and VimentinMigration and invasionAbsence of gene interference validation
LUAD (293)E-cadherin, N-cadherin and VimentinTumor growthAbsence of transcriptional regulation validation
CCA (294)N-cadherin, Vimentin, ZEB1 and SnailTumor growthUnvalidated in multiple tumor cell lines
ECa (295)N-cadherin, Vimentin and Snail2Migration and invasionAbsence of in vivo experiments
EC (296)E-cadherin, Vimentin, Slug and Snail2Lung metastasisAbsence of transcriptional regulation validation
CRC (297)N-cadherin, Vimentin and SnailLymph node and liver metastasesAbsence of gene interference validation

[i] HCC, hepatocellular carcinoma; GBM, glioblastoma multiforme; TNBC, triple negative breast cancer; PC, prostatic cancer; OV, ovarian cancer; BLCA, bladder cancer; LUAD, lung adenocarcinoma; CCA, cholangiocarcinoma; ECa, endometrial cancer; EC, esophageal cancer; CRC, colorectal cancer; EMT, epithelial-mesenchymal transition; TFs, transcription factors.

Cholesterol and immunity

Recent studies have demonstrated that modulation of systemic cholesterol metabolism could improve responses to immunotherapy (16,136,236). Cholesterol depletion is a key metabolic basis for tumor-associated macrophages (TAMs) acquiring an immunosuppressive phenotype (6). In patients with CRC, cholesterol metabolism promotes macrophage polarization towards a pro-tumor phenotype and is associated with poorer prognosis (237). Notably, replenishing cholesterol or adding 7-DHC to mouse macrophages was shown to inhibit interferon-β production (238). In a murine melanoma model, cholesterol accumulation in the TME induced functional exhaustion of CD8+ T cells (239). Cholesterol metabolism exhibits different effects on different T cell subsets. Cholesterol enhances receptor signaling and immune synapse formation in CD8+ T cells by binding to T cell receptor β (240). Another in vivo study demonstrated that inhibition of ACAT1 by knockdown or pharmacological inhibition can inhibit intracellular cholesterol esterification in T cells and enhance the proliferative and effector capacity of CD8+ T cells, whereas CD4+ T cells are unaffected (241). In C57BL/6J mice, genes involved in cholesterol esterification (ACAT1/2, lecithin cholesterol acyltransferase and cholesterol ester transfer protein) and utilization (CYP11A1, CYP17A1 and CYP7A1) are upregulated in γδ T cells compared with αβ T cells (242). Moreover, cholesterol depletion using methyl-β-cyclodextrin reduces the activation phenotype of γδ T cells (243). However, the impact of hypercholesterolemia or lipid-lowering treatments on T cell signaling in human patients remains unclear.

Cholesterol has also been identified as a key immunomodulatory factor, whose accumulation promotes tumor immune escape. In the TME, cholesterol attenuates the efficacy of PD-L1 blockade by activating the signal transducer and activator of transcription 3/nuclear factor κB pathway, driving macrophages toward an M2-like phenotype that accelerates CRC progression (244). Additionally, cholesterol upregulates PD-L1 expression on cancer cells, facilitating immune evasion (245,246). Hyaluronic acid oligomers secreted by tumor cells increase cholesterol efflux from TAMs, promoting M2 polarization and tumor progression (6). Although IL-9-secreting CD8+ T cells (Tc9 cells) exhibit potent antitumor activity, cholesterol suppresses IL-9 expression and impairs Tc9 cell function through LXR activation (247). In microsatellite-stable CRC, tumor cells secrete distal cholesterol precursors that polarize T cells into Th17 cells, promoting tumor growth (248).

Cholesterol derivatives similarly modulate immune responses. 27-HC promotes the development of the BC pre-metastatic microenvironment by attracting polymorphonuclear neutrophils and γδ-T cells at metastatic sites and depleting CD8+ T cells (249). Furthermore, 27-HC induces T cell cholesterol deficiency by inhibiting SREBP2 and activating LXR, leading to autophagy-mediated T cell apoptosis (14). In HCC, oxidized sterols activate LXRα in dendritic cells (DCs), downregulating CC chemokine receptor 7 expression and impairing DC migration to lymphoid organs, thereby suppressing antitumor immunity (250).

In summary, the conflicting results of cholesterol in modulating immunotherapy suggest that a deeper understanding of the regulatory mechanisms of cholesterol metabolism in the interactions between tumor cells and immune cells in the TME will contribute to the development of new cancer immunotherapy strategies (Fig. 6). Targeting cholesterol metabolism in the tumor immune microenvironment represents a promising approach to enhance antitumor immunity.

Impact of cholesterol and derivatives
on the adaptive immune system. Cholesterol regulates T cell
function through direct binding to the TCR or through indirect
mechanisms, influencing immune synapse formation. Excessive
cholesterol impairs T cell activity; it also promotes M2 macrophage
polarization via the STAT3/NF-κB/PD-L1 axis. The oxysterol 27-HC
induces T cell apoptosis through SREBP inhibition and LXR
activation. Genes involved in cholesterol transport and metabolism
(such as ACAT1/2, LCAT, CETP, CYP11A1, CYP17A1 and CYP7A1) modulate
cholesterol levels in γδ T cells. Cholesterol enhances IL-9
secretion in macrophages but inhibits CCR7 in DCs; it also promotes
Th17 cell differentiation and suppresses IFN-γ production in
macrophages. TCR, T-cell receptor; STAT3, signal transducer and
activator of transcription 3; NF-κB, nuclear factor κB; PD-L1,
programmed death-1; 27-HC, 27-hydroxycholesterol; SREBP, sterol
regulatory binding protein; LXR, liver X receptor; ACAT1/2,
acetyl-CoA acetyltransferase 1/2; LCAT, lecithin cholesterol
acyltransferase; CETP, cholesteryl ester transfer protein; CYP11A1,
enzyme cytochrome P450 family 11 subfamily A member 1; CYP17A1,
enzyme cytochrome P450 family 17 subfamily A member 1; CYP7A1,
cholesterol 7α-hydroxylase; IL-9, interleukin-9; CCR7, chemokine
receptor 7; DCs, dendritic cells; IFN-γ, interferon-γ; Th17,
intestinal IL-17-producing T helper; 7-DHC,
7-dehydrocholesterol.

Figure 6

Impact of cholesterol and derivatives on the adaptive immune system. Cholesterol regulates T cell function through direct binding to the TCR or through indirect mechanisms, influencing immune synapse formation. Excessive cholesterol impairs T cell activity; it also promotes M2 macrophage polarization via the STAT3/NF-κB/PD-L1 axis. The oxysterol 27-HC induces T cell apoptosis through SREBP inhibition and LXR activation. Genes involved in cholesterol transport and metabolism (such as ACAT1/2, LCAT, CETP, CYP11A1, CYP17A1 and CYP7A1) modulate cholesterol levels in γδ T cells. Cholesterol enhances IL-9 secretion in macrophages but inhibits CCR7 in DCs; it also promotes Th17 cell differentiation and suppresses IFN-γ production in macrophages. TCR, T-cell receptor; STAT3, signal transducer and activator of transcription 3; NF-κB, nuclear factor κB; PD-L1, programmed death-1; 27-HC, 27-hydroxycholesterol; SREBP, sterol regulatory binding protein; LXR, liver X receptor; ACAT1/2, acetyl-CoA acetyltransferase 1/2; LCAT, lecithin cholesterol acyltransferase; CETP, cholesteryl ester transfer protein; CYP11A1, enzyme cytochrome P450 family 11 subfamily A member 1; CYP17A1, enzyme cytochrome P450 family 17 subfamily A member 1; CYP7A1, cholesterol 7α-hydroxylase; IL-9, interleukin-9; CCR7, chemokine receptor 7; DCs, dendritic cells; IFN-γ, interferon-γ; Th17, intestinal IL-17-producing T helper; 7-DHC, 7-dehydrocholesterol.

Epidemiologic relationship between cholesterol and tumors

Although extensive basic and preclinical studies have demonstrated the involvement of cholesterol in cancer development and the potential of targeting its metabolism, the relationship between serum cholesterol levels and tumor risk remains controversial, with epidemiological studies reporting conflicting findings (3). Several large-scale studies have indicated a positive association between elevated serum cholesterol and cancer risk. For example, two prospective cohort studies provided evidence that higher cholesterol levels were positively associated with the risk of high-grade PC (251,252). Another study revealed an association between the use of statins to lower serum cholesterol levels and the risk of developing or dying from 13 tumor types, including HCC, MM, BC, ECa and CRC (253). A 10 mg/dl increase in cholesterol was correlated with a 9% increase in PC recurrence (128). In OV, elevated preoperative LDL-C was significantly associated with a poorer OS, whereas high HDL-C was correlated with improved progression-free survival (254). Patients with advanced-stage OV are typically accompanied by the presence of malignant ascites (MAs) and high cholesterol levels in these MAs (255). Moreover, high cholesterol levels promote drug resistance in OV cells by increasing the expression of multidrug resistance protein 1 (256). Dietary cholesterol and tumor risk also deserve attention. According to population-based cohort studies, tumorigenesis is closely related to dietary structure (257), and changes in dietary structure effectively prevent tumorigenesis and progression (258,259). Increased dietary cholesterol intake is positively associated with the risk of BC (260). Dietary cholesterol is a risk factor for the development of ECa, with a 35% increased risk of tumor development for every 150 mg/kcal of cholesterol consumed (261). Esophageal cancer (EC) ranks as the ninth most common cancer worldwide, with genetic factors, dietary factors and environmental risk factors potentially influencing its progression (262). A meta-analysis that investigated the association between high-cholesterol diets and EC revealed that dietary cholesterol intake may increase the risk of EC (summarized odds ratio, 1.424; 95% CI, 1.191-1.704) (263). Pancreatic cancer (PCa) risk is associated with elevated serum cholesterol levels, which are partially influenced by diet (264). A study involving 258 patients with PCa and 551 controls demonstrated that a plant-based cholesterol-lowering diet is associated with a reduced risk of PCa (265). In addition, higher cumulative dietary cholesterol intake is positively associated with the risk of GC (266), HCC (267) and OV (267). The incidence of OV is significantly lower by 40% in individuals consuming a low-cholesterol diet than those on a conventional diet (268). However, studies on dietary cholesterol and lung cancer risk have consistently shown no correlation (115,269,270).

Conversely, other epidemiological studies reported no clear association or even an inverse relationship between cholesterol and cancer. For instance, a large prospective cohort study from South Korea found that the association between serum TC levels and cancer was dependent on the cancer type (251). Further comparison across cancer types revealed that elevated serum TC levels showed a significant positive association with the risk of PC and CRC in men, as well as BC in women. Conversely, there was a significant negative association with the overall risk of HCC and GC in the general population. Moreover, a population-based cohort study has suggested that serum TC levels may be negatively associated with the risk of OV and that circulating lipid levels are not strongly correlated with the risk of OV (271). These studies showed that the positive correlation and negative correlation coexisted, indicating that the association was cancer type specific. Similarly, our latest study showed that higher daily dietary cholesterol intake was significantly positively associated with the risk of OV, whereas abnormal lipid levels were not associated with the risk of OV (33).

These conflicting epidemiological results suggest that the role of cholesterol in cancer development remains uncertain, warranting further investigation into its underlying mechanisms. Key issues include the reliability of dietary study data, the limitations of animal models in simulating human disease states and the potentially opposing effects of serum cholesterol across different tumor stages. The current epidemiological evidence regarding cholesterol and cancer risk is indeed contradictory, stemming from multiple confounding factors and methodological challenges. The primary concern is reverse causality: Actively growing tumors consume large amounts of cholesterol to build membrane structures and synthesize signaling molecules, potentially leading to reduced serum cholesterol levels. Thus, observed low cholesterol levels may result from undetected tumors rather than cause them. This is particularly notable in studies of malignancies such as liver and stomach cancer, potentially completely reversing the apparent 'protective association' (272,273). Cancer type specificity further complicates this relationship as metabolic dependencies vary significantly across tumor types: Hormone-sensitive cancers such as PC and OV may rely more heavily on cholesterol as a steroid hormone precursor, showing positive correlations; whereas other tumor types may exhibit no clear association or even negative correlations (274,275). Methodological limitations also warrant attention: Dietary cholesterol studies rely on self-reporting, introducing recall bias; single serum cholesterol measurements may fail to reflect long-term exposure levels; and statin use confounds the association between serum cholesterol and cancer risk. Most critically, correlation does not imply causation. Observational studies reveal statistical associations but cannot establish biological mechanisms. These conflicting findings suggest systemic cholesterol levels may be unreliable cancer risk indicators, while targeted interventions affecting intracellular cholesterol metabolism may hold greater therapeutic promise than systemic cholesterol reduction. Recently, we suggested that low levels of cholesterol promote tumor progression, that high cholesterol may inhibit tumor cell growth and that chronically high levels of cholesterol ultimately screen for more proliferative and invasive tumor cells (122). Evidence from these studies suggests that the dysregulation of cholesterol homeostasis is an important factor in cancer development, but its true link to tumor progression remains unclear. Studies are needed to link population epidemiological data, molecular mechanisms and clinical data more broadly to more effectively unravel the processes by which cholesterol affects cancer. The associations of serum cholesterol and dietary cholesterol with the risk of cancer development are summarized in Table III.

Table III

Epidemiological evidence of cholesterol-cancer associations.

Table III

Epidemiological evidence of cholesterol-cancer associations.

HCD or serum TC level Phenotype/effectRelationshipCancer types
HCDLung metastasisPositiveHCC (298)
HCDCancer riskPositiveECa (299)
HCDAssociated with a longer overall survivalPositiveBC (300,301)
HCDCancer riskPositiveGC (302)
HCDCancer riskUnrelatedOV (267,303)
HCDCancer riskPositivePCa (265)
HCDCancer riskUnrelatedLC (269)
HCDCancer riskPositiveEC (263)
High serum TCAssociated with lymph node metastasisPositivePNENs (304)
Low serum TCAssociated with more advanced tumor gradingPositiveRCC (305)
High serum TCCancer riskUnrelatedMM (306)
High serum TCAssociated with the diagnosis of high-grade PCPositivePC (26)
High serum TCAssociated with CRC incidence ratePositiveCRC (307,308)
High serum TCAssociated with longer overall survivalPositiveNSCLC (309)
High serum TCCancer riskUnrelatedBC (310)
High serum TCAssociated with longer overall survivalPositiveCCA (311)
High serum TC5-year survival rate reducedNegativeHGG (312)
High serum TCCancer riskNegativeGC (31)
High serum TCCancer recurrencePositiveNB (313)
High serum TCCancer riskPositivePCOS (314)
High serum TCRate of prevalencePositiveOSCC (315)
High serum TCTotal mortality rateNegativeOS (316)
High serum TCCancer riskUnrelatedOV (33)

[i] HCC, hepatocellular carcinoma; PNENs, pancreatic neuroendocrine neoplasms; RCC, renal cell carcinoma; MM, multiple myeloma; PC, prostate cancer; CRC, colorectal cancer; CCA, cholangiocarcinoma; HGG, high-grade glioma; PCa, pancreatic cancer; GC, gastric cancer; NB, neuroblastoma; PCOS, polycystic ovary syndrome; OSCC, oral squamous cell carcinoma; OS, osteosarcoma; OV, ovarian cancer; EC, esophageal cancer; TC, total cholesterol; HCD, high cholesterol diet; LC, lung cancer; ECa, endometrial cancer; BC, breast cancer; NSCLC, non-small cell lung cancer.

Conclusions and perspectives

The present review describes the role of cholesterol in cancer progression from a multidimensional perspective. First, how cholesterol activates oncogenes and oncogenic signals was discussed. Then, the molecular mechanisms regulated by cholesterol in cancer were summarized. Finally, the ongoing controversies regarding cholesterol levels and cancer risk within epidemiological studies were addressed. Although substantial evidence supports a definitive role for cholesterol metabolism in tumorigenesis and progression, several critical questions remain unresolved. For example, whether the specific roles of cholesterol metabolites or derivatives are consistent, what the causal relationship is between abnormal cholesterol metabolism and tumorigenesis and how cholesterol metabolism influences other metabolic processes such as glucose metabolism. Notably, studies on the heterogeneity of cholesterol metabolism (such single-cell metabolomics) and comparative analyses across cancer types (such as BC vs. liver cancer) are fewer and deserve focused attention. This complexity suggests that not only do we need to modulate from a single cholesterol metabolic pathway but also interfere with the activation of oncogenes or oncogenic signals and that a combination of different drugs or inhibitors may be a more effective antitumor regimen.

In conclusion, dysregulation of cholesterol metabolism is increasingly recognized as a critical facilitator of cancer development. While population-based epidemiological findings have been inconsistent, both basic and clinical studies indicate that targeting cholesterol metabolism can significantly inhibit tumor progression. These insights provide a robust foundation for the development of cholesterol-focused therapeutics and offer promising new strategies for cancer prevention and treatment.

Availability of data and materials

Not applicable.

Authors' contributions

ZH was responsible for conceptualization, writing the original draft and visualization. LZ and SG helped to revise the manuscript and collect literature. GX and XY were responsible for supervision, writing, reviewing and editing. Data authentication is not applicable. All authors read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Abbreviations:

TME

tumor microenvironment

EMT

epithelial-to-mesenchymal transition

HMGCR

HMG-CoA reductase

SREBPs

sterol regulatory element binding proteins

SQLE

squalene epoxidase

ACAT1

acetyl-CoA acetyltransferase 1

ABCA1

ATP-binding cassette transporter A1

LDLR

low-density lipoprotein receptor

TC

total cholesterol

VLDL

very low-density lipoprotein

HDL-C

high-density lipoprotein cholesterol

MVA

mevalonate

IPP

isopentenyl pyrophosphate

FDPS

farnesyl diphosphate synthase

GGPP

geranylgeranyl pyrophosphate

FPP

farnesyl pyrophosphate

25-HC

25-hydroxycholesterol

LR

lipid raft

GPX4

glutathione peroxidase 4

PD-L1

programmed death-ligand 1

FSP1

ferroptosis suppressor protein 1

HCD

high cholesterol diet

Acknowledgements

Not applicable.

Funding

This review was funded by the Research Fund of Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital (grant no. 24QNPY025), in part by the Natural Science Foundation of Sichuan Province (grant no. 2025YFHZ0308).

References

1 

Luo J, Yang H and Song BL: Mechanisms and regulation of cholesterol homeostasis. Nat Rev Mol Cell Biol. 21:225–245. 2020. View Article : Google Scholar

2 

Ouimet M, Barrett TJ and Fisher EA: HDL and reverse cholesterol transport. Circ Res. 124:1505–1518. 2019. View Article : Google Scholar : PubMed/NCBI

3 

Kuzu OF, Noory MA and Robertson GP: The role of cholesterol in cancer. Cancer Res. 76:2063–2070. 2016. View Article : Google Scholar : PubMed/NCBI

4 

Cao D and Liu H: Dysregulated cholesterol regulatory genes in hepatocellular carcinoma. Eur J Med Res. 28:5802023. View Article : Google Scholar : PubMed/NCBI

5 

Geng F and Guo D: SREBF1/SREBP-1 concurrently regulates lipid synthesis and lipophagy to maintain lipid homeostasis and tumor growth. Autophagy. 20:1183–1185. 2024. View Article : Google Scholar :

6 

Goossens P, Rodriguez-Vita J, Etzerodt A, Masse M, Rastoin O, Gouirand V, Ulas T, Papantonopoulou O, Van Eck M, Auphan-Anezin N, et al: Membrane cholesterol efflux drives tumor-associated macrophage reprogramming and tumor progression. Cell Metab. 29:1376–1389.e4. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Spann NJ and Glass CK: Sterols and oxysterols in immune cell function. Nat Immunol. 14:893–900. 2013. View Article : Google Scholar : PubMed/NCBI

8 

Miller WL and Auchus RJ: The molecular biology, biochemistry, and physiology of human steroidogenesis and its disorders. Endocr Rev. 32:81–151. 2011. View Article : Google Scholar

9 

Simons K and Toomre D: Lipid rafts and signal transduction. Nat Rev Mol Cell Biol. 1:31–39. 2000. View Article : Google Scholar

10 

Riscal R, Skuli N and Simon MC: Even cancer cells watch their cholesterol! Mol Cell. 76:220–231. 2019. View Article : Google Scholar : PubMed/NCBI

11 

Ma X, Xiao L, Liu L, Ye L, Su P, Bi E, Wang Q, Yang M, Qian J and Yi Q: CD36-mediated ferroptosis dampens intratumoral CD8+ T cell effector function and impairs their antitumor ability. Cell Metab. 33:1001–1012.e5. 2021. View Article : Google Scholar

12 

Gabitova-Cornell L, Surumbayeva A, Peri S, Franco-Barraza J, Restifo D, Weitz N, Ogier C, Goldman AR, Hartman TR, Francescone R, et al: Cholesterol pathway inhibition induces TGF-β signaling to promote basal differentiation in pancreatic cancer. Cancer Cell. 38:567–583.e11. 2020. View Article : Google Scholar

13 

Liu W, Chakraborty B, Safi R, Kazmin D, Chang CY and McDonnell DP: Dysregulated cholesterol homeostasis results in resistance to ferroptosis increasing tumorigenicity and metastasis in cancer. Nat Commun. 12:51032021. View Article : Google Scholar : PubMed/NCBI

14 

Yan C, Zheng L, Jiang S, Yang H, Guo J, Jiang LY, Li T, Zhang H, Bai Y, Lou Y, et al: Exhaustion-associated cholesterol deficiency dampens the cytotoxic arm of antitumor immunity. Cancer Cell. 41:1276–1293.e11. 2023. View Article : Google Scholar : PubMed/NCBI

15 

Luo M, Bao L, Chen Y, Xue Y, Wang Y, Zhang B, Wang C, Corley CD, McDonald JG, Kumar A, et al: ZMYND8 is a master regulator of 27-hydroxycholesterol that promotes tumorigenicity of breast cancer stem cells. Sci Adv. 8:eabn52952022. View Article : Google Scholar : PubMed/NCBI

16 

Hu C, Qiao W, Li X, Ning ZK, Liu J, Dalangood S, Li H, Yu X, Zong Z, Wen Z and Gui J: Tumor-secreted FGF21 acts as an immune suppressor by rewiring cholesterol metabolism of CD8+T cells. Cell Metab. 36:630–647.e8. 2024. View Article : Google Scholar

17 

Yarmolinsky J, Bull CJ, Vincent EE, Robinson J, Walther A, Smith GD, Lewis SJ, Relton CL and Martin RM: Association between genetically proxied inhibition of HMG-CoA reductase and epithelial ovarian cancer. JAMA. 323:646–655. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Kawamura S, Matsushita Y, Kurosaki S, Tange M, Fujiwara N, Hayata Y, Hayakawa Y, Suzuki N, Hata M, Tsuboi M, et al: Inhibiting SCAP/SREBP exacerbates liver injury and carcinogenesis in murine nonalcoholic steatohepatitis. J Clin Invest. 132:e1518952022. View Article : Google Scholar : PubMed/NCBI

19 

Li C, Wang Y, Liu D, Wong CC, Coker OO, Zhang X, Liu C, Zhou Y, Liu Y, Kang W, et al: Squalene epoxidase drives cancer cell proliferation and promotes gut dysbiosis to accelerate colorectal carcinogenesis. Gut. 71:2253–2265. 2022. View Article : Google Scholar : PubMed/NCBI

20 

Fan J, Lin R, Xia S, Chen D, Elf SE, Liu S, Pan Y, Xu H, Qian Z, Wang M, et al: Tetrameric Acetyl-CoA acetyltransferase 1 is important for tumor growth. Mol Cell. 64:859–874. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Viaud M, Abdel-Wahab O, Gall J, Ivanov S, Guinamard R, Sore S, Merlin J, Ayrault M, Guilbaud E, Jacquel A, et al: ABCA1 exerts tumor-suppressor function in myeloproliferative neoplasms. Cell Rep. 30:3397–3410.e5. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Roundhill EA, Jabri S and Burchill SA: ABCG1 and Pgp identify drug resistant, self-renewing osteosarcoma cells. Cancer Lett. 453:142–157. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Kong W, Wei J, Abidi P, Lin M, Inaba S, Li C, Wang Y, Wang Z, Si S, Pan H, et al: Berberine is a novel cholesterol-lowering drug working through a unique mechanism distinct from statins. Nat Med. 10:1344–1351. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Ioannou GN, Morrow OB, Connole ML and Lee SP: Association between dietary nutrient composition and the incidence of cirrhosis or liver cancer in the United States population. Hepatology. 50:175–184. 2009. View Article : Google Scholar : PubMed/NCBI

25 

Xu R, Shen J, Song Y, Lu J, Liu Y, Cao Y, Wang Z and Zhang J: Exploration of the application potential of serum multi-biomarker model in colorectal cancer screening. Sci Rep. 14:101272024. View Article : Google Scholar : PubMed/NCBI

26 

Jamnagerwalla J, Howard LE, Allott EH, Vidal AC, Moreira DM, Castro-Santamaria R, Andriole GL, Freeman MR and Freedland SJ: Serum cholesterol and risk of high-grade prostate cancer: Results from the REDUCE study. Prostate Cancer Prostatic Dis. 21:252–259. 2018. View Article : Google Scholar :

27 

Zhao B, Gan L, Graubard BI, Männistö S, Albanes D and Huang J: Associations of dietary cholesterol, serum cholesterol, and egg consumption with overall and cause-specific mortality: Systematic review and updated meta-analysis. Circulation. 145:1506–1520. 2022. View Article : Google Scholar : PubMed/NCBI

28 

Demierre MF, Higgins PD, Gruber SB, Hawk E and Lippman SM: Statins and cancer prevention. Nat Rev Cancer. 5:930–942. 2005. View Article : Google Scholar : PubMed/NCBI

29 

Siemianowicz K, Gminski J, Stajszczyk M, Wojakowski W, Goss M, Machalski M, Telega A, Brulinski K and Magiera-Molendowska H: Serum total cholesterol and triglycerides levels in patients with lung cancer. Int J Mol Med. 5:201–205. 2000.PubMed/NCBI

30 

Zhou P, Li B, Liu B, Chen T and Xiao J: Prognostic role of serum total cholesterol and high-density lipoprotein cholesterol in cancer survivors: A systematic review and meta-analysis. Clin Chim Acta. 477:94–104. 2018. View Article : Google Scholar

31 

Oh MJ, Han K, Kim B, Lim JH, Kim B, Kim SG and Cho SJ: Risk of gastric cancer in relation with serum cholesterol profiles: A nationwide population-based cohort study. Medicine (Baltimore). 102:e362602023. View Article : Google Scholar : PubMed/NCBI

32 

Zhang X, Coker OO, Chu ES, Fu K, Lau HCH, Wang YX, Chan AWH, Wei H, Yang X, Sung JJY and Yu J: Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut. 70:761–774. 2021. View Article : Google Scholar

33 

Zhang X, Ding HM, Deng LF, Chen GC, Li J, He ZY, Fu L, Li JF, Jiang F, Zhang ZL and Li BY: Dietary fats and serum lipids in relation to the risk of ovarian cancer: A meta-analysis of observational studies. Front Nutr. 10:11539862023. View Article : Google Scholar : PubMed/NCBI

34 

Ikonen E and Olkkonen VM: Intracellular cholesterol trafficking. Cold Spring Harb Perspect Biol. 15:a0414042023. View Article : Google Scholar : PubMed/NCBI

35 

von Eckardstein A, Nordestgaard BG, Remaley AT and Catapano AL: High-density lipoprotein revisited: Biological functions and clinical relevance. Eur Heart J. 44:1394–1407. 2023. View Article : Google Scholar :

36 

Dang EV and Reboldi A: Cholesterol sensing and metabolic adaptation in tissue immunity. Trends Immunol. 45:861–870. 2024. View Article : Google Scholar : PubMed/NCBI

37 

Griffiths WJ and Wang Y: Cholesterol metabolism: From lipidomics to immunology. J Lipid Res. 63:1001652022. View Article : Google Scholar :

38 

Long T, Debler EW and Li X: Structural enzymology of cholesterol biosynthesis and storage. Curr Opin Struct Biol. 74:1023692022. View Article : Google Scholar : PubMed/NCBI

39 

Schade DS, Shey L and Eaton RP: Cholesterol review: A metabolically important molecule. Endocr Pract. 26:1514–1523. 2020. View Article : Google Scholar

40 

Halimi H and Farjadian S: Cholesterol: An important actor on the cancer immune scene. Front Immunol. 13:10575462022. View Article : Google Scholar : PubMed/NCBI

41 

Goicoechea L, Conde de la Rosa L, Torres S, García-Ruiz C and Fernández-Checa JC: Mitochondrial cholesterol: Metabolism and impact on redox biology and disease. Redox Biol. 61:1026432023. View Article : Google Scholar : PubMed/NCBI

42 

Meng Y, Heybrock S, Neculai D and Saftig P: Cholesterol handling in lysosomes and beyond. Trends Cell Biol. 30:452–466. 2020. View Article : Google Scholar : PubMed/NCBI

43 

Feingold KR: Lipid and lipoprotein metabolism. Endocrinol Metab Clin North Am. 51:437–458. 2022. View Article : Google Scholar : PubMed/NCBI

44 

Chandel NS: Lipid metabolism. Cold Spring Harb Perspect Biol. 13:a0405762021. View Article : Google Scholar : PubMed/NCBI

45 

Xiao X, Kennelly JP, Ferrari A, Clifford BL, Whang E, Gao Y, Qian K, Sandhu J, Jarrett KE, Brearley-Sholto MC, et al: Hepatic nonvesicular cholesterol transport is critical for systemic lipid homeostasis. Nat Metab. 5:165–181. 2023. View Article : Google Scholar : PubMed/NCBI

46 

Kennelly JP and Tontonoz P: Cholesterol transport to the endoplasmic reticulum. Cold Spring Harb Perspect Biol. 15:a0412632023. View Article : Google Scholar

47 

Faulkner RA, Yang Y, Tsien J, Qin T and DeBose-Boyd RA: Direct binding to sterols accelerates endoplasmic reticulum-associated degradation of HMG CoA reductase. Proc Natl Acad Sci USA. 121:e23188221212024. View Article : Google Scholar : PubMed/NCBI

48 

Chen L, Zhao ZW, Zeng PH, Zhou YJ and Yin WJ: Molecular mechanisms for ABCA1-mediated cholesterol efflux. Cell Cycle. 21:1121–1139. 2022. View Article : Google Scholar : PubMed/NCBI

49 

Hu X, Chen F, Jia L, Long A, Peng Y, Li X, Huang J, Wei X, Fang X, Gao Z, et al: A gut-derived hormone regulates cholesterol metabolism. Cell. 187:1685–1700.e18. 2024. View Article : Google Scholar : PubMed/NCBI

50 

Saher G: Cholesterol metabolism in aging and age-related disorders. Annu Rev Neurosci. 46:59–78. 2023. View Article : Google Scholar : PubMed/NCBI

51 

Xu H, Zhou S, Tang Q, Xia H and Bi F: Cholesterol metabolism: New functions and therapeutic approaches in cancer. Biochim Biophys Acta Rev Cancer. 1874:1883942020. View Article : Google Scholar : PubMed/NCBI

52 

Mittal S, Nenwani M, Pulikkal Kadamberi I, Kumar S, Animasahun O, George J, Tsaih SW, Gupta P, Singh M, Geethadevi A, et al: eIF4E enriched extracellular vesicles induce immunosuppressive macrophages through HMGCR-mediated metabolic rewiring. Adv Sci (Weinh). e063072025.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

53 

Ashida S, Kawada C and Inoue K: Stromal regulation of prostate cancer cell growth by mevalonate pathway enzymes HMGCS1 and HMGCR. Oncol Lett. 14:6533–6542. 2017.PubMed/NCBI

54 

Martin OP, Wallace MS, Oetheimer C, Patel HB, Butler MD, Wong LP, Huang P, Elbaz J, Costentin C, Salloum S, et al: Single-cell atlas of human liver and blood immune cells across fatty liver disease stages reveals distinct signatures linked to liver dysfunction and fibrogenesis. Nat Immunol. 26:1596–1611. 2025. View Article : Google Scholar : PubMed/NCBI

55 

Saito Y, Yin D, Kubota N, Wang X, Filliol A, Remotti H, Nair A, Fazlollahi L, Hoshida Y, Tabas I, et al: A therapeutically targetable TAZ-TEAD2 pathway drives the growth of hepatocellular carcinoma via ANLN and KIF23. Gastroenterology. 164:1279–1292. 2023. View Article : Google Scholar : PubMed/NCBI

56 

Li Z, Liu C, Wang M, Wei R, Li R, Huang K, Liang H, Li G and Zhao L: Cholesterol confers resistance to Apatinib-mediated ferroptosis in gastric cancer. Cell Biosci. 15:952025. View Article : Google Scholar : PubMed/NCBI

57 

Li MX, Hu S, Lei HH, Yuan M, Li X, Hou WK, Huang XJ, Xiao BW, Yu TX, Zhang XH, et al: Tumor-derived miR-9-5p-loaded EVs regulate cholesterol homeostasis to promote breast cancer liver metastasis in mice. Nat Commun. 15:105392024. View Article : Google Scholar : PubMed/NCBI

58 

Xue X, He Z, Liu F, Wang Q, Chen Z, Lin L, Chen D, Yuan Y, Huang Z and Wang Y: Taurochenodeoxycholic acid suppresses the progression of glioblastoma via HMGCS1/HMGCR/GPX4 signaling pathway in vitro and in vivo. Cancer Cell Int. 25:1602025. View Article : Google Scholar : PubMed/NCBI

59 

Nakagawa H: Lipogenesis and MASLD: Re-thinking the role of SREBPs. Arch Toxicol. 99:2299–2312. 2025. View Article : Google Scholar : PubMed/NCBI

60 

Li Y, Wu S, Zhao X, Hao S, Li F, Wang Y, Liu B, Zhang D, Wang Y and Zhou H: Key events in cancer: Dysregulation of SREBPs. Front Pharmacol. 14:11307472023. View Article : Google Scholar : PubMed/NCBI

61 

Geng F, Zhong Y, Su H, Lefai E, Magaki S, Cloughesy TF, Yong WH, Chakravarti A and Guo D: SREBP-1 upregulates lipophagy to maintain cholesterol homeostasis in brain tumor cells. Cell Rep. 42:1127902023. View Article : Google Scholar : PubMed/NCBI

62 

Chen Y, Deng X, Li Y, Han Y, Peng Y, Wu W, Wang X, Ma J, Hu E, Zhou X, et al: Comprehensive molecular classification predicted microenvironment profiles and therapy response for HCC. Hepatology. 80:536–551. 2024. View Article : Google Scholar : PubMed/NCBI

63 

Zhou J, Qu G, Zhang G, Wu Z, Liu J, Yang D, Li J, Chang M, Zeng H, Hu J, et al: Glycerol kinase 5 confers gefitinib resistance through SREBP1/SCD1 signaling pathway. J Exp Clin Cancer Res. 38:962019. View Article : Google Scholar : PubMed/NCBI

64 

Su F and Koeberle A: Regulation and targeting of SREBP-1 in hepatocellular carcinoma. Cancer Metastasis Rev. 43:673–708. 2024. View Article : Google Scholar :

65 

Chen F, Li H, Wang Y, Tang X, Lin K, Li Q, Meng C, Shi W, Leo J, Liang X, et al: CHD1 loss reprograms SREBP2-driven cholesterol synthesis to fuel androgen-responsive growth and castration resistance in SPOP-mutated prostate tumors. Nat Cancer. 6:854–873. 2025. View Article : Google Scholar : PubMed/NCBI

66 

Gao W, Guo X, Sun L, Gai J, Cao Y and Zhang S: PKMYT1 knockdown inhibits cholesterol biosynthesis and promotes the drug sensitivity of triple-negative breast cancer cells to atorvastatin. PeerJ. 12:e177492024. View Article : Google Scholar : PubMed/NCBI

67 

Wu R, Li N, Huang W, Yang Y, Zang R, Song H, Shi J, Zhu S and Liu Q: Melittin suppresses ovarian cancer growth by regulating SREBP1-mediated lipid metabolism. Phytomedicine. 137:1563672025. View Article : Google Scholar : PubMed/NCBI

68 

Wen J, Zhang X, Wong CC, Zhang Y, Pan Y, Zhou Y, Cheung AH, Liu Y, Ji F, Kang X, et al: Targeting squalene epoxidase restores anti-PD-1 efficacy in metabolic dysfunction-associated steatohepatitis-induced hepatocellular carcinoma. Gut. 73:2023–2036. 2024. View Article : Google Scholar : PubMed/NCBI

69 

Li J, Yang T, Wang Q, Li Y, Wu H, Zhang M, Qi H, Zhang H and Li J: Upregulation of SQLE contributes to poor survival in head and neck squamous cell carcinoma. Int J Biol Sci. 18:3576–3591. 2022. View Article : Google Scholar : PubMed/NCBI

70 

Liu Q, Zhang Y, Li H, Gao H, Zhou Y, Luo D, Shan Z, Yang Y, Weng J, Li Q, et al: Squalene epoxidase promotes the chemoresistance of colorectal cancer via (S)-2,3-epoxysqualene-activated NF-κB. Cell Commun Signal. 22:2782024. View Article : Google Scholar

71 

Wu J, Hu W, Yang W, Long Y, Chen K, Li F, Ma X and Li X: Knockdown of SQLE promotes CD8+ T cell infiltration in the tumor microenvironment. Cell Signal. 114:1109832024. View Article : Google Scholar

72 

Xu M, Pan G, Zhang Q, Huang J, Wu Y and Ashan Y: FOXM1 boosts glycolysis by upregulating SQLE to inhibit anoikis in breast cancer cells. J Cancer Res Clin Oncol. 151:1622025. View Article : Google Scholar : PubMed/NCBI

73 

Shen T, Lu Y and Zhang Q: High squalene epoxidase in tumors predicts worse survival in patients with hepatocellular carcinoma: Integrated bioinformatic analysis on NAFLD and HCC. Cancer Control. 27:10732748209146632020. View Article : Google Scholar : PubMed/NCBI

74 

Zhang Z, Wu W, Jiao H, Chen Y, Ji X, Cao J, Yin F and Yin W: Squalene epoxidase promotes hepatocellular carcinoma development by activating STRAP transcription and TGF-β/SMAD signalling. Br J Pharmacol. 180:1562–1581. 2023. View Article : Google Scholar

75 

Kanmalar M, Abdul Sani SF, Kamri NINB, Said NABM, Jamil AHBA, Kuppusamy S, Mun KS and Bradley DA: Raman spectroscopy biochemical characterisation of bladder cancer cisplatin resistance regulated by FDFT1: A review. Cell Mol Biol Lett. 27:92022. View Article : Google Scholar : PubMed/NCBI

76 

Cai D, Zhong GC, Dai X, Zhao Z, Chen M, Hu J, Wu Z, Cheng L, Li S and Gong J: Targeting FDFT1 reduces cholesterol and bile acid production and delays hepatocellular carcinoma progression through the HNF4A/ALDOB/AKT1 axis. Adv Sci (Weinh). 12:e24117192025. View Article : Google Scholar : PubMed/NCBI

77 

Wu Z, Duan W, Xiong Y, Liu J, Wen X, Zhao F, Xiang D, Wang J, Kasim V and Wu S: NeuroD1 drives a KAT2A-FDFT1 signaling axis to promote cholesterol biosynthesis and hepatocellular carcinoma progression via histone H3K27 acetylation. Oncogene. Sep 1–2025.Epub ahead of print.

78 

Yang C, Huang S, Cao F and Zheng Y: A lipid metabolism-related genes prognosis biomarker associated with the tumor immune microenvironment in colorectal carcinoma. BMC Cancer. 21:11822021. View Article : Google Scholar : PubMed/NCBI

79 

Zhang HL, Zhao R, Wang D, Mohd Sapudin SN, Yahaya BH, Harun MSR, Zhang ZW, Song ZJ, Liu YT, Doblin S and Lu P: Candida albicans and colorectal cancer: A paradoxical role revealed through metabolite profiling and prognostic modeling. World J Clin Oncol. 16:1041822025. View Article : Google Scholar : PubMed/NCBI

80 

Weng ML, Chen WK, Chen XY, Lu H, Sun ZR, Yu Q, Sun PF, Xu YJ, Zhu MM, Jiang N, et al: Fasting inhibits aerobic glycolysis and proliferation in colorectal cancer via the Fdft1-mediated AKT/mTOR/HIF1α pathway suppression. Nat Commun. 11:18692020. View Article : Google Scholar

81 

Goudarzi A: The recent insights into the function of ACAT1: A possible anti-cancer therapeutic target. Life Sci. 232:1165922019. View Article : Google Scholar : PubMed/NCBI

82 

Gu L, Zhu Y, Lin X, Tan X, Lu B and Li Y: Stabilization of FASN by ACAT1-mediated GNPAT acetylation promotes lipid metabolism and hepatocarcinogenesis. Oncogene. 39:2437–2449. 2020. View Article : Google Scholar : PubMed/NCBI

83 

Wang T, Wang G, Shan D, Fang Y, Zhou F, Yu M, Ju L, Li G, Xiang W, Qian K, et al: ACAT1 promotes proliferation and metastasis of bladder cancer via AKT/GSK3β/c-Myc signaling pathway. J Cancer. 15:3297–3312. 2024. View Article : Google Scholar :

84 

Sun S, Qi G, Chen H, He D, Ma D, Bie Y, Xu L, Feng B, Pang Q, Guo H and Zhang R: Ferroptosis sensitization in glioma: Exploring the regulatory mechanism of SOAT1 and its therapeutic implications. Cell Death Dis. 14:7542023. View Article : Google Scholar : PubMed/NCBI

85 

Wei C, Liao K, Chen HJ, Xiao ZX, Meng Q, Liu ZK, Lu YX, Sheng H, Mo HY, Wu QN, et al: Nuclear mitochondrial acetyl-CoA acetyltransferase 1 orchestrates natural killer cell-dependent antitumor immunity in colorectal cancer. Signal Transduct Target Ther. 10:1382025. View Article : Google Scholar : PubMed/NCBI

86 

Kwon RJ, Park EJ, Lee SY, Lee Y, Hwang C, Kim C and Cho YH: Expression and prognostic significance of Niemann-Pick C1-Like 1 in colorectal cancer: A retrospective cohort study. Lipids Health Dis. 20:1042021. View Article : Google Scholar : PubMed/NCBI

87 

Yin W, Ao Y, Jia Q, Zhang C, Yuan L, Liu S, Xiao W, Luo G, Shi X, Xin C, et al: Integrated singlecell and bulk RNA-seq analysis identifies a prognostic signature related to inflammation in colorectal cancer. Sci Rep. 15:8742025. View Article : Google Scholar : PubMed/NCBI

88 

Yin Y, Wu C, Zhou Y, Zhang M, Mai S, Chen M and Wang HY: Ezetimibe INDUCES PARAPTOSIS THROUGH NIEMANN-PICK C1-like 1 inhibition of mammalian-target-of-rapamycin signaling in hepatocellular carcinoma cells. Genes (Basel). 15:42023. View Article : Google Scholar

89 

Liu X, Lv M, Zhang W and Zhan Q: Dysregulation of cholesterol metabolism in cancer progression. Oncogene. 42:3289–3302. 2023. View Article : Google Scholar : PubMed/NCBI

90 

Guo XJ, Zhu BB, Li J, Guo P, Niu YB, Shi JL, Yokoyama W, Huang QS and Shao DY: Cholesterol metabolism in tumor immunity: Mechanisms and therapeutic opportunities for cancer. Biochem Pharmacol. 234:1168022025. View Article : Google Scholar : PubMed/NCBI

91 

Pan Z, Wang K, Wang X, Jia Z, Yang Y, Duan Y, Huang L, Wu ZX, Zhang JY and Ding X: Cholesterol promotes EGFR-TKIs resistance in NSCLC by inducing EGFR/Src/Erk/SP1 signaling-mediated ERRα re-expression. Mol Cancer. 21:772022. View Article : Google Scholar

92 

Rademaker G, Hernandez GA, Seo Y, Dahal S, Miller-Phillips L, Li AL, Peng XL, Luan C, Qiu L, Liegeois MA, et al: PCSK9 drives sterol-dependent metastatic organ choice in pancreatic cancer. Nature. 643:1381–1390. 2025. View Article : Google Scholar : PubMed/NCBI

93 

Fu R, Xue W, Liang J, Li X, Zheng J, Wang L, Zhang M and Meng J: SOAT1 regulates cholesterol metabolism to induce EMT in hepatocellular carcinoma. Cell Death Dis. 15:3252024. View Article : Google Scholar : PubMed/NCBI

94 

Alvarado-Ortiz E, de la Cruz-López KG, Becerril-Rico J, Sarabia-Sánchez MA, Ortiz-Sánchez E and García-Carrancá A: Mutant p53 gain-of-function: Role in cancer development, progression, and therapeutic approaches. Front Cell Dev Biol. 8:6076702021. View Article : Google Scholar : PubMed/NCBI

95 

Freed-Pastor WA, Mizuno H, Zhao X, Langerød A, Moon SH, Rodriguez-Barrueco R, Barsotti A, Chicas A, Li W, Polotskaia A, et al: Mutant p53 disrupts mammary tissue architecture via the mevalonate pathway. Cell. 148:244–258. 2012. View Article : Google Scholar : PubMed/NCBI

96 

Moon SH, Huang CH, Houlihan SL, Regunath K, Freed-Pastor WA, Morris JP IV, Tschaharganeh DF, Kastenhuber ER, Barsotti AM, Culp-Hill R, et al: p53 represses the mevalonate pathway to mediate tumor suppression. Cell. 176:564–580.e19. 2019. View Article : Google Scholar :

97 

Zhang Y, Mohibi S, Vasilatis DM, Chen M, Zhang J and Chen X: Ferredoxin reductase and p53 are necessary for lipid homeostasis and tumor suppression through the ABCA1-SREBP pathway. Oncogene. 41:1718–1726. 2022. View Article : Google Scholar : PubMed/NCBI

98 

Fan S, Guo J, Nie H, Xiong H and Xia Y: Aberrant energy metabolism in tumors and potential therapeutic targets. Genes Chromosomes Cancer. 63:e700082024. View Article : Google Scholar : PubMed/NCBI

99 

Da Cruz Paula A, Zhu Y, Brown DN, Issa Bhaloo S, Pareja F, Hoang TJ, Green H, Basili T, Dopeso H, Selenica P, et al: Evolution and co-occurrence of PI3K pathway gene mutations in endometrial carcinoma molecular subtypes at the single-cell level. Clin Cancer Res. August 20–2025.Epub ahead of print. View Article : Google Scholar : PubMed/NCBI

100 

Kaysudu I, Gungul TB, Atici S, Yilmaz S, Bayram E, Guven G, Cizmecioglu NT, Sahin O, Yesiloz G, Haznedaroglu BZ and Cizmecioglu O: Cholesterol biogenesis is a PTEN-dependent actionable node for the treatment of endocrine therapy-refractory cancers. Cancer Sci. 114:4365–4375. 2023. View Article : Google Scholar : PubMed/NCBI

101 

Fitzgerald TL, Lertpiriyapong K, Cocco L, Martelli AM, Libra M, Candido S, Montalto G, Cervello M, Steelman L, Abrams SL and McCubrey JA: Roles of EGFR and KRAS and their downstream signaling pathways in pancreatic cancer and pancreatic cancer stem cells. Adv Biol Regul. 59:65–81. 2015. View Article : Google Scholar : PubMed/NCBI

102 

Aylon Y and Oren M: The Hippo pathway, p53 and cholesterol. Cell Cycle. 15:2248–2255. 2016. View Article : Google Scholar : PubMed/NCBI

103 

Eid W, Dauner K, Courtney KC, Gagnon A, Parks RJ, Sorisky A and Zha X: mTORC1 activates SREBP-2 by suppressing cholesterol trafficking to lysosomes in mammalian cells. Proc Natl Acad Sci USA. 114:7999–8004. 2017. View Article : Google Scholar : PubMed/NCBI

104 

Deng YZ, Cai Z, Shi S, Jiang H, Shang YR, Ma N, Wang JJ, Guan DX, Chen TW, Rong YF, et al: Cilia loss sensitizes cells to transformation by activating the mevalonate pathway. J Exp Med. 215:177–195. 2018. View Article : Google Scholar :

105 

Das S, Parigi SM, Luo X, Fransson J, Kern BC, Okhovat A, Diaz OE, Sorini C, Czarnewski P, Webb AT, et al: Liver X receptor unlinks intestinal regeneration and tumorigenesis. Nature. 637:1198–1206. 2025. View Article : Google Scholar :

106 

Bakiri L, Hamacher R, Graña O, Guío-Carrión A, Campos-Olivas R, Martinez L, Dienes HP, Thomsen MK, Hasenfuss SC and Wagner EF: Liver carcinogenesis by FOS-dependent inflammation and cholesterol dysregulation. J Exp Med. 214:1387–1409. 2017. View Article : Google Scholar : PubMed/NCBI

107 

Deng CM, Zhang GG, Liu QW, Xu JJ, Liu ZC, Yang J, Xu TY, Li ZG, Zhang F and Li B: ANO1 reprograms cholesterol metabolism and the tumor microenvironment to promote cancer metastasis. Cancer Res. 83:1851–1865. 2023. View Article : Google Scholar : PubMed/NCBI

108 

Zhou YX, Wei J, Deng G, Hu A, Sun PY, Zhao X, Song BL and Luo J: Delivery of low-density lipoprotein from endocytic carriers to mitochondria supports steroidogenesis. Nat Cell Biol. 25:937–949. 2023. View Article : Google Scholar : PubMed/NCBI

109 

He W, Wang M, Zhang X, Wang Y, Zhao D, Li W, Lei F, Peng M, Zhang Z, Yuan Y and Huang Z: Estrogen induces LCAT to maintain cholesterol homeostasis and suppress hepatocellular carcinoma development. Cancer Res. 84:2417–2431. 2024. View Article : Google Scholar : PubMed/NCBI

110 

Xian M, Wang Q, Xiao L, Zhong L, Xiong W, Ye L, Su P, Zhang C, Li Y, Orlowski RZ, et al: Leukocyte immunoglobulin-like receptor B1 (LILRB1) protects human multiple myeloma cells from ferroptosis by maintaining cholesterol homeostasis. Nat Commun. 15:57672024. View Article : Google Scholar : PubMed/NCBI

111 

De Oliveira-Gomes D, Joshi PH, Peterson ED, Rohatgi A, Khera A and Navar AM: Apolipoprotein B: Bridging the gap between evidence and clinical practice. Circulation. 150:62–79. 2024. View Article : Google Scholar : PubMed/NCBI

112 

Gilliland T, Dron JS, Selvaraj MS, Trinder M, Paruchuri K, Urbut SM, Haidermota S, Bernardo R, Uddin MM, Honigberg MC, et al: Genetic architecture and clinical outcomes of combined lipid disturbances. Circ Res. 135:265–276. 2024. View Article : Google Scholar : PubMed/NCBI

113 

Lee G, Jeong YS, Kim DW, Kwak MJ, Koh J, Joo EW, Lee JS, Kah S, Sim YE and Yim SY: Clinical significance of APOB inactivation in hepatocellular carcinoma. Exp Mol Med. 50:1–12. 2018. View Article : Google Scholar : PubMed/NCBI

114 

Liu Z, Yuan H, Wang Y, Li K, Suo C, Jin L, Ding C and Chen X: Proteogenomic analysis identifies a causal association between plasma apolipoprotein B levels and liver cancer risk. J Proteome Res. 23:4055–4066. 2024. View Article : Google Scholar : PubMed/NCBI

115 

Borgquist S, Butt T, Almgren P, Shiffman D, Stocks T, Orho-Melander M, Manjer J and Melander O: Apolipoproteins, lipids and risk of cancer. Int J Cancer. 138:2648–2656. 2016. View Article : Google Scholar : PubMed/NCBI

116 

Guan Y, Liu X, Yang Z, Zhu X, Liu M, Du M, Pan X and Wang Y: PCSK9 promotes LDLR degradation by preventing SNX17-mediated LDLR recycling. Circulation. 151:1512–1526. 2025. View Article : Google Scholar : PubMed/NCBI

117 

Yuan J, Cai T, Zheng X, Ren Y, Qi J, Lu X, Chen H, Lin H, Chen Z, Liu M, et al: Potentiating CD8+ T cell antitumor activity by inhibiting PCSK9 to promote LDLR-mediated TCR recycling and signaling. Protein Cell. 12:240–260. 2021. View Article : Google Scholar : PubMed/NCBI

118 

Ma L, Zhang L, Li L and Zhao L: The function of lncRNA EMX2OS/miR-653-5p and its regulatory mechanism in lung adenocarcinoma. Open Med (Wars). 18:202306862023. View Article : Google Scholar : PubMed/NCBI

119 

Chen Z, Chen L, Sun B, Liu D, He Y, Qi L, Li G, Han Z, Zhan L, Zhang S, et al: LDLR inhibition promotes hepatocellular carcinoma proliferation and metastasis by elevating intracellular cholesterol synthesis through the MEK/ERK signaling pathway. Mol Metab. 51:1012302021. View Article : Google Scholar : PubMed/NCBI

120 

Zhu T, Tan JZA, Zhang L, Huang H, Das SS, Cheng F, Padmanabhan P, Jones MJK, Lee M, Lee A, et al: FTO suppresses DNA repair by inhibiting PARP1. Nat Commun. 16:29252025. View Article : Google Scholar : PubMed/NCBI

121 

Alemasova EE and Lavrik OI: Poly(ADP-ribosyl)ation by PARP1: Reaction mechanism and regulatory proteins. Nucleic Acids Res. 47:3811–3827. 2019. View Article : Google Scholar : PubMed/NCBI

122 

He Z, Gong S, Zhang X, Li J, Xue J, Zeng Q, Nie J, Zhang Z, Ding H, Pei H and Li B: Activated PARP1/FAK/COL5A1 signaling facilitates the tumorigenesis of cholesterol-resistant ovarian cancer cells through promoting EMT. Cell Signal. 124:1114192024. View Article : Google Scholar : PubMed/NCBI

123 

Diestel A, Aktas O, Hackel D, Hake I, Meier S, Raine CS, Nitsch R, Zipp F and Ullrich O: Activation of microglial poly(ADP-ribose)-polymerase-1 by cholesterol breakdown products during neuroinflammation: A link between demyelination and neuronal damage. J Exp Med. 198:1729–1740. 2003. View Article : Google Scholar : PubMed/NCBI

124 

Farez MF, Quintana FJ, Gandhi R, Izquierdo G, Lucas M and Weiner HL: Toll-like receptor 2 and poly(ADP-ribose) polymerase 1 promote central nervous system neuroinflammation in progressive EAE. Nat Immunol. 10:958–964. 2009. View Article : Google Scholar : PubMed/NCBI

125 

Lee DH, Nam YJ, Lee MS, Sohn DS and Lee CS: Rotundarpene attenuates cholesterol oxidation product-induced apoptosis by suppressing the mitochondrial pathway and the caspase-8- and bid-dependent pathways. Eur J Pharmacol. 749:39–48. 2015. View Article : Google Scholar : PubMed/NCBI

126 

Batnasan E, Wang R, Wen J, Ke Y, Li X, Bohio AA, Zeng X, Huo H, Han L, Boldogh I and Ba X: 17-Beta estradiol inhibits oxidative stress-induced accumulation of AIF into nucleolus and PARP1-dependent cell death via estrogen receptor alpha. Toxicol Lett. 232:1–9. 2015. View Article : Google Scholar

127 

Wang D, Li Y, Wang N, Luo G, Wang J, Luo C, Yu W and Hao L: 1α,25-Dihydroxyvitamin D3 prevents renal oxidative damage via the PARP1/SIRT1/NOX4 pathway in Zucker diabetic fatty rats. Am J Physiol Endocrinol Metab. 318:E343–E356. 2020. View Article : Google Scholar

128 

Allott EH, Howard LE, Cooperberg MR, Kane CJ, Aronson WJ, Terris MK, Amling CL and Freedland SJ: Serum lipid profile and risk of prostate cancer recurrence: Results from the SEARCH database. Cancer Epidemiol Biomarkers Prev. 23:2349–2356. 2014. View Article : Google Scholar : PubMed/NCBI

129 

Xu R, Song J, Ruze R, Chen Y, Yin X, Wang C and Zhao Y: SQLE promotes pancreatic cancer growth by attenuating ER stress and activating lipid rafts-regulated Src/PI3K/Akt signaling pathway. Cell Death Dis. 14:4972023. View Article : Google Scholar : PubMed/NCBI

130 

Zhang Z, Huang H, Chen Z, Yan M, Lu C, Xu Z and Li Z: Helicobacter pylori promotes gastric cancer through CagA-mediated mitochondrial cholesterol accumulation by targeting CYP11A1 redistribution. Int J Biol Sci. 20:4007–4028. 2024. View Article : Google Scholar : PubMed/NCBI

131 

Dhanasekaran R, Deutzmann A, Mahauad-Fernandez WD, Hansen AS, Gouw AM and Felsher DW: The MYC oncogene-the grand orchestrator of cancer growth and immune evasion. Nat Rev Clin Oncol. 19:23–36. 2022. View Article : Google Scholar

132 

Yang F, Kou J, Liu Z, Li W and Du W: MYC enhances cholesterol biosynthesis and supports cell proliferation through SQLE. Front Cell Dev Biol. 9:6558892021. View Article : Google Scholar : PubMed/NCBI

133 

Tan SH, Tan TK, Yokomori R, Liao M, Huang XZ, Yeoh AEJ and Sanda T: TAL1 hijacks MYCN enhancer that induces MYCN expression and dependence on mevalonate pathway in T-cell acute lymphoblastic leukemia. Leukemia. 37:1969–1981. 2023. View Article : Google Scholar : PubMed/NCBI

134 

Wu Y, Chen K and Liu X, Huang L, Zhao D, Li L, Gao M, Pei D, Wang C and Liu X: Srebp-1 interacts with c-Myc to enhance somatic cell reprogramming. Stem Cells. 34:83–92. 2016. View Article : Google Scholar

135 

Zhou X, Wang G, Tian C, Du L, Prochownik EV and Li Y: Inhibition of DUSP18 impairs cholesterol biosynthesis and promotes anti-tumor immunity in colorectal cancer. Nat Commun. 15:58512024. View Article : Google Scholar : PubMed/NCBI

136 

Liu F, Chen W, Zhang Z, Zeng W, Hu H, Ning S, Liao Z, Liu Y, Zhang H, Fu Q, et al: Targeting Aurora kinase B regulates cholesterol metabolism and enhances chemoimmunotherapy in cholangiocarcinoma. Gut. July 31–2025.Epub ahead of print. View Article : Google Scholar

137 

Gui L, Chen K, Yan J, Chen P, Gao WQ and Ma B: Targeting the mevalonate pathway potentiates NUAK1 inhibition-induced immunogenic cell death and antitumor immunity. Cell Rep Med. 6:1019132025. View Article : Google Scholar : PubMed/NCBI

138 

Singh PK and Mehla K: LXR signaling-mediated cholesterol metabolism reprogramming regulates cancer cell metastasis. Cancer Res. 83:1759–1761. 2023. View Article : Google Scholar : PubMed/NCBI

139 

Mullen PJ, Yu R, Longo J, Archer MC and Penn LZ: The interplay between cell signalling and the mevalonate pathway in cancer. Nat Rev Cancer. 16:718–731. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Seidah NG and Prat A: The multifaceted biology of PCSK9. Endocr Rev. 43:558–582. 2022. View Article : Google Scholar : PubMed/NCBI

141 

Collins R, Reith C, Emberson J, Armitage J, Baigent C, Blackwell L, Blumenthal R, Danesh J, Smith GD, DeMets D, et al: Interpretation of the evidence for the efficacy and safety of statin therapy. Lancet. 388:2532–2561. 2016. View Article : Google Scholar : PubMed/NCBI

142 

Xie W, Peng M, Liu Y, Zhang B, Yi L and Long Y: Simvastatin induces pyroptosis via ROS/caspase-1/GSDMD pathway in colon cancer. Cell Commun Signal. 21:3292023. View Article : Google Scholar : PubMed/NCBI

143 

Choi J, Nguyen VH, Przybyszewski E, Song J, Carroll A, Michta M, Almazan E, Simon TG and Chung RT: Statin use and risk of hepatocellular carcinoma and liver fibrosis in chronic liver disease. JAMA Intern Med. 185:522–530. 2025. View Article : Google Scholar : PubMed/NCBI

144 

Nowakowska MK, Lei X, Thompson MT, Shaitelman SF, Wehner MR, Woodward WA, Giordano SH and Nead KT: Association of statin use with clinical outcomes in patients with triple-negative breast cancer. Cancer. 127:4142–4150. 2021. View Article : Google Scholar : PubMed/NCBI

145 

Hemmati S, Saeidikia Z, Seradj H and Mohagheghzadeh A: Immunomodulatory peptides as vaccine adjuvants and antimicrobial agents. Pharmaceuticals (Basel). 17:2012024. View Article : Google Scholar : PubMed/NCBI

146 

Pospiech M, Owens SE, Miller DJ, Austin-Muttitt K, Mullins JGL, Cronin JG, Allemann RK and Sheldon IM: Bisphosphonate inhibitors of squalene synthase protect cells against cholesterol-dependent cytolysins. FASEB J. 35:e216402021. View Article : Google Scholar : PubMed/NCBI

147 

Toyota Y, Yoshioka H, Sagimori I, Hashimoto Y and Ohgane K: Bisphosphonate esters interact with HMG-CoA reductase membrane domain to induce its degradation. Bioorg Med Chem. 28:1155762020. View Article : Google Scholar : PubMed/NCBI

148 

Van Acker HH, Anguille S, Willemen Y, Smits EL and Van Tendeloo VF: Bisphosphonates for cancer treatment: Mechanisms of action and lessons from clinical trials. Pharmacol Ther. 158:24–40. 2016. View Article : Google Scholar

149 

Yoneda T, Michigami T, Yi B, Williams PJ, Niewolna M and Hiraga T: Actions of bisphosphonate on bone metastasis in animal models of breast carcinoma. Cancer. 88:2979–2988. 2000. View Article : Google Scholar : PubMed/NCBI

150 

D'Oronzo S, Gregory W, Nicholson S, Chong YK, Brown J and Coleman R: Natural history of stage II/III breast cancer, bone metastasis and the impact of adjuvant zoledronate on distribution of recurrences. J Bone Oncol. 28:1003672021. View Article : Google Scholar : PubMed/NCBI

151 

Liu D, Wong CC, Fu L, Chen H, Zhao L, Li C, Zhou Y, Zhang Y, Xu W, Yang Y, et al: Squalene epoxidase drives NAFLD-induced hepatocellular carcinoma and is a pharmaceutical target. Sci Transl Med. 10:eaap98402018. View Article : Google Scholar : PubMed/NCBI

152 

Wang Y, Ma X, Xu E, Huang Z, Yang C, Zhu K, Dong Y and Zhang C: Identifying squalene epoxidase as a metabolic vulnerability in high-risk osteosarcoma using an artificial intelligence-derived prognostic index. Clin Transl Med. 14:e15862024. View Article : Google Scholar : PubMed/NCBI

153 

Ma L, Huang W, Liang X, Bai G, Wang X, Jiang H, Xin Y, Hu L, Chen X and Liu C: Inhibition of squalene epoxidase linking with PI3K/AKT signaling pathway suppresses endometrial cancer. Cancer Sci. 114:3595–3607. 2023. View Article : Google Scholar : PubMed/NCBI

154 

Kalogirou C, Linxweiler J, Schmucker P, Snaebjornsson MT, Schmitz W, Wach S, Krebs M, Hartmann E, Puhr M, Müller A, et al: MiR-205-driven downregulation of cholesterol biosynthesis through SQLE-inhibition identifies therapeutic vulnerability in aggressive prostate cancer. Nat Commun. 12:50662021. View Article : Google Scholar : PubMed/NCBI

155 

Hu LP, Huang W, Wang X, Xu C, Qin WT, Li D, Tian G, Li Q, Zhou Y, Chen S, et al: Terbinafine prevents colorectal cancer growth by inducing dNTP starvation and reducing immune suppression. Mol Ther. 30:3284–3299. 2022. View Article : Google Scholar : PubMed/NCBI

156 

Zhang EB, Zhang X, Wang K, Zhang F, Chen TW, Ma N, Ni QZ, Wang YK, Zheng QW, Cao HJ, et al: Antifungal agent terbinafine restrains tumor growth in preclinical models of hepatocellular carcinoma via AMPK-mTOR axis. Oncogene. 40:5302–5313. 2021. View Article : Google Scholar : PubMed/NCBI

157 

Loibl S, Poortmans P, Morrow M, Denkert C and Curigliano G: Breast cancer. Lancet. 397:1750–1769. 2021. View Article : Google Scholar : PubMed/NCBI

158 

He Q, Kong L, Shi W, Ma D, Liu K, Yang S, Xin Q, Jiang C and Wu J: Ezetimibe inhibits triple-negative breast cancer proliferation and promotes cell cycle arrest by targeting the PDGFR/AKT pathway. Heliyon. 9:e213432023. View Article : Google Scholar : PubMed/NCBI

159 

Wang Y, You S, Su S, Yeon A, Lo EM, Kim S, Mohler JL, Freeman MR and Kim HL: Cholesterol-lowering intervention decreases mTOR complex 2 signaling and enhances antitumor immunity. Clin Cancer Res. 28:414–424. 2022. View Article : Google Scholar :

160 

Bovenga F, Sabbà C and Moschetta A: Uncoupling nuclear receptor LXR and cholesterol metabolism in cancer. Cell Metab. 21:517–526. 2015. View Article : Google Scholar : PubMed/NCBI

161 

Liang H and Shen X: LXR activation radiosensitizes non-small cell lung cancer by restricting myeloid-derived suppressor cells. Biochem Biophys Res Commun. 528:330–335. 2020. View Article : Google Scholar : PubMed/NCBI

162 

Wan W, Hou Y, Wang K, Cheng Y, Pu X and Ye X: The LXR-623-induced long non-coding RNA LINC01125 suppresses the proliferation of breast cancer cells via PTEN/AKT/p53 signaling pathway. Cell Death Dis. 10:2482019. View Article : Google Scholar : PubMed/NCBI

163 

Munir MT, Ponce C, Santos JM, Sufian HB, Al-Harrasi A, Gollahon LS, Hussain F and Rahman SM: VD3 and LXR agonist (T0901317) combination demonstrated greater potency in inhibiting cholesterol accumulation and inducing apoptosis via ABCA1-CHOP-BCL-2 cascade in MCF-7 breast cancer cells. Mol Biol Rep. 47:7771–7782. 2020. View Article : Google Scholar : PubMed/NCBI

164 

Tavazoie MF, Pollack I, Tanqueco R, Ostendorf BN, Reis BS, Gonsalves FC, Kurth I, Andreu-Agullo C, Derbyshire ML, Posada J, et al: LXR/ApoE activation restricts innate immune suppression in cancer. Cell. 172:825–840.e18. 2018. View Article : Google Scholar : PubMed/NCBI

165 

Gronich N and Rennert G: Beyond aspirin-cancer prevention with statins, metformin and bisphosphonates. Nat Rev Clin Oncol. 10:625–642. 2013. View Article : Google Scholar : PubMed/NCBI

166 

Kamal A, Boerner J, Assad H, Chen W and Simon MS: The effect of statins on markers of breast cancer proliferation and apoptosis in women with in situ or early-stage invasive breast cancer. Int J Mol Sci. 25:95872024. View Article : Google Scholar : PubMed/NCBI

167 

Graf H, Jüngst C, Straub G, Dogan S, Hoffmann RT, Jakobs T, Reiser M, Waggershauser T, Helmberger T, Walter A, et al: Chemoembolization combined with pravastatin improves survival in patients with hepatocellular carcinoma. Digestion. 78:34–38. 2008. View Article : Google Scholar : PubMed/NCBI

168 

Kim Y, Kim TW, Han SW, Ahn JB, Kim ST, Lee J, Park JO, Park YS, Lim HY and Kang WK: A single arm, phase II study of simvastatin plus XELOX and bevacizumab as first-line chemotherapy in metastatic colorectal cancer patients. Cancer Res Treat. 51:1128–1134. 2019. View Article : Google Scholar :

169 

Jian-Yu E, Graber JM, Lu SE, Lin Y, Lu-Yao G and Tan XL: Effect of metformin and statin use on survival in pancreatic cancer patients: A systematic literature review and meta-analysis. Curr Med Chem. 25:2595–2607. 2018. View Article : Google Scholar

170 

Nguyen MKL, Jose J, Wahba M, Bernaus-Esqué M, Hoy AJ, Enrich C, Rentero C and Grewal T: Linking late endosomal cholesterol with cancer progression and anticancer drug resistance. Int J Mol Sci. 23:72062022. View Article : Google Scholar : PubMed/NCBI

171 

Du X, Zhang Y, Jo SR, Liu X, Qi Y, Osborne B, Byrne FL, Smith GC, Turner N, Hoehn KL, et al: Akt activation increases cellular cholesterol by promoting the proteasomal degradation of Niemann-Pick C1. Biochem J. 471:243–253. 2015. View Article : Google Scholar : PubMed/NCBI

172 

Chen K, Zhang X, Sun G, Fang Z, Liao L, Zhong Y, Huang F, Dong M and Luo S: Focusing on the abnormal events of NPC1, NPC2, and NPC1L1 in pan-cancer and further constructing LUAD and KICH prediction models. J Proteome Res. 23:449–464. 2024. View Article : Google Scholar

173 

Liang B, Wu Q, Wang Y, Shi Y, Sun F, Huang Q, Li G, Liu Y, Zhang S, Xu X, et al: Cdc42-driven endosomal cholesterol transport promotes collateral resistance in HER2-positive gastric cancer. Cancer Lett. 587:2167022024. View Article : Google Scholar : PubMed/NCBI

174 

Lin MH, Chao TC, Lee CC, Tung CJ, Yeh CY and Hong JH: Measurement-based Monte Carlo dose calculation system for IMRT pretreatment and on-line transit dose verifications. Med Phys. 36:1167–1175. 2009. View Article : Google Scholar : PubMed/NCBI

175 

Liang D, Minikes AM and Jiang X: Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell. 82:2215–2227. 2022. View Article : Google Scholar : PubMed/NCBI

176 

Jiang X, Stockwell BR and Conrad M: Ferroptosis: Mechanisms, biology and role in disease. Nat Rev Mol Cell Biol. 22:266–282. 2021. View Article : Google Scholar : PubMed/NCBI

177 

Lei G, Zhuang L and Gan B: Targeting ferroptosis as a vulnerability in cancer. Nat Rev Cancer. 22:381–396. 2022. View Article : Google Scholar : PubMed/NCBI

178 

Li Y, Ran Q, Duan Q, Jin J, Wang Y, Yu L, Wang C, Zhu Z, Chen X, Weng L, et al: 7-Dehydrocholesterol dictates ferroptosis sensitivity. Nature. 626:411–418. 2024. View Article : Google Scholar : PubMed/NCBI

179 

Song YH, Lei HX, Yu D, Zhu H, Hao MZ, Cui RH, Meng XS, Sheng XH and Zhang L: Endogenous chemicals guard health through inhibiting ferroptotic cell death. Biofactors. 50:266–293. 2024. View Article : Google Scholar

180 

Lee J and Roh JL: Cholesterol-ferroptosis nexus: Unveiling novel cancer therapeutic avenues. Cancer Lett. 597:2170462024. View Article : Google Scholar : PubMed/NCBI

181 

Mao X, Xiong J, Cai M, Wang C, He Q, Wang B, Chen J, Xiao Z, Wang B, Han S and Zhang Y: SCARB1 links cholesterol metabolism-mediated ferroptosis inhibition to radioresistance in tumor cells. J Adv Res. January 18–2025.Epub ahead of print. View Article : Google Scholar

182 

Garcia-Bermudez J, Baudrier L, Bayraktar EC, Shen Y, La K, Guarecuco R, Yucel B, Fiore D, Tavora B, Freinkman E, et al: Squalene accumulation in cholesterol auxotrophic lymphomas prevents oxidative cell death. Nature. 567:118–122. 2019. View Article : Google Scholar : PubMed/NCBI

183 

Wang Y, Calvert AE, Cardenas H, Rink JS, Nahotko D, Qiang W, Ndukwe CE, Chen F, Keathley R, Zhang Y, et al: Nanoparticle targeting in chemo-resistant ovarian cancer reveals dual axis of therapeutic vulnerability involving cholesterol uptake and cell redox balance. Adv Sci (Weinh). 11:e23052122024. View Article : Google Scholar : PubMed/NCBI

184 

Riscal R, Bull CJ, Mesaros C, Finan JM, Carens M, Ho ES, Xu JP, Godfrey J, Brennan P, Johansson M, et al: Cholesterol auxotrophy as a targetable vulnerability in clear cell renal cell carcinoma. Cancer Discovery. 11:3106–3125. 2021. View Article : Google Scholar : PubMed/NCBI

185 

Huang B, Song BL and Xu C: Cholesterol metabolism in cancer: Mechanisms and therapeutic opportunities. Nat Metab. 2:132–141. 2020. View Article : Google Scholar : PubMed/NCBI

186 

Yuan J, Lv T, Yang J, Wu Z, Yan L, Yang J and Shi Y: HDLBP-stabilized lncFAL inhibits ferroptosis vulnerability by diminishing Trim69-dependent FSP1 degradation in hepatocellular carcinoma. Redox Biol. 58:1025462022. View Article : Google Scholar : PubMed/NCBI

187 

Jin H, Zhu M, Zhang D, Liu X, Guo Y, Xia L, Chen Y, Chen Y, Xu R, Liu C, et al: B7H3 increases ferroptosis resistance by inhibiting cholesterol metabolism in colorectal cancer. Cancer Sci. 114:4225–4236. 2023. View Article : Google Scholar : PubMed/NCBI

188 

Noguchi N, Saito Y and Niki E: Lipid peroxidation, ferroptosis, and antioxidants. Free Radic Biol Med. 237:228–238. 2025. View Article : Google Scholar : PubMed/NCBI

189 

Zhao X, Lian X, Xie J and Liu G: Accumulated cholesterol protects tumours from elevated lipid peroxidation in the microenvironment. Redox Biol. 62:1026782023. View Article : Google Scholar : PubMed/NCBI

190 

Li Y, Li Z, Ran Q and Wang P: Sterols in ferroptosis: From molecular mechanisms to therapeutic strategies. Trends Mol Med. 31:36–49. 2025. View Article : Google Scholar

191 

Bai T, Xue P, Shao S, Yan S and Zeng X: Cholesterol depletion-enhanced ferroptosis and immunotherapy via engineered nanozyme. Adv Sci (Weinh). 11:e24058262024. View Article : Google Scholar : PubMed/NCBI

192 

Vitalakumar D, Sharma A and Flora SJS: Ferroptosis: A potential therapeutic target for neurodegenerative diseases. J Biochem Mol Toxicol. 35:e228302021. View Article : Google Scholar : PubMed/NCBI

193 

Shimada K, Skouta R, Kaplan A, Yang WS, Hayano M, Dixon SJ, Brown LM, Valenzuela CA, Wolpaw AJ and Stockwell BR: Global survey of cell death mechanisms reveals metabolic regulation of ferroptosis. Nat Chem Biol. 12:497–503. 2016. View Article : Google Scholar : PubMed/NCBI

194 

Yao X, Xie R, Cao Y, Tang J, Men Y, Peng H and Yang W: Simvastatin induced ferroptosis for triple-negative breast cancer therapy. J Nanobiotechnology. 19:3112021. View Article : Google Scholar : PubMed/NCBI

195 

Tang WJ, Xu D, Liang MX, Wo GQ, Chen WQ, Tang JH and Zhang W: Pitavastatin induces autophagy-dependent ferroptosis in MDA-MB-231 cells via the mevalonate pathway. Heliyon. 10:e270842024. View Article : Google Scholar

196 

Zhang R, Zhang L, Fan S and Wang L, Wang B and Wang L: Squalene monooxygenase (SQLE) protects ovarian cancer cells from ferroptosis. Sci Rep. 14:226462024. View Article : Google Scholar : PubMed/NCBI

197 

Mao X, Wang L, Chen Z, Huang H, Chen J, Su J, Li Z, Shen G, Ren Y, Li Z, et al: SCD1 promotes the stemness of gastric cancer stem cells by inhibiting ferroptosis through the SQLE/cholesterol/mTOR signalling pathway. Int J Biol Macromol. 275:1336982024. View Article : Google Scholar : PubMed/NCBI

198 

Debnath J, Gammoh N and Ryan KM: Autophagy and autophagy-related pathways in cancer. Nat Rev Mol Cell Biol. 24:560–575. 2023. View Article : Google Scholar : PubMed/NCBI

199 

Vargas JNS, Hamasaki M, Kawabata T, Youle RJ and Yoshimori T: The mechanisms and roles of selective autophagy in mammals. Nat Rev Mol Cell Biol. 24:167–185. 2023. View Article : Google Scholar

200 

Yamamoto H, Zhang S and Mizushima N: Autophagy genes in biology and disease. Nat Rev Genet. 24:382–400. 2023. View Article : Google Scholar : PubMed/NCBI

201 

Rybstein MD, Bravo-San Pedro JM, Kroemer G and Galluzzi L: The autophagic network and cancer. Nat Cell Biol. 20:243–251. 2018. View Article : Google Scholar : PubMed/NCBI

202 

Castellano BM, Thelen AM, Moldavski O, Feltes M, van der Welle RE, Mydock-McGrane L, Jiang X, van Eijkeren RJ, Davis OB, Louie SM, et al: Lysosomal cholesterol activates mTORC1 via an SLC38A9-Niemann-Pick C1 signaling complex. Science. 355:1306–1311. 2017. View Article : Google Scholar : PubMed/NCBI

203 

Liu F, Tian T, Zhang Z, Xie S, Yang J, Zhu L, Wang W, Shi C, Sang L, Guo K, et al: Long non-coding RNA SNHG6 couples cholesterol sensing with mTORC1 activation in hepatocellular carcinoma. Nat Metab. 4:1022–1040. 2022. View Article : Google Scholar : PubMed/NCBI

204 

Podar K, Tai YT, Cole CE, Hideshima T, Sattler M, Hamblin A, Mitsiades N, Schlossman RL, Davies FE, Morgan GJ, et al: Essential role of caveolae in interleukin-6- and insulin-like growth factor I-triggered Akt-1-mediated survival of multiple myeloma cells. J Biol Chem. 278:5794–5801. 2003. View Article : Google Scholar

205 

Vilimanovich U, Bosnjak M, Bogdanovic A, Markovic I, Isakovic A, Kravic-Stevovic T, Mircic A, Trajkovic V and Bumbasirevic V: Statin-mediated inhibition of cholesterol synthesis induces cytoprotective autophagy in human leukemic cells. Eur J Pharmacol. 765:415–428. 2015. View Article : Google Scholar : PubMed/NCBI

206 

Yi J, Zhu J, Wu J, Thompson CB and Jiang X: Oncogenic activation of PI3K-AKT-mTOR signaling suppresses ferroptosis via SREBP-mediated lipogenesis. Proc Natl Acad Sci USA. 117:31189–31197. 2020. View Article : Google Scholar : PubMed/NCBI

207 

Guo Y, Zhao M, Bo T, Ma S, Yuan Z, Chen W, He Z, Hou X, Liu J, Zhang Z, et al: Blocking FSH inhibits hepatic cholesterol biosynthesis and reduces serum cholesterol. Cell Res. 29:151–166. 2019. View Article : Google Scholar :

208 

Jun I, Choi YJ, Kim BR, Lee HK, Seo KY and Kim TI: Activation of the mTOR pathway enhances PPARγ/SREBP-mediated lipid synthesis in human meibomian gland epithelial cells. Sci Rep. 14:281182024. View Article : Google Scholar

209 

Hsu JL, Leu WJ, Zhong NS and Guh JH: Autophagic activation and decrease of plasma membrane cholesterol contribute to anticancer activities in non-small cell lung cancer. Molecules. 26:59672021. View Article : Google Scholar : PubMed/NCBI

210 

Zhong Y, Geng F, Mazik L, Yin X, Becker AP, Mohammed S, Su H, Xing E, Kou Y, Chiang CY, et al: Combinatorial targeting of glutamine metabolism and lysosomal-based lipid metabolism effectively suppresses glioblastoma. Cell Rep Med. 5:1017062024. View Article : Google Scholar : PubMed/NCBI

211 

Ng MYW, Charsou C, Lapao A, Singh S, Trachsel-Moncho L, Schultz SW, Nakken S, Munson MJ and Simonsen A: The cholesterol transport protein GRAMD1C regulates autophagy initiation and mitochondrial bioenergetics. Nat Commun. 13:62832022. View Article : Google Scholar : PubMed/NCBI

212 

Shi Y, Ye Z, Lu G, Yang N, Zhang J, Wang L, Cui J, Del Pozo MA, Wu Y, Xia D and Shen HM: Cholesterol-enriched membrane micro-domaindeficiency induces doxorubicin resistancevia promoting autophagy in breast cancer. Mol Ther Oncolytics. 23:311–329. 2021. View Article : Google Scholar : PubMed/NCBI

213 

Shao WQ, Zhu WW, Luo MJ, Fan MH, Li Q, Wang SH, Lin ZF, Zhao J, Zheng Y, Dong QZ, et al: Cholesterol suppresses GOLM1-dependent selective autophagy of RTKs in hepatocellular carcinoma. Cell Rep. 39:1107122022. View Article : Google Scholar : PubMed/NCBI

214 

Yang J, Peng S and Zhang K: ARL4C depletion suppresses the resistance of ovarian cancer to carboplatin by disrupting cholesterol transport and autophagy via notch-RBP-Jκ-H3K4Me3-OSBPL5. Hum Exp Toxicol. 41:96032712211350642022. View Article : Google Scholar

215 

McBrearty N, Cho C, Chen J, Zahedi F, Peck AR, Radaelli E, Assenmacher CA, Pavlak C, Devine A, Yu P, et al: Tumor-suppressive and immune-stimulating roles of cholesterol 25-hydroxylase in pancreatic cancer cells. Mol Cancer Res. 21:228–239. 2023. View Article : Google Scholar :

216 

Liu C, Sun L, Niu N, Hou P, Chen G, Wang H, Zhang Z, Jiang X, Xu Q, Zhao Y, et al: Molecular classification of hormone receptor-positive/HER2-positive breast cancer reveals potential neoadjuvant therapeutic strategies. Signal Transduct Target Ther. 10:972025. View Article : Google Scholar

217 

Leignadier J, Dalenc F, Poirot M and Silvente-Poirot S: Improving the efficacy of hormone therapy in breast cancer: The role of cholesterol metabolism in SERM-mediated autophagy, cell differentiation and death. Biochem Pharmacol. 144:18–28. 2017. View Article : Google Scholar : PubMed/NCBI

218 

de Medina P, Paillasse MR, Ségala G, Khallouki F, Brillouet S, Dalenc F, Courbon F, Record M, Poirot M and Silvente-Poirot S: Importance of cholesterol and oxysterols metabolism in the pharmacology of tamoxifen and other AEBS ligands. Chem Phys Lipids. 164:432–437. 2011. View Article : Google Scholar : PubMed/NCBI

219 

Mittal V: Epithelial mesenchymal transition in tumor metastasis. Annu Rev Pathol. 13:395–412. 2018. View Article : Google Scholar : PubMed/NCBI

220 

Huang Y, Hong W and Wei X: The molecular mechanisms and therapeutic strategies of EMT in tumor progression and metastasis. J Hematol Oncol. 15:1292022. View Article : Google Scholar : PubMed/NCBI

221 

Jiang S, Wang X, Song D, Liu X, Gu Y, Xu Z, Wang X, Zhang X, Ye Q, Tong Z, et al: Cholesterol induces epithelial-to-mesenchymal transition of prostate cancer cells by suppressing degradation of EGFR through APMAP. Cancer Res. 79:3063–3075. 2019. View Article : Google Scholar : PubMed/NCBI

222 

Shapira KE, Ehrlich M and Henis YI: Cholesterol depletion enhances TGF-β Smad signaling by increasing c-Jun expression through a PKR-dependent mechanism. Mol Biol Cell. 29:2494–2507. 2018. View Article : Google Scholar : PubMed/NCBI

223 

Zheng S, Lin J, Pang Z, Zhang H, Wang Y, Ma L, Zhang H, Zhang X, Chen M, Zhang X, et al: Aberrant cholesterol metabolism and Wnt/β-catenin signaling coalesce via Frizzled5 in supporting cancer growth. Adv Sci (Weinh). 9:e22007502022. View Article : Google Scholar

224 

Ehmsen S and Ditzel HJ: Signaling pathways essential for triple-negative breast cancer stem-like cells. Stem Cells. 39:133–143. 2021. View Article : Google Scholar

225 

Wang XD, Kim C, Zhang Y, Rindhe S, Cobb MH and Yu Y: Cholesterol regulates the tumor adaptive resistance to MAPK pathway inhibition. J Proteome Res. 20:5379–5391. 2021. View Article : Google Scholar : PubMed/NCBI

226 

Aguirre-Portolés C, Feliu J, Reglero G and Ramírez de Molina A: ABCA1 overexpression worsens colorectal cancer prognosis by facilitating tumour growth and caveolin-1-dependent invasiveness, and these effects can be ameliorated using the BET inhibitor apabetalone. Mol Oncol. 12:1735–1752. 2018. View Article : Google Scholar : PubMed/NCBI

227 

Zhan J, Wang P, Li S, Song J, He H, Wang Y, Liu Z, Wang F, Bai H, Fang W, et al: HOXB13 networking with ABCG1/EZH2/Slug mediates metastasis and confers resistance to cisplatin in lung adenocarcinoma patients. Theranostics. 9:2084–2099. 2019. View Article : Google Scholar : PubMed/NCBI

228 

Bu L, Zhang Z, Chen J, Fan Y, Guo J, Su Y, Wang H, Zhang X, Wu X, Jiang Q, et al: High-fat diet promotes liver tumorigenesis via palmitoylation and activation of AKT. Gut. 73:1156–1168. 2024. View Article : Google Scholar : PubMed/NCBI

229 

Dong J, Kong L, Wang S, Xia M, Zhang Y, Wu J, Yang F, Zuo S and Wei J: Oncolytic adenovirus encoding apolipoprotein A1 suppresses metastasis of triple-negative breast cancer in mice. J Exp Clin Cancer Res. 43:1022024. View Article : Google Scholar : PubMed/NCBI

230 

Li Y, Zhou Y, Huang M, Wang Z, Liu D, Liu J, Fu X, Yang S, Shan S, Yang L, et al: DHCR7 promotes tumorigenesis via activating PI3K/AKT/mTOR signalling pathway in bladder cancer. Cell Signal. 102:1105532023. View Article : Google Scholar

231 

Wan S, He QY, Yang Y, Liu F, Zhang X, Guo X, Niu H, Wang Y, Liu YX, Ye WL, et al: SPARC stabilizes ApoE to induce cholesterol-dependent invasion and sorafenib resistance in hepatocellular carcinoma. Cancer Res. 84:1872–1888. 2024. View Article : Google Scholar : PubMed/NCBI

232 

Tashiro J, Sugiura A, Warita T, Irie N, Dwi Cahyadi D, Ishikawa T and Warita K: CYP11A1 silencing suppresses HMGCR expression via cholesterol accumulation and sensitizes CRPC cell line DU-145 to atorvastatin. J Pharmacol Sci. 153:104–112. 2023. View Article : Google Scholar : PubMed/NCBI

233 

Gómez-López S, Alhendi ASN, Przybilla MJ, Bordeu I, Whiteman ZE, Butler T, Rouhani MJ, Kalinke L, Uddin I, Otter KEJ, et al: Aberrant basal cell clonal dynamics shape early lung carcinogenesis. Science. 388:eads91452025. View Article : Google Scholar : PubMed/NCBI

234 

Lampropoulou DI, Papadimitriou M, Papadimitriou C, Filippou D, Kourlaba G, Aravantinos G and Gazouli M: The role of EMT-related lncRNAs in ovarian cancer. Int J Mol Sci. 24:100792023. View Article : Google Scholar : PubMed/NCBI

235 

Warita K, Ishikawa T, Sugiura A, Tashiro J, Shimakura H, Hosaka YZ, Ohta KI, Warita T and Oltvai ZN: Concomitant attenuation of HMGCR expression and activity enhances the growth inhibitory effect of atorvastatin on TGF-β-treated epithelial cancer cells. Sci Rep. 11:127632021. View Article : Google Scholar

236 

Xiao J, Wang S, Chen L, Ding X, Dang Y, Han M, Zheng Y, Shen H, Wu S, Wang M, et al: 25-Hydroxycholesterol regulates lysosome AMP kinase activation and metabolic reprogramming to educate immunosuppressive macrophages. Immunity. 57:1087–1104.e7. 2024. View Article : Google Scholar : PubMed/NCBI

237 

Sun Z, Xu Y, Shao B, Dang P, Hu S, Sun H, Chen C, Wang C, Liu J, Liu Y and Hu J: Exosomal circPOLQ promotes macrophage M2 polarization via activating IL-10/STAT3 axis in a colorectal cancer model. J Immunother Cancer. 12:e0084912024. View Article : Google Scholar : PubMed/NCBI

238 

Ma Z, Guo L, Pan M, Jiang C, Liu D, Gao Y, Bai J, Jiang P and Liu X: Inhibition of pseudorabies virus replication via upregulated interferon response by targeting 7-dehydrocholesterol reductase. Vet Microbiol. 290:1100002024. View Article : Google Scholar : PubMed/NCBI

239 

Ma X, Bi E, Lu Y, Su P, Huang C, Liu L, Wang Q, Yang M, Kalady MF, Qian J, et al: Cholesterol induces CD8+ T cell exhaustion in the tumor microenvironment. Cell Metab. 30:143–156.e5. 2019. View Article : Google Scholar

240 

Yang W, Bai Y, Xiong Y, Zhang J, Chen S, Zheng X, Meng X, Li L, Wang J, Xu C, et al: Potentiating the antitumour response of CD8(+) T cells by modulating cholesterol metabolism. Nature. 531:651–655. 2016. View Article : Google Scholar : PubMed/NCBI

241 

Sugi T, Katoh Y, Ikeda T, Seta D, Iwata T, Nishio H, Sugawara M, Kato D, Katoh K, Kawana K, et al: SCD1 inhibition enhances the effector functions of CD8+ T cells via ACAT1-dependent reduction of esterified cholesterol. Cancer Sci. 115:48–58. 2024. View Article : Google Scholar

242 

Xu H, Exner BG, Cramer DE, Tanner MK, Mueller YM and Ildstad ST: CD8(+), alphabeta-TCR(+), and gammadelta-TCR(+) cells in the recipient hematopoietic environment mediate resistance to engraftment of allogeneic donor bone marrow. J Immunol. 168:1636–1643. 2002. View Article : Google Scholar : PubMed/NCBI

243 

Lü HZ, Zhu AY, Chen Y, Tang J and Li BQ: Formation and aggregation of lipid rafts in γδ T cells following stimulation with Mycobacterium tuberculosis antigens. Tohoku J Exp Med. 223:193–198. 2011. View Article : Google Scholar

244 

Júnior RFA, Lira GA, Schomann T, Cavalcante RS, Vilar NF, de Paula RCM, Gomes RF, Chung CK, Jorquera-Cordero C, Vepris O, et al: Retinoic acid-loaded PLGA nanocarriers targeting cell cholesterol potentialize the antitumour effect of PD-L1 antibody by preventing epithelial-mesenchymal transition mediated by M2-TAM in colorectal cancer. Transl Oncol. 31:1016472023. View Article : Google Scholar : PubMed/NCBI

245 

Wang Q, Cao Y, Shen L, Xiao T, Cao R, Wei S, Tang M, Du L, Wu H, Wu B, et al: Regulation of PD-L1 through direct binding of cholesterol to CRAC motifs. Sci Adv. 8:eabq47222022. View Article : Google Scholar : PubMed/NCBI

246 

Sun S, Ma J, Zuo T, Shi J, Sun L, Meng C, Shu W, Yang Z, Yao H and Zhang Z: Inhibition of PCSK9: A promising enhancer for Anti-PD-1/PD-L1 immunotherapy. Research (Wash D C). 7:04882024.PubMed/NCBI

247 

Ma X, Bi E, Huang C, Lu Y, Xue G, Guo X, Wang A, Yang M, Qian J, Dong C and Yi Q: Cholesterol negatively regulates IL-9-producing CD8+ T cell differentiation and antitumor activity. J Exp Med. 215:1555–1569. 2018. View Article : Google Scholar : PubMed/NCBI

248 

Bai Y, Li T, Wang Q, You W, Yang H, Xu X, Li Z, Zhang Y, Yan C, Yang L, et al: Shaping immune landscape of colorectal cancer by cholesterol metabolites. EMBO Mol Med. 16:334–360. 2024. View Article : Google Scholar : PubMed/NCBI

249 

Baek AE, Yu YA, He S, Wardell SE, Chang CY, Kwon S, Pillai RV, McDowell HB, Thompson JW, Dubois LG, et al: The cholesterol metabolite 27 hydroxycholesterol facilitates breast cancer metastasis through its actions on immune cells. Nat Commun. 8:8642017. View Article : Google Scholar : PubMed/NCBI

250 

Bruckner M, Dickel D, Singer E and Legler DF: Converse regulation of CCR7-driven human dendritic cell migration by prostaglandin E2 and liver X receptor activation. Eur J Immunol. 42:2949–2958. 2012. View Article : Google Scholar : PubMed/NCBI

251 

Kitahara CM, Berrington de González A, Freedman ND, Huxley R, Mok Y, Jee SH and Samet JM: Total cholesterol and cancer risk in a large prospective study in Korea. J Clin Oncol. 29:1592–1598. 2011. View Article : Google Scholar : PubMed/NCBI

252 

Mondul AM, Clipp SL, Helzlsouer KJ and Platz EA: Association between plasma total cholesterol concentration and incident prostate cancer in the CLUE II cohort. Cancer Causes Control. 21:61–68. 2010.

253 

Nielsen SF, Nordestgaard BG and Bojesen SE: Statin use and reduced cancer-related mortality. N Engl J Med. 367:1792–1802. 2012. View Article : Google Scholar : PubMed/NCBI

254 

Lin Q, Liu W, Xu S and Sun L: Associations of preoperative serum high-density lipoprotein cholesterol and low-density lipoprotein cholesterol levels with the prognosis of ovarian cancer. Arch Gynecol Obstet. 305:683–691. 2022. View Article : Google Scholar

255 

Penet MF, Krishnamachary B, Wildes FB, Mironchik Y, Hung CF, Wu TC and Bhujwalla ZM: Ascites volumes and the ovarian cancer microenvironment. Front Oncol. 8:5952018. View Article : Google Scholar

256 

Lu L, Katsaros D, Wiley A, Rigault de la Longrais IA, Puopolo M and Yu H: Expression of MDR1 in epithelial ovarian cancer and its association with disease progression. Oncol Res. 16:395–403. 2007. View Article : Google Scholar : PubMed/NCBI

257 

Mentella MC, Scaldaferri F, Ricci C, Gasbarrini A and Miggiano GAD: Cancer and mediterranean diet: A review. Nutrients. 11:20592019. View Article : Google Scholar : PubMed/NCBI

258 

Mittelman SD: The role of diet in cancer prevention and chemotherapy efficacy. Annu Rev Nutr. 40:273–297. 2020. View Article : Google Scholar : PubMed/NCBI

259 

Nencioni A, Caffa I, Cortellino S and Longo VD: Fasting and cancer: molecular mechanisms and clinical application. Nat Rev Cancer. 18:707–719. 2018. View Article : Google Scholar : PubMed/NCBI

260 

Li C, Yang L, Zhang D and Jiang W: Systematic review and meta-analysis suggest that dietary cholesterol intake increases risk of breast cancer. Nutr Res. 36:627–635. 2016. View Article : Google Scholar : PubMed/NCBI

261 

Bandera EV, Kushi LH, Moore DF, Gifkins DM and McCullough ML: Dietary lipids and endometrial cancer: The current epidemiologic evidence. Cancer Causes Control. 18:687–703. 2007.PubMed/NCBI

262 

Yang H, Wang F, Hallemeier CL, Lerut T and Fu J: Oesophageal cancer. Lancet. 404:1991–2005. 2024. View Article : Google Scholar : PubMed/NCBI

263 

Jin Y, Yang T, Li D and Ding W: Effect of dietary cholesterol intake on the risk of esophageal cancer: A meta-analysis. J Int Med Res. 47:4059–4068. 2019. View Article : Google Scholar : PubMed/NCBI

264 

Wang QL, Khil J, Hong S, Lee DH, Ha KH, Keum N, Kim HC and Giovannucci EL: Temporal association of total serum cholesterol and pancreatic cancer incidence. Nutrients. 14:49382022. View Article : Google Scholar : PubMed/NCBI

265 

Di Maso M, Augustin LSA, Jenkins DJA, Crispo A, Toffolutti F, Negri E, La Vecchia C, Ferraroni M and Polesel J: Adherence to a cholesterol-lowering diet and the risk of pancreatic cancer: A case-control study. Nutrients. 16:25082024. View Article : Google Scholar : PubMed/NCBI

266 

Hu J, La Vecchia C, de Groh M, Negri E, Morrison H and Mery L; Canadian Cancer Registries Epidemiology Research Group: Dietary cholesterol intake and cancer. Ann Oncol. 23:491–500. 2012. View Article : Google Scholar

267 

Rice MS, Poole EM, Willett WC and Tworoger SS: Adult dietary fat intake and ovarian cancer risk. Int J Cancer. 146:2756–2772. 2020. View Article : Google Scholar :

268 

Chlebowski RT, Anderson GL, Manson JE, Prentice RL, Aragaki AK, Snetselaar L, Beresford SAA, Kuller LH, Johnson K, Lane D, et al: Low-fat dietary pattern and cancer mortality in the women's health initiative (WHI) randomized controlled trial. JNCI Cancer Spectr. 2:pky0652019. View Article : Google Scholar : PubMed/NCBI

269 

Lin X, Liu L, Fu Y, Gao J, He Y, Wu Y and Lian X: Dietary cholesterol intake and risk of lung cancer: A meta-analysis. Nutrients. 10:1852018. View Article : Google Scholar : PubMed/NCBI

270 

Wu Y, Zheng W, Sellers TA, Kushi LH, Bostick RM and Potter JD: Dietary cholesterol, fat, and lung cancer incidence among older women: The Iowa women's health study (United States). Cancer Causes Control. 5:395–400. 1994.PubMed/NCBI

271 

Habis M, Wroblewski K, Bradaric M, Ismail N, Yamada SD, Litchfield L, Lengyel E and Romero IL: Statin therapy is associated with improved survival in patients with non-serous-papillary epithelial ovarian cancer: A retrospective cohort analysis. PLoS One. 9:e1045212014. View Article : Google Scholar : PubMed/NCBI

272 

Iyengar NM, Gucalp A, Dannenberg AJ and Hudis CA: Obesity and cancer mechanisms: Tumor microenvironment and inflammation. J Clin Oncol. 34:4270–4276. 2016. View Article : Google Scholar : PubMed/NCBI

273 

Yan P and Zhao D: Association between serum total cholesterol and the development of gastric cancer: A two-way two-sample Mendelian randomization study. Medicine (Baltimore). 103:e389002024. View Article : Google Scholar : PubMed/NCBI

274 

Gruenbacher G and Thurnher M: Mevalonate metabolism in immuno-oncology. Front Immunol. 8:17142017. View Article : Google Scholar : PubMed/NCBI

275 

Madan B, Virshup DM, Nes WD and Leaver DJ: Unearthing the Janus-face cholesterogenesis pathways in cancer. Biochem Pharmacol. 196:1146112022. View Article : Google Scholar

276 

Murto MO, Simolin N, Arponen O, Siltari A, Artama M, Visvanathan K, Jukkola A and Murtola TJ: Statin use, cholesterol level, and mortality among females with breast cancer. JAMA Netw Open. 6:e23438612023. View Article : Google Scholar : PubMed/NCBI

277 

Kansal V, Burnham AJ, Kinney BLC, Saba NF, Paulos C, Lesinski GB, Buchwald ZS and Schmitt NC: Statin drugs enhance responses to immune checkpoint blockade in head and neck cancer models. J Immunother Cancer. 11:e0059402023. View Article : Google Scholar : PubMed/NCBI

278 

Zhang Y, Wu K, Chan AT, Meyerhardt JA and Giovannucci EL: Long-term statin use, total cholesterol level, and risk of colorectal cancer: A prospective cohort study. Am J Gastroenterol. 117:158–166. 2022. View Article : Google Scholar :

279 

Morote J, Celma A, Planas J, Placer J, de Torres I, Olivan M, Carles J, Reventós J and Doll A: Role of serum cholesterol and statin use in the risk of prostate cancer detection and tumor aggressiveness. Int J Mol Sci. 15:13615–13623. 2014. View Article : Google Scholar : PubMed/NCBI

280 

Hillis AL, Martin TD, Manchester HE, Högström J, Zhang N, Lecky E, Kozlova N, Lee J, Persky NS, Root DE, et al: Targeting cholesterol biosynthesis with statins synergizes with AKT inhibitors in triple-negative breast cancer. Cancer Res. 84:3250–3266. 2024. View Article : Google Scholar : PubMed/NCBI

281 

Trotta F, Avena P, Chimento A, Rago V, De Luca A, Sculco S, Nocito MC, Malivindi R, Fallo F, Pezzani R, et al: Statins reduce intratumor cholesterol affecting adrenocortical cancer growth. Mol Cancer Ther. 19:1909–1921. 2020. View Article : Google Scholar : PubMed/NCBI

282 

Qiu W, Su W, Xu J, Liang M, Ma X, Xue P, Kang Y, Sun ZJ and Xu Z: Immunomodulatory-photodynamic nanostimulators for invoking pyroptosis to augment tumor immunotherapy. Adv Healthc Mater. 11:e22012332022. View Article : Google Scholar : PubMed/NCBI

283 

Nguyen TTT, Ishida CT, Shang E, Shu C, Torrini C, Zhang Y, Bianchetti E, Sanchez-Quintero MJ, Kleiner G, Quinzii CM, et al: Activation of LXRβ inhibits tumor respiration and is synthetically lethal with Bcl-xL inhibition. EMBO Mol Med. 11:e107692019. View Article : Google Scholar

284 

Nguyen TTT, Ishida CT, Shang E, Shu C, Bianchetti E, Karpel-Massler G and Siegelin MD: Activation of LXR receptors and inhibition of TRAP1 causes synthetic lethality in solid tumors. Cancers (Basel). 11:7882019. View Article : Google Scholar : PubMed/NCBI

285 

Cai J, Ye Z, Hu Y, Ye L, Gao L, Wang Y, Sun Q, Tong S, Zhang S, Wu L, et al: Fatostatin induces ferroptosis through inhibition of the AKT/mTORC1/GPX4 signaling pathway in glioblastoma. Cell Death Dis. 14:2112023. View Article : Google Scholar : PubMed/NCBI

286 

Gao S, Shi Z, Li X, Li W, Wang Y, Liu Z and Jiang J: Fatostatin suppresses growth and enhances apoptosis by blocking SREBP-regulated metabolic pathways in endometrial carcinoma. Oncol Rep. 39:1919–1929. 2018.PubMed/NCBI

287 

Lee HJ, Li J, Vickman RE, Li J, Liu R, Durkes AC, Elzey BD, Yue S, Liu X, Ratliff TL and Cheng JX: Cholesterol esterification inhibition suppresses prostate cancer metastasis by impairing the Wnt/β-catenin pathway. Mol Cancer Res. 16:974–985. 2018. View Article : Google Scholar : PubMed/NCBI

288 

Zhang Y, Li L, Chu F, Wu H, Xiao X, Ye J and Li K: Itraconazole inhibits tumor growth via CEBPB-mediated glycolysis in colorectal cancer. Cancer Sci. 115:1154–1169. 2024. View Article : Google Scholar : PubMed/NCBI

289 

Aftab BT, Dobromilskaya I, Liu JO and Rudin CM: Itraconazole inhibits angiogenesis and tumor growth in non-small cell lung cancer. Cancer Res. 71:6764–6772. 2011. View Article : Google Scholar : PubMed/NCBI

290 

Wang S, Link F, Han M, Chaudhary R, Asimakopoulos A, Liebe R, Yao Y, Hammad S, Dropmann A, Krizanac M, et al: The interplay of TGF-β1 and cholesterol orchestrating hepatocyte cell fate, EMT, and signals for HSC activation. Cell Mol Gastroenterol Hepatol. 17:567–587. 2024. View Article : Google Scholar

291 

Wang Y, Zhou X, Lei Y, Chu Y, Yu X, Tong Q, Zhu T, Yu H, Fang S, Li G, et al: NNMT contributes to high metastasis of triple negative breast cancer by enhancing PP2A/MEK/ERK/c-Jun/ABCA1 pathway mediated membrane fluidity. Cancer Lett. 547:2158842022. View Article : Google Scholar : PubMed/NCBI

292 

Brindisi M, Frattaruolo L, Fiorillo M, Dolce V, Sotgia F, Lisanti MP and Cappello AR: New insights into cholesterol-mediated ERRα activation in breast cancer progression and pro-tumoral microenvironment orchestration. FEBS J. 290:1481–1501. 2023. View Article : Google Scholar

293 

Jin H, He Y, Zhao P, Hu Y, Tao J, Chen J and Huang Y: Targeting lipid metabolism to overcome EMT-associated drug resistance via integrin β3/FAK pathway and tumor-associated macrophage repolarization using legumain-activatable delivery. Theranostics. 9:265–278. 2019. View Article : Google Scholar :

294 

Gao CQ, Chu ZZ, Zhang D, Xiao Y, Zhou XY, Wu JR, Yuan H, Jiang YC, Chen D, Zhang JC, et al: Serine/threonine kinase TBK1 promotes cholangiocarcinoma progression via direct regulation of β-catenin. Oncogene. 42:1492–1507. 2023. View Article : Google Scholar : PubMed/NCBI

295 

Wen Q, Xie X, Chen C, Wen B, Liu Y, Zhou J, Lin X, Jin H and Shi K: Lipid reprogramming induced by the NNMT-ABCA1 axis enhanced membrane fluidity to promote endometrial cancer progression. Aging (Albany NY). 15:11860–11874. 2023. View Article : Google Scholar : PubMed/NCBI

296 

Dong G, Huang X, Jiang S, Ni L, Ma L, Zhu C and Chen S: SCAP mediated GDF15-induced invasion and EMT of esophageal cancer. Front Oncol. 10:5647852020. View Article : Google Scholar : PubMed/NCBI

297 

Chen W, Zhang Q, Dai X, Chen X, Zhang C, Bai R, Chen Y, Zhang K, Duan X, Qiao Y, et al: PGC-1α promotes colorectal carcinoma metastasis through regulating ABCA1 transcription. Oncogene. 42:2456–2470. 2023. View Article : Google Scholar : PubMed/NCBI

298 

Qin WH, Yang ZS, Li M, Chen Y, Zhao XF, Qin YY, Song JQ, Wang BB, Yuan B, Cui XL, et al: High serum levels of cholesterol increase antitumor functions of nature killer Cells and reduce growth of liver tumors in mice. Gastroenterology. 158:1713–1727. 2020. View Article : Google Scholar : PubMed/NCBI

299 

Esposito G, Augustin LSA, Jenkins DJA, Ferraroni M, Parazzini F, Crispo A, Dal Maso L, Negri E, La Vecchia C, Polesel J and Di Maso M: Adherence to a cholesterol-lowering diet and the risk of female hormone-related cancers: An analysis from a case-control study network. BJOG. 132:1791–1801. 2025. View Article : Google Scholar : PubMed/NCBI

300 

Furberg AS, Veierød MB, Wilsgaard T, Bernstein L and Thune I: Serum high-density lipoprotein cholesterol, metabolic profile, and breast cancer risk. J Natl Cancer Inst. 96:1152–1160. 2004. View Article : Google Scholar : PubMed/NCBI

301 

Di Maso M, Dal Maso L, Augustin LSA, Puppo A, Falcini F, Stocco C, Mattioli V, Serraino D and Polesel J: Adherence to the mediterranean diet and mortality after breast cancer. Nutrients. 12:36492020. View Article : Google Scholar : PubMed/NCBI

302 

Maddineni G, Xie JJ, Brahmbhatt B and Mutha P: Diet and carcinogenesis of gastric cancer. Curr Opin Gastroenterol. 38:588–591. 2022. View Article : Google Scholar : PubMed/NCBI

303 

Bidoli E, La Vecchia C, Montella M, Maso LD, Conti E, Negri E, Scarabelli C, Carbone A, Decarli A and Franceschi S: Nutrient intake and ovarian cancer: An Italian case-control study. Cancer Causes Control. 13:255–261. 2002.PubMed/NCBI

304 

Quan L, Liu Y, Cui W, Wang X, Zhang W, Wang Z, Guo C, Lu C, Hu F and Chen X: The associations between serum high-density lipoprotein cholesterol levels and malignant behavior in pancreatic neuroendocrine neoplasms. Lipids Health Dis. 21:582022. View Article : Google Scholar : PubMed/NCBI

305 

de Martino M, Leitner CV, Seemann C, Hofbauer SL, Lucca I, Haitel A, Shariat SF and Klatte T: Preoperative serum cholesterol is an independent prognostic factor for patients with renal cell carcinoma (RCC). BJU Int. 115:397–404. 2015. View Article : Google Scholar

306 

Li L, Yu Z, Ren J and Niu T: Low cholesterol levels are associated with increasing risk of plasma cell neoplasm: A UK biobank cohort study. Cancer Med. 12:20964–20975. 2023. View Article : Google Scholar : PubMed/NCBI

307 

Yang Z, Tang H, Lu S, Sun X and Rao B: Relationship between serum lipid level and colorectal cancer: A systemic review and meta-analysis. BMJ Open. 12:e0523732022. View Article : Google Scholar : PubMed/NCBI

308 

Yuan F, Wen W, Jia G, Long J, Shu XO and Zheng W: Serum lipid profiles and cholesterol-lowering medication use in relation to subsequent risk of colorectal cancer in the UK biobank cohort. Cancer Epidemiol Biomarkers Prev. 32:524–530. 2023. View Article : Google Scholar : PubMed/NCBI

309 

Karayama M, Inui N, Inoue Y, Yoshimura K, Mori K, Hozumi H, Suzuki Y, Furuhashi K, Fujisawa T, Enomoto N, et al: Increased serum cholesterol and long-chain fatty acid levels are associated with the efficacy of nivolumab in patients with non-small cell lung cancer. Cancer Immunol Immunother. 71:203–217. 2022. View Article : Google Scholar :

310 

Narii N, Zha L, Komatsu M, Kitamura T, Sobue T and Ogawa T: Cholesterol and breast cancer risk: A cohort study using health insurance claims and health checkup databases. Breast Cancer Res Treat. 199:315–322. 2023. View Article : Google Scholar : PubMed/NCBI

311 

Yang Z, Zhang D, Sima X, Fu Y, Zeng H, Hu Z, Hou J, Pan Y, Zhang Y, Zhou Z, et al: Levels of pretreatment serum lipids predict responses to PD-1 inhibitor treatment in advanced intrahepatic cholangiocarcinoma. Int Immunopharmacol. 115:1096872023. View Article : Google Scholar : PubMed/NCBI

312 

Huang F, Li S, Wang X, Wang C, Pan X, Chen X, Zhang W and Hong J: Serum lipids concentration on prognosis of high-grade glioma. Cancer Causes Control. 34:801–811. 2023.PubMed/NCBI

313 

Min J, Wu Y, Huang S, Li Y, Lv X, Tang R, Zhao H and Wang J: Serum cholesterol level as a predictive biomarker for prognosis of neuroblastoma. BMC Pediatr. 24:2052024. View Article : Google Scholar : PubMed/NCBI

314 

Zhang C, Li Y, Wang Y, Hu S, Liu Y, Liang X, Chen ZJ, Zhang Y and Zhao H: Genetic associations of metabolic factors and therapeutic drug targets with polycystic ovary syndrome. J Adv Res. 75:581–590. 2025. View Article : Google Scholar

315 

Qian L, Qian B, Xu J, Yang J, Wu G, Zhao Y, Liu Q, Yuan Z, Fan Y and Li H: Clinical relevance of serum lipids in the carcinogenesis of oral squamous cell carcinoma. BMC Oral Health. 23:2002023. View Article : Google Scholar : PubMed/NCBI

316 

Leeper H, Viall A, Ruaux C and Bracha S: Preliminary evaluation of serum total cholesterol concentrations in dogs with osteosarcoma. J Small Anim Pract. 58:562–569. 2017. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
He Z, Zhang L, Gong S, Yang X and Xu G: Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review). Int J Mol Med 56: 226, 2025.
APA
He, Z., Zhang, L., Gong, S., Yang, X., & Xu, G. (2025). Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review). International Journal of Molecular Medicine, 56, 226. https://doi.org/10.3892/ijmm.2025.5667
MLA
He, Z., Zhang, L., Gong, S., Yang, X., Xu, G."Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review)". International Journal of Molecular Medicine 56.6 (2025): 226.
Chicago
He, Z., Zhang, L., Gong, S., Yang, X., Xu, G."Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 226. https://doi.org/10.3892/ijmm.2025.5667
Copy and paste a formatted citation
x
Spandidos Publications style
He Z, Zhang L, Gong S, Yang X and Xu G: Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review). Int J Mol Med 56: 226, 2025.
APA
He, Z., Zhang, L., Gong, S., Yang, X., & Xu, G. (2025). Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review). International Journal of Molecular Medicine, 56, 226. https://doi.org/10.3892/ijmm.2025.5667
MLA
He, Z., Zhang, L., Gong, S., Yang, X., Xu, G."Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review)". International Journal of Molecular Medicine 56.6 (2025): 226.
Chicago
He, Z., Zhang, L., Gong, S., Yang, X., Xu, G."Cholesterol metabolism and cancer: Molecular mechanisms, immune regulation and an epidemiological perspective (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 226. https://doi.org/10.3892/ijmm.2025.5667
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team