Open Access

Therapeutic efficacy of ferroptosis in the treatment of colorectal cancer (Review)

  • Authors:
    • Zhao Guo
    • Haoyan Zhuang
    • Xuewen Shi
  • View Affiliations

  • Published online on: September 26, 2024     https://doi.org/10.3892/ol.2024.14697
  • Article Number: 563
  • Copyright: © Guo et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is the third most common malignancy worldwide, and the second leading cause of cancer‑associated mortality. The incidence and mortality rates of CRC remain high, posing a significant threat to humans and overall quality of life. Current therapeutic strategies, such as surgery and chemotherapy, are limited due to disease recurrence, chemotherapeutic drug resistance and toxicity. Thus, research is focused on the development of novel treatment approaches. In 2012, ferroptosis was identified as a form of regulated cell death that is iron‑dependent and driven by lipid peroxidation. Notably, therapies targeting ferroptosis exhibit potential in the treatment of disease; however, their role in CRC treatment remains controversial. The present study aimed to systematically review the mechanisms and signaling pathways of ferroptosis in CRC, and the specific role within the tumor microenvironment. Moreover, the present study aimed to review the role of ferroptosis in drug resistance, offering novel perspectives for the diagnosis and treatment of CRC.

Introduction

Colorectal cancer (CRC) is a prevalent malignancy of the gastrointestinal tract, with high rates of morbidity and mortality. In 2018, the GLOBOCAN database published an analysis of the incidence and mortality rates for 36 cancers across 185 countries, and the results revealed that CRC is the third most common cancer and the second leading cause of cancer-associated mortalities worldwide (1,2). The etiology of CRC is associated with various factors, including age, sex, inflammatory bowel disease, lifestyle and environmental factors (35). Current treatment options for CRC include surgery, chemotherapy, radiotherapy, immunotherapy and targeted biological therapies (6). However, the absence of highly specific biomarkers for the diagnosis of CRC and the development of chemoresistance in advanced stages of treatment may impact the quality of life of patients with late-stage disease (7). Thus, research is focused on the development of novel effective targeted therapies for CRC. Notably, ferroptosis may exhibit potential as an emerging strategy for CRC treatment.

Regulated cell death, including apoptosis, pyroptosis, necroptosis, ferroptosis, autophagy-dependent cell death and neoplastic cell death, is a crucial mechanism for maintaining the internal homeostasis of the human body and preserving tissue function and morphology (8,9). The primary method for distinguishing between different forms of cell death is based on the associated morphological characteristics (Table I).

Table I.

Characteristics of ferroptosis, apoptosis, pyroptosis, necroptosis and autophagy.

Table I.

Characteristics of ferroptosis, apoptosis, pyroptosis, necroptosis and autophagy.

Regulated form of cell deathMorphological characteristicBiochemical characteristic(Refs.)
FerroptosisDecreased mitochondrial volume, reduced number of cristae, increased mitochondrial membrane density and elevated rates of mitochondrial membrane ruptureIron accumulation and lipid peroxidation(8)
ApoptosisCell shrinkage, nuclear condensation and preservation of cytoplasmic membrane integrityDNA fragmentation and macromolecular synthesis(13)
PyroptosisCell swelling and expansion, formation of numerous vesicles in necrotic cells, chromatin degradation, and the release of pro-inflammatory componentsInflammatory vesicle formation(1416)
NecroptosisPlasma membrane permeabilization, cellular swelling and loss of organelle integrityDecreased levels of adenosine triphosphate(17,18)
AutophagyFormation of autophagic lysosomes with double-membrane structuresIncreased lysosomal activity(19)

Ferroptosis, initially described by Dixon in 2012, is a novel form of regulated cell death (9). It is distinct from other forms of regulated cell death in terms of morphology, biochemical features and gene expression (10). Notably, ferroptosis is characterized by unique morphological features, including mitochondrial shrinkage, increased mitochondrial membrane density, reduced or absent mitochondrial cristae, rupture of the outer mitochondrial membrane, and preservation of nuclear integrity (11,12). The main morphological characteristics of apoptosis include cell shrinkage, nuclear condensation and the maintenance of plasma membrane integrity, while its biochemical features are primarily DNA fragmentation and macromolecule synthesis (13). The key morphological traits of pyroptosis involve cell swelling, the formation of numerous bubbles on the pyroptotic cells, and chromatin degradation, leading to the formation of inflammasomes (1416). The primary morphological characteristics of necroptosis include increased cell membrane permeability, cell swelling and loss of organelle integrity, while its biochemical hallmark is a decrease in adenosine triphosphate levels (17,18). Autophagy is characterized by the formation of double-membraned autophagosomes, and its biochemical feature is an increase in lysosomal activity (19).

Ferroptosis has been investigated in a variety of diseases, including Parkinson's disease (20,21), Alzheimer's disease (22,23), liver injury (24,25), brain injury (26,27), spinal cord injury (2830), kidney injury (31,32), cardiovascular disease (3335) and gynecological disorders (36,37) (Fig. 1). At present, research is focused on the role of ferroptosis in the treatment of cancer. Yan et al (38) demonstrated that ferroptosis plays a key role in the development of CRC, breast cancer, ovarian cancer, renal cancer, lymphoma and melanoma (39). However, results of a previous study revealed that ferroptosis may also play a role in CRC progression and metastasis, through the activation of different signaling pathways (40). Thus, previous studies highlight that ferroptosis may exert contrasting effects in CRC, demonstrating the requirement for further investigations. In addition, the development of novel treatment approaches is required to mitigate the adverse effects of ferroptosis on CRC. This article systematically reviews the mechanisms and signaling pathways of ferroptosis in CRC, the role of ferroptosis within the tumor microenvironment, and the progress in research on ferroptosis in reducing drug resistance. It aims to provide new insights for the diagnosis and treatment of CRC.

Mechanism of action of ferroptosis

Ferroptosis is primarily characterized by the excessive accumulation of iron and lipid reactive oxygen species (10). Previous studies have identified mechanisms that may be involved with ferroptosis, including amino acid, lipid and iron metabolism (41). Key mechanisms involved in ferroptosis are displayed in Fig. 2.

Induction of ferroptosis through inhibition of the cystine/glutamate antiporter system (System Xc-)

System Xc- is a critical component of the cellular antioxidant system that is widely distributed in the phospholipid bilayer. System Xc- is a heterodimer composed of two subunits: Namely, solute carrier family 7 member 11 (SLC7A11) and SLC3A2. System Xc- facilitates the 1:1 exchange of cystine and glutamate across the cell membrane (9). Once inside the cell, cystine is utilized to synthesize glutathione, which, in the presence of glutathione peroxidase, reduces the production of lipid reactive oxygen species. Therefore, inhibition of System Xc- leads to reduced glutathione peroxidase activity, decreased cellular antioxidant capacity, accumulation of lipid reactive oxygen species and ferroptosis (9). A well-established oncogene, P53, may induce ferroptosis via downregulation of SLC7A11 expression, thereby inhibiting System Xc- activity (42,43). Moreover, sorafenib (44), sulfasalazine (45) and erastin (46) induce ferroptosis via inhibition of System Xc-.

Induction of ferroptosis through inhibition of glutathione peroxidase 4 (GPX4)

GPX4 plays a critical role in the cellular peroxidase system, inhibiting the formation of peroxides and thus acting as a key regulator of ferroptosis. GPX4 catalyzes the conversion of glutathione to oxidized glutathione and reduces cytotoxic lipid peroxides to their corresponding alcohols (12). Yang et al (47) demonstrated that reduced GPX4 expression may lead to the induction of ferroptosis, whereas increased GPX4 expression may inhibit ferroptosis. RAS-selective lethal 3 (RSL3) is a widely established inhibitor of GPX4 and an inducer of ferroptosis. RSL3 directly targets GPX4, thereby inhibiting its activity and leading to reductions in cellular antioxidant capacity, the accumulation of lipid reactive oxygen species and the induction of ferroptosis (48). Costa et al (49) identified ML162 and ML210 as GPX4 inhibitors that induce ferroptosis, through a mechanism that is comparable with that of RSL3.

Lipid metabolism

Lipid peroxidation is critical in the induction of ferroptosis. Lipids are essential components of cell membranes, and lipid peroxidation disrupts these membranes, thereby triggering ferroptosis (50). Polyunsaturated fatty acids (PUFAs), such as arachidonic acid and adrenic acid, are long-chain fatty acids with multiple double bonds that are highly susceptible to lipid peroxidation. Phosphatidylethanolamine containing arachidonic and adrenic acids is a key phospholipid in the induction of cellular ferroptosis. There are two lipid-metabolizing enzymes associated with ferroptosis; namely, acyl-CoA synthetase long-chain family member 4 (ACSL4) and lysophosphatidylcholine acyltransferase 3, which are involved in the biosynthesis of phosphatidylethanolamine. These enzymes activate PUFAs and impact their transmembrane properties, leading to ferroptosis (51). In addition, cytochrome P450 oxidoreductase-mediated lipid peroxidation may play a key role in the induction of ferroptosis (52). Cytochrome P450 oxidoreductase transfers electrons from reduced nicotinamide adenine dinucleotide phosphate to oxygen, producing hydrogen peroxide. Hydrogen peroxide subsequently reacts with iron to generate reactive hydroxyl radicals, which peroxidize the polyunsaturated fatty acid chains of membrane phospholipids. This process disrupts the integrity of cellular membranes during iron accumulation, ultimately leading to ferroptosis (53,54).

Iron metabolism

Iron accumulation is closely associated with ferroptosis, and primarily involves iron absorption and reduction processes (55). Iron is a critical raw material for the production of hemoglobin and myoglobin. Moreover, iron plays a vital role in various cellular metabolic processes, such as oxygen storage and transport, DNA and RNA synthesis, cellular differentiation, and enzymatic reactions (56). Healthy individuals exhibit iron levels of 3–5 g, with >50% circulating in red blood cells as hemoglobin or stored as ferritin. In addition, small amounts of iron bind to transferrin in the plasma (57,58).

Intracellular iron homeostasis is maintained through a balance of iron absorption, utilization, storage and excretion. Fe3+ is absorbed in the duodenum and jejunum, and transported into cells via transferrin receptor 1, where it is reduced to Fe2+ by metal reductase in the endoplasmic reticulum. For biological activation, a small amount of Fe2+ is released into a labile iron pool in the cytoplasm via the divalent metal transporter 1, while the remainder is recycled or stored as ferritin (59,60). The results of a previous study revealed that cancer cells require higher levels of iron than healthy cells, and are more susceptible to iron depletion, also known as iron addiction (61). Notably, in the presence of a large number of cancer cells, ferritin is degraded through autophagy. This process is carried out via autophagy-related proteins 5 and 7, and the nuclear receptor coactivator 4 (NCOA4) signaling pathways. NCOA4 binds to lysosomes, degrades ferritin and releases Fe2+, leading to an abnormal increase in Fe2+ levels (6264). Excessive Fe2+ accumulation triggers the Fenton reaction (Fig. 3), initiating ferroptosis and further contributing to the accumulation of reactive oxygen species (65).

Ferroptosis in CRC

At present, research is focused on the role of drug- (Table II), gene-, protein- (Table III), and RNA-induced ferroptosis (Table IV). Each of these mechanisms play distinct roles in CRC.

Table II.

Drug-induced ferroptosis may lead to inhibition of CRC development and progression.

Table II.

Drug-induced ferroptosis may lead to inhibition of CRC development and progression.

Chemotherapeutic agentMechanism(Refs.)
CetuximabEnhanced RSL3-induced ferroptosis through inhibition of the Nrf2/HO-1 signaling pathway(66)
ApatinibPromotion of ferroptosis through the ELOVL6/ACSL4 signaling pathway(67)
IrrutinibEnhanced sensitivity to ferroptosis in CRC cells via the BTK/nRF2 signaling pathway(68)
PropofolDownregulation of STAT3 expression(69)
AspirinEnhanced RSL3-induced ferroptosis through inhibition of the mTOR/SREBP-1/SCD1 signaling pathway(70)
EmodinInduction of ferroptosis in CRC cells via NCOA4-mediated ferritin autophagy and the NF-κb pathway(71)
Ginsenoside Rh3Induction of ferroptosis in CRC cells via the Stat3/p53/NRF2 axis(72)
CurcuminPromotion of ferroptosis through regulation of the oncogene, P53, and the SLC7A11/glutathione/GPX4 axis(73)
Curcumin in combination with Andrographis paniculataPromotion of ferroptosis through downregulation of GPX4 and iron regulatory protein 1 expression levels(74)
EsculinInduction of endoplasmic reticulum stress via apoptosis and ferroptosis, through regulation of eIF2α/CHOP and Nrf2/HO-1 via the PERK signaling pathway(75)
BaicaleinPromotion of ferroptosis through inhibition of the JAK2/STAT3/GPX4 signaling pathway(76)

[i] RSL3, RAS-selective lethal 3; Nrf2, nuclear factor erythroid 2-related factor 2; ELOVL6, elongation of very long chain fatty acids protein 6; ACSL4, Acyl-CoA synthetase long-chain family member 4; STAT3, signal transducer and activator of transcription 3; mTOR, mechanistic target of rapamycin; SREBP-1, sterol regulatory element-binding protein 1; SCD1, stearoyl-CoA desaturase 1; NCOA4, Nuclear receptor coactivator 4; SLC7A11, solute carrier family 7 member 11; GPX4, glutathione peroxidase 4; PERK, protein kinase R-like endoplasmic reticulum kinase; JAK2, Janus kinase 2; CRC, colorectal cancer.

Table III.

Gene- and protein-induced ferroptosis in CRC.

Table III.

Gene- and protein-induced ferroptosis in CRC.

A, Inhibition of CRC development and progression

Gene or proteinMechanism(Refs.)
Kruppel-like factor 2Induction of ferroptosis through inhibition of the PI3K/AKT signaling pathway(77)
BromelainInhibition of proliferation and induction of ferroptosis in KRAS-mutant CRC cells through ACSL4(78)
Tagitinin CInduction of ferroptosis through the PERK/Nrf2/HO-1 signaling pathway(79)
TIGARIncreased sensitivity of CRC cells to ferroptosis via the ROS/AMPK/SCD1 signaling pathway(80,81)

B, Promotion of colorectal carcinogenesis and progression

Gene or protein Mechanism(Refs.)

Lipid transport protein 2Induction of ferroptosis through increased expression of glutathione peroxidase 4 and the cystine/glutamate antiporter component, xCT(82)
FOXA2Inhibition of ferroptosis through inhibition of the Nrf2/GPX4 signaling pathway(83)
Uridine cytidine kinase-like 1Inhibition of ferroptosis through the UCKL1/Nrf2/SLC7A11 axis(84)

[i] PI3K, phosphoinositide 3-kinase; KRAS-mutant, Kirsten rat sarcoma viral oncogene homolog; ACSL4, acyl-CoA synthetase long-chain family member 4; ROS, reactive oxygen species; AMPK, AMP-activated protein kinase; SCD1, stearoyl-CoA desaturase 1; xCT, cystine/glutamate transporter; Nrf2, nuclear factor erythroid 2-related factor 2; GPX4, glutathione peroxidase 4; UCKL1, uridine-cytidine kinase-like 1; SLC7A11, solute carrier family 7 member 11; CRC, colorectal cancer.

Table IV.

RNA-regulated ferroptosis in CRC.

Table IV.

RNA-regulated ferroptosis in CRC.

A, Inhibition of CRC development and progression

RNAMechanism(Refs.)
miR-148a-3pActivation of ferroptosis via targeting of SLC7A11(85)
miR-509-5pPromotion of ferroptosis via targeting of SLC7A1(86)
miR-15a-3pActivation of ferroptosis via direct targeting of GPX4(87)

B, Promotion of colorectal carcinogenesis and progression

RNA Mechanism(Refs.)

miR-19aInhibition of ferroptosis via targeting of IREB2(88)
miR-545Inhibition of ferroptosis through inhibition of transferrin(89)
lncRNA LINC00239Inhibition of ferroptosis through interaction with Kelch-like ECH-related protein 1(90)

[i] SLC7A11, solute carrier family 7 member 11; GPX4, glutathione peroxidase 4; IREB2, iron-responsive element binding protein 2; miR, microRNA; CRC, colorectal cancer.

Drug-induced ferroptosis

In 2021, Yang et al (66) reported that cetuximab inhibits the progression of KRAS-mutant CRC through the promotion of RSL3-induced ferroptosis via inhibition of the Nrf2/heme oxygenase 1 (HO-1) signaling pathway. Results of a previous study demonstrated that apatinib promotes ferroptosis in CRC cells via the ELOVL6/ACSL4 signaling pathway, highlighting the potential role of ferroptosis in CRC treatment (67). Ibrutinib enhances the sensitivity of CRC cells to ferroptosis through the BTK/nRF2 signaling pathway, thereby inhibiting CRC progression (68). In addition, Zhao et al (69) demonstrated that propofol induces ferroptosis in CRC cells via downregulation of STAT3 expression. The results of a previous study revealed that aspirin promotes RSL3-induced ferroptosis through inhibition of the mTOR/SREBP-1/SCD1 signaling pathway, highlighting the potential role of ferroptosis in the treatment of PIK3CA-mutant CRC (70).

In addition, herbs and plant extracts may induce ferroptosis. Emodin, a natural anthraquinone derivative extracted from various herbs, may induce ferroptosis in CRC cells through NCOA4-mediated ferritin autophagy and NF-κB pathways, thereby inhibiting CRC progression (71). Ginsenoside Rh3, a semi-natural product isolated from Panax ginseng, may induce ferroptosis in CRC cells via the Stat3/p53/NRF2 axis, demonstrating potential in the treatment of cancer (72). Moreover, curcumin may inhibit CRC through the induction of ferroptosis. Results of a previous study demonstrated that curcumin played a role in the regulation of oncogenes, such as P53, and the SLC7A11/glutathione/GPX4 axis (73). The combination of curcumin and Andrographis paniculata may induce ferroptosis in CRC, through the downregulation of GPX4 and iron regulatory protein 1 (74). In addition, esculin induces endoplasmic reticulum stress through the regulation of eukaryotic translation initiation factor 2α/CHOP and Nrf2/HO-1 pathways via the PERK signaling pathway; thus, promoting apoptosis and ferroptosis in CRC cells, ultimately inhibiting CRC occurrence and progression (75). Results of a previous study revealed that baicalein promotes ferroptosis through inhibition of the JAK2/STAT3/GPX4 signaling pathway; thus, exerting an inhibitory effect on CRC (76).

Gene- and protein-induced ferroptosis

Kruppel-like factor 2 (KLF2) is an oncogene that may also inhibit CRC progression. Notably, KLF2 suppresses the PI3K/AKT signaling pathway, thereby inducing ferroptosis (77). Results of a previous study demonstrated that bromelain inhibits the proliferation of KRAS-mutant CRC cells and induces ferroptosis via ACSL4 (78). Wei et al (79) revealed that Tagitinin C, a novel inducer of ferroptosis, acts as a potent chemosensitizer that enhances the efficacy of chemotherapeutic agents. Notably, Tagitinin C induces ferroptosis through the PERK/Nrf2/HO-1 signaling pathway. TIGAR, a TP53-induced regulator of glycolysis and apoptosis, plays a crucial role in energy metabolism, autophagy, stem cell differentiation and cell survival (80). Liu et al (81) demonstrated that TIGAR inhibits CRC progression through increasing the sensitivity of CRC cells to ferroptosis via the ROS/AMPK/SCD1 signaling pathway. Chaudhary et al (82) revealed that lipid carrier protein 2 inhibits ferroptosis through upregulation of GPX4 expression and the cystine/glutamate antiporter component, xCT; thus, promoting CRC progression. Moreover, increased expression of lipid carrier protein 2 may lead to resistance to 5-fluorouracil in CRC cells, further contributing to chemotherapeutic resistance. In a recent study, it has been reported that TRIM36-mediated FOXA2 promotes colorectal cancer by inhibiting the Nrf2/GPX4 signaling pathway and suppressing ferroptosis. However, the specific mechanism by which FOXA2 regulates Nrf2, whether directly or indirectly, remains unclear (83). Results of a previous study demonstrated that uridine-cytidine kinase-like 1 (UCKL1) enhances the proliferation and metastasis of CRC cells via the UCKL1/Nrf2/SLC7A11 axis, ultimately inhibiting ferroptosis and promoting CRC development and progression (84). These findings suggest that reduced lipid carrier protein 2, FOXA2 and UCKL1 expression levels may promote ferroptosis; thus, acting as an effective strategy for the treatment of CRC.

RNA-regulated ferroptosis

MicroRNAs (miRNAs/miRs) may play a role in the regulation of ferroptosis. Results of a previous study have revealed that miR-148a-3p acts as a tumor suppressor in CRC through targeting SLC7A11 and activating ferroptosis (85). In addition, miR-509-5p promotes ferroptosis through targeting SLC7A1 (86), while miR-15a-3p induces ferroptosis through the direct targeting of GPX4 (87). The oncogenic miRNA, miR-19a, promotes the proliferation, migration and invasion of CRC cells. Iron-responsive element binding protein 2 (IREB2) is a direct target of miR-19a. Thus, targeting IREB2 may lead to the inhibition of ferroptosis via miR-19a, exhibiting potential as a novel target for CRC treatment (88). In addition, results of a previous study have revealed that miR-545 suppresses transferrin, promoting CRC cell survival through the inhibition of ferroptosis (89).

Long non-coding RNAs (lncRNAs) may also regulate ferroptosis. The results of a previous study have revealed that LINC00239 may act as a ferroptosis inhibitor in CRC, through interaction with Kelch-like ECH-associated protein 1. This leads to a reduction in the antitumor activity of Erastin and RSL3, and the promotion of CRC progression (90).

Ferroptosis and the tumor microenvironment (TME)

Ferroptotic damage induces the activation of immune responses within the TME (45). Thus, further investigations into the interaction between ferroptosis and the TME may provide a basis for the development of effective strategies for CRC treatment (91).

The TME is a complex environment containing tumor cells, stromal cells, immune cells, adipocytes, endothelial cells and the extracellular matrix (92). Dai et al (93) revealed that autophagic degradation-mediated ferroptosis leads to the release of cancer cell components into the TME and tumor-associated macrophage polarization. Moreover, Ma et al (94) revealed that CD36 mediates fatty acid uptake via CD8+ T cells within the TME induced ferroptosis, leading to a reduction in CD8+ T cell effector function and antitumor activity. In addition, cancer-associated fibroblasts in the TME may impair the antitumor capacity of natural killer cells through the induction of ferroptosis (95).

Notably, CD8+ T cells resist tumor growth and metastasis via multiple mechanisms (9698). The results of a previous study have demonstrated that CD8+ T cell-derived IFN-γ promotes ferroptosis in cancer cells. This process leads to the release of a variety of tumor antigens into the TME, and antigen-presenting cell-mediated activation of CD8+ T cells, thereby enhancing anticancer immunity. However, CD8+ T cells in tumors exhibit increased levels of sensitivity to ferroptosis compared with cancer cells, which may limit the use of ferroptosis inducers in the treatment of cancer (99).

Inducing ferroptosis may attenuate drug resistance in CRC

Mitigating CRC-associated drug resistance to chemotherapy

Chemotherapy is the most common post-operative treatment for patients with CRC. The results of our previous study demonstrated that oxaliplatin and 5-fluorouracil are highly associated with ferroptosis in CRC (82,100) (Table V).

Table V.

Attenuating chemotherapeutic resistance to oxaliplatin and 5-fluorouracil through ferroptosis in colorectal cancer.

Table V.

Attenuating chemotherapeutic resistance to oxaliplatin and 5-fluorouracil through ferroptosis in colorectal cancer.

A, Direct attenuation of chemotherapeutic resistance

DrugMechanism(Refs.)
OxaliplatinInduction of ferroptosis through inhibition of the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway(100)
Increased intracellular reactive oxygen species levels and promotion of ferroptosis through inhibition of cysteine desulfurase expression levels(101)

B, Indirect attenuation of chemotherapeutic resistance

Drug Mechanism(Refs.)

OxaliplatinPromotion of ferroptosis through inhibition of RNA-binding motif single-stranded interacting protein 1 expression(102)
Promotion of ferroptosis in Candida nucleatum(103)
Promotion of ferroptosis through inhibition of cyclin-dependent kinase 1 expression(104)
5-FluorouracilPromotion of ferroptosis through inhibition of lipid transport protein 2 expression(82)
Promotion of ferroptosis through inhibition of serine protease 1 expression(105)

[i] KIF20A, kinesin family member 20A; NUAK1, NUAK family kinase 1; Nrf2, nuclear factor erythroid 2-related factor 2; GPX4, glutathione peroxidase 4.

The results of a previous study revealed that the induction of ferroptosis through the inhibition of the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway enhances sensitivity to oxaliplatin, thereby improving the quality of life of patients with CRC (100). In addition, inhibition of cysteine desulfurase expression leads to increased intracellular reactive oxygen species levels, the promotion of ferroptosis and reduced levels of resistance to oxaliplatin (101). Overexpression of RNA-binding motif single-stranded interacting protein 1 (RBMS1) inhibits ferroptosis; therefore, suppressing RBMS1 expression may increase the sensitivity of CRC cells to oxaliplatin (102). Inhibition of Candida nucleata also promotes ferroptosis and decreases CRC cell resistance to oxaliplatin (103). The results of a previous study also demonstrated that oxaliplatin resistance is reversed following inhibition of cyclin-dependent kinase 1 expression (104).

Overexpression of lipid transport protein 2 leads to inhibition of ferroptosis, which ultimately increases the resistance to 5-fluorouracil. Therefore, targeting lipid transport protein 2 levels may attenuate resistance to 5-fluorouracil (82). In addition, serine protease 1 is associated with chemotherapeutic resistance in CRC. This protein is highly expressed in CRC cells and is negatively associated with the prognosis of patients. Moreover, Liu et al (105) demonstrated that serine protease 1 interacts with SLC7A11 to increase expression levels, ultimately inhibiting ferroptosis, leading to increased resistance to 5-fluorouracil. Thus, targeting serine protease 1 may exhibit potential in the treatment of CRC.

Mitigating CRC-associated drug resistance to targeted therapies

When targeting cancer cells, chemotherapy may also target healthy cells. Thus, targeted therapy is considered an alternative treatment option to chemotherapy. Mu et al (106) demonstrated that 3-bromopyruvic acid and cetuximab may induce ferroptosis; thus, exhibiting potential in mitigating cetuximab resistance in CRC. In addition, the combination of β-elemene, a natural product derived from turmeric, and cetuximab may exert effects on metastatic CRC cells with Kras mutations, inducing ferroptosis and reducing cetuximab resistance (107).

Conclusions

At present, research is focused on the role of ferroptosis in the physiological and pathological processes of numerous diseases, leading to the development of novel treatment approaches. The present study aimed to review the specific mechanisms underlying ferroptosis in CRC, including drug-, gene-, protein- and RNA-induced ferroptosis. The aforementioned forms of ferroptosis play distinct roles in CRC. For example, various signaling pathways directly facilitate drug-induced ferroptosis, thereby impeding CRC onset and progression. By contrast, gene-, protein- and RNA-induced ferroptosis involve specific signaling pathways and mechanisms that may exhibit potential as effective strategies for targeted CRC treatment. Notably, mechanisms may include targeting of GPX4 expression and xCT, the Nrf2/GPX4 signaling pathway, the UCKL1/Nrf2/SLC7A11 signaling pathway, miR-19a, miR-545, and lncRNA LINC00239. In addition, the association between ferroptosis and the TME further highlighted that induction of ferroptosis may attenuate drug resistance in CRC.

Results of the present study demonstrated that induction of ferroptosis may inhibit CRC progression. In addition, genes, proteins and RNAs that inhibit ferroptosis may promote CRC development and progression. However, the clinical application of ferroptosis is limited at present, as the specific mechanisms and signaling pathways through which ferroptosis promotes or inhibits CRC development are yet to be fully elucidated. Thus, further investigations into the specific role of ferroptosis in CRC are required for the development of effective targeted therapies.

In conclusion, ferroptosis may exhibit potential in the treatment of CRC. Further investigations are required to elucidate the signaling pathways involved in ferroptosis, and to identify the genes, proteins and signaling pathways that inhibit ferroptosis in CRC. Moreover, further investigations should focus on determining alternative therapeutic modalities that may increase the therapeutic effects of ferroptosis, and on assessing the specific anti-tumor effects of ferroptosis in CRC progression. An increased understanding of ferroptosis in CRC may lead to the effective implementation of treatment in clinical practice.

Acknowledgements

The figures were created using Figdraw.

Funding

This article is funded by Shandong Province's Traditional Chinese Medicine Science and Technology Development Project (grant no. 2019-0187) and Qilu Chinese Medicine Advantageous Specialty Cluster Construction Project (grant no. YWC2022ZKJQ0003).

Availability of data and materials

Not applicable.

Authors' contributions

ZG, HZ and XS contributed to the conception and the main idea of the work. ZG and HZ drafted the manuscript, figures and tables. XS reviewed and modified the manuscript. All authors read and approved the final version of the manuscript. Data authentication is not applicable.

Ethics approval and consent to participate

Not applicable.

Ethics approval and consent to participate

Not applicable.

Competing interests

All authors declare that they have no competing interests.

References

1 

Favoriti P, Carbone G, Greco M, Pirozzi F, Pirozzi RE and Corcione F: Worldwide burden of colorectal cancer: A review. Updates Surg. 68:7–11. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

3 

Bouvard V, Loomis D, Guyton KZ, Grosse Y, Ghissassi FE, Benbrahim-Tallaa L, Guha N, Mattock H and Straif K; International Agency for Research on Cancer Monograph Working Group, : Carcinogenicity of consumption of red and processed meat. Lancet Oncol. 16:1599–1600. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Johnson CH, Dejea CM, Edler D, Hoang LT, Santidrian AF, Felding BH, Ivanisevic J, Cho K, Wick EC, Hechenbleikner EM, et al: Metabolism links bacterial biofilms and colon carcinogenesis. Cell Metab. 21:891–897. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Dejea CM, Wick EC, Hechenbleikner EM, White JR, Mark Welch JL, Rossetti BJ, Peterson SN, Snesrud EC, Borisy GG, Lazarev M, et al: Microbiota organization is a distinct feature of proximal colorectal cancers. Proc Natl Acad Sci USA. 111:18321–18326. 2014. View Article : Google Scholar : PubMed/NCBI

6 

Benson AB, Venook AP, Al-Hawary MM, Cederquist L, Chen YJ, Ciombor KK, Cohen S, Cooper HS, Deming D, Engstrom PF, et al: NCCN guidelines insights: Colon cancer, version 2.2018. J Natl Compr Canc Netw. 16:359–369. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Guo J, Xu B, Han Q, Zhou H, Xia Y, Gong C, Dai X, Li Z and Wu G: Ferroptosis: A novel Anti-tumor action for cisplatin. Cancer Res Treat. 50:445–460. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Conrad M, Dixon SJ, Fulda S, Gascón S, Hatzios SK, Kagan VE, et al: Ferroptosis: A regulated cell death nexus linking metabolism, redox biology, and disease. Cell. 171:273–285. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, et al: Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell. 149:1060–1072. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Xu S, He Y, Lin L, Chen P, Chen M and Zhang S: The emerging role of ferroptosis in intestinal disease. Cell Death Dis. 12:2892021. View Article : Google Scholar : PubMed/NCBI

11 

Yagoda N, von Rechenberg M, Zaganjor E, Bauer AJ, Yang WS, Fridman DJ, Wolpaw AJ, Smukste I, Peltier JM, Boniface JJ, et al: RAS-RAF-MEK-dependent oxidative cell death involving voltage-dependent anion channels. Nature. 447:865–869. 2007. View Article : Google Scholar

12 

Li J, Cao F, Yin HL, Huang ZJ, Lin ZT, Mao N, Sun B and Wang G: Ferroptosis: Past, present and future. Cell Death Dis. 11:882020. View Article : Google Scholar : PubMed/NCBI

13 

Ketelut-Carneiro N and Fitzgerald KA: Apoptosis, pyroptosis, and Necroptosis-oh my! The many ways a cell can die. J Mol Biol. 434:1673782022. View Article : Google Scholar : PubMed/NCBI

14 

Aglietti RA and Dueber EC: Recent insights into the molecular mechanisms underlying pyroptosis and gasdermin family functions. Trends Immunol. 38:261–271. 2017. View Article : Google Scholar : PubMed/NCBI

15 

Fang Y, Tian S, Pan Y, Li W, Wang Q, Tang Y, Yu T, Wu X, Shi Y, Ma P and Shu Y: Pyroptosis: A new frontier in cancer. Biomed Pharmacother. 121:1095952020. View Article : Google Scholar : PubMed/NCBI

16 

Liu X, Zhang Z, Ruan J, Pan Y, Magupalli VG, Wu H and Lieberman J: Inflammasome-activated gasdermin D causes pyroptosis by forming membrane pores. Nature. 535:153–158. 2016. View Article : Google Scholar : PubMed/NCBI

17 

Sun L, Wang H, Wang Z, He S, Chen S, Liao D, Wang L, Yan J, Liu W, Lei X and Wang X: Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell. 148:213–227. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Negroni A, Colantoni E, Cucchiara S and Stronati L: Necroptosis in intestinal inflammation and cancer: New concepts and therapeutic perspectives. Biomolecules. 10:14312020. View Article : Google Scholar : PubMed/NCBI

19 

Liu S, Yao S, Yang H, Liu S and Wang Y: Autophagy: Regulator of cell death. Cell Death Dis. 14:6482023. View Article : Google Scholar : PubMed/NCBI

20 

Guiney SJ, Adlard PA, Bush AI, Finkelstein DI and Ayton S: Ferroptosis and cell death mechanisms in Parkinson's disease. Neurochem Int. 104:34–48. 2017. View Article : Google Scholar : PubMed/NCBI

21 

Do Van B, Gouel F, Jonneaux A, Timmerman K, Gelé P, Pétrault M, Bastide M, Laloux C, Moreau C, Bordet R, et al: Ferroptosis, a newly characterized form of cell death in Parkinson's disease that is regulated by PKC. Neurobiol Dis. 94:169–178. 2016. View Article : Google Scholar : PubMed/NCBI

22 

Yan N and Zhang J: Iron metabolism, ferroptosis, and the links with Alzheimer's disease. Front Neurosci. 13:14432020. View Article : Google Scholar : PubMed/NCBI

23 

Cong L, Dong X, Wang Y, Deng Y, Li B and Dai R: On the role of synthesized hydroxylated chalcones as dual functional amyloid-β aggregation and ferroptosis inhibitors for potential treatment of Alzheimer's disease. Eur J Med Chem. 166:11–21. 2019. View Article : Google Scholar : PubMed/NCBI

24 

Deng G, Li Y, Ma S, Gao Z, Zeng T, Chen L, Ye H, Yang M, Shi H, Yao X, et al: Caveolin-1 dictates ferroptosis in the execution of acute immune-mediated hepatic damage by attenuating nitrogen stress. Free Radic Biol Med. 148:151–161. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Park SJ, Cho SS, Kim KM, Yang JH, Kim JH, Jeong EH, Yang JW, Han CY, Ku SK, Cho IJ and Ki SH: Protective effect of sestrin2 against iron overload and ferroptosis-induced liver injury. Toxicol Appl Pharmacol. 379:1146652019. View Article : Google Scholar : PubMed/NCBI

26 

Zhang Z, Wu Y, Yuan S, Zhang P, Zhang J, Li H, Li X, Shen H, Wang Z and Chen G: Glutathione peroxidase 4 participates in secondary brain injury through mediating ferroptosis in a rat model of intracerebral hemorrhage. Brain Res. 1701:112–125. 2018. View Article : Google Scholar : PubMed/NCBI

27 

Kenny EM, Fidan E, Yang Q, Anthonymuthu TS, New LA, Meyer EA, Wang H, Kochanek PM, Dixon CE, Kagan VE and Bayir H: Ferroptosis contributes to neuronal death and functional outcome after traumatic brain injury. Crit Care Med. 47:410–418. 2019. View Article : Google Scholar : PubMed/NCBI

28 

Yao X, Zhang Y, Hao J, Duan HQ, Zhao CX, Sun C, Li B, Fan BY, Wang X, Li WX, et al: Deferoxamine promotes recovery of traumatic spinal cord injury by inhibiting ferroptosis. Neural Regen Res. 14:5322019. View Article : Google Scholar : PubMed/NCBI

29 

Zhang Y, Sun C, Zhao C, Hao J, Zhang Y, Fan B, Li B, Duan H, Liu C, Kong X, et al: Ferroptosis inhibitor SRS 16–86 attenuates ferroptosis and promotes functional recovery in contusion spinal cord injury. Brain Res. 1706:48–57. 2019. View Article : Google Scholar : PubMed/NCBI

30 

Shi Z, Yuan S, Shi L, Li J, Ning G, Kong X and Feng S: Programmed cell death in spinal cord injury pathogenesis and therapy. Cell Prolif. 54:e129922021. View Article : Google Scholar : PubMed/NCBI

31 

Martin-Sanchez D, Ruiz-Andres O, Poveda J, Carrasco S, Cannata-Ortiz P, Sanchez-Niño MD, Ruiz Ortega M, Egido J, Linkermann A, Ortiz A and Sanz AB: Ferroptosis, but not necroptosis, is important in nephrotoxic folic Acid-induced AKI. J Am Soc Nephrol. 28:218–229. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Müller T, Dewitz C, Schmitz J, Schröder AS, Bräsen JH, Stockwell BR, Murphy JM, Kunzendorf U and Krautwald S: Necroptosis and ferroptosis are alternative cell death pathways that operate in acute kidney failure. Cell Mol Life Sci. 74:3631–3645. 2017. View Article : Google Scholar : PubMed/NCBI

33 

Liu B, Zhao C, Li H, Chen X, Ding Y and Xu S: Puerarin protects against heart failure induced by pressure overload through mitigation of ferroptosis. Biochem Biophys Res Commun. 497:233–240. 2018. View Article : Google Scholar : PubMed/NCBI

34 

Chen X, Xu S, Zhao C and Liu B: Role of TLR4/NADPH oxidase 4 pathway in promoting cell death through autophagy and ferroptosis during heart failure. Biochem Biophys Res Commun. 516:37–43. 2019. View Article : Google Scholar : PubMed/NCBI

35 

Li D, Pi W, Sun Z, Liu X and Jiang J: Ferroptosis and its role in cardiomyopathy. Biomed Pharmacother. 153:1132792022. View Article : Google Scholar : PubMed/NCBI

36 

Ng SW, Norwitz SG and Norwitz ER: The impact of iron overload and ferroptosis on reproductive disorders in humans: Implications for preeclampsia. Int J Mol Sci. 20:32832019. View Article : Google Scholar : PubMed/NCBI

37 

Ng SW, Norwitz SG, Taylor HS and Norwitz ER: Endometriosis: The role of iron overload and ferroptosis. Reprod Sci. 27:1383–1390. 2020. View Article : Google Scholar : PubMed/NCBI

38 

Yan HF, Zou T, Tuo QZ, Xu S, Li H, Belaidi AA and Lei P: Ferroptosis: Mechanisms and links with diseases. Signal Transduct TargetTher. 6:492021. View Article : Google Scholar

39 

Yan H, Talty R, Aladelokun O, Bosenberg M and Johnson CH: Ferroptosis in colorectal cancer: A future target? Br J Cancer. 128:1439–1451. 2023. View Article : Google Scholar : PubMed/NCBI

40 

Wu T, Wan J, Qu X, Xia K, Wang F, Zhang Z, Yang M, Wu X, Gao R, Yuan X, et al: Nodal promotes colorectal cancer survival and metastasis through regulating SCD1-mediated ferroptosis resistance. Cell Death Dis. 14:2292023. View Article : Google Scholar : PubMed/NCBI

41 

Wang Y, Zhang Z, Sun W, Zhang J, Xu Q, Zhou X and Mao L: Ferroptosis in colorectal cancer: Potential mechanisms and effective therapeutic targets. Biomed Pharmacother. 153:1135242022. View Article : Google Scholar : PubMed/NCBI

42 

Jiang L, Hickman JH, Wang SJ and Gu W: Dynamic roles of p53-mediated metabolic activities in ROS-induced stress responses. Cell Cycle. 14:2881–2885. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Jiang L, Kon N, Li T, Wang SJ, Su T, Hibshoosh H, Baer R and Gu W: Ferroptosis as a p53-mediated activity during tumour suppression. Nature. 520:57–62. 2015. View Article : Google Scholar : PubMed/NCBI

44 

Li Q, Chen K, Zhang T, Jiang D, Chen L, Jiang J, Zhang C and Li S: Understanding Sorafenib-induced ferroptosis and resistance mechanisms: Implications for cancer therapy. Eur J Pharmacol. 955:1759132023. View Article : Google Scholar : PubMed/NCBI

45 

Chen X, Kang R, Kroemer G and Tang D: Broadening horizons: The role of ferroptosis in cancer. Nat Rev Clin Oncol. 18:280–296. 2021. View Article : Google Scholar : PubMed/NCBI

46 

Wang L, Liu Y, Du T, Yang H, Lei L, Guo M, Ding HF, Zhang J, Wang H, Chen X and Yan C: ATF3 promotes Erastin-induced ferroptosis by suppressing system Xc. Cell Death Differ. 27:662–675. 2020. View Article : Google Scholar : PubMed/NCBI

47 

Yang WS and Stockwell BR: Synthetic lethal screening identifies compounds activating Iron-dependent, nonapoptotic cell death in oncogenic-RAS-harboring cancer cells. Chem Biol. 15:234–245. 2008. View Article : Google Scholar : PubMed/NCBI

48 

Sui X, Zhang R, Liu S, Duan T, Zhai L, Zhang M, Han X, Xiang Y, Huang X, Lin H and Xie T: RSL3 drives ferroptosis through GPX4 inactivation and ROS production in colorectal cancer. Front Pharmacol. 9:13712018. View Article : Google Scholar : PubMed/NCBI

49 

Costa I, Barbosa DJ, Benfeito S, Silva V, Chavarria D, Borges F, Remião F and Silva R: Molecular mechanisms of ferroptosis and their involvement in brain diseases. Pharmacol Ther. 244:1083732023. View Article : Google Scholar : PubMed/NCBI

50 

Harayama T and Riezman H: Understanding the diversity of membrane lipid composition. Nat Rev Mol Cell Biol. 19:281–296. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH, Liu B, Tyurin VA, Ritov VB, et al: Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat Chem Biol. 13:81–90. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Koppula P, Zhuang L and Gan B: Cytochrome P450 reductase (POR) as a ferroptosis fuel. Protein Cell. 12:675–679. 2021. View Article : Google Scholar : PubMed/NCBI

53 

Yan B, Ai Y, Sun Q, Ma Y, Cao Y, Wang J, Zhang Z and Wang X: Membrane damage during ferroptosis is caused by oxidation of phospholipids catalyzed by the oxidoreductases POR and CYB5R1. Mol Cell. 81:355–369.e10. 2021. View Article : Google Scholar : PubMed/NCBI

54 

Zou Y, Li H, Graham ET, Deik AA, Eaton JK, Wang W, Sandoval-Gomez G, Clish CB, Doench JG and Schreiber SL: Cytochrome P450 oxidoreductase contributes to phospholipid peroxidation in ferroptosis. Nat Chem Biol. 16:302–309. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Ning X, Qi H, Yuan Y, Li R, Wang Y, Lin Z and Yin Y: Identification of a new small molecule that initiates ferroptosis in cancer cells by inhibiting the system Xc to deplete GSH. Eur J Pharmacol. 934:1753042022. View Article : Google Scholar : PubMed/NCBI

56 

Fan X, Li A, Yan Z, Geng X, Lian L, Lv H, Gao D and Zhang J: From iron metabolism to ferroptosis: Pathologic changes in coronary heart disease. Oxid Med Cell Longev. 2022:62918892022. View Article : Google Scholar : PubMed/NCBI

57 

Zhou L, Zhao B, Zhang L, Wang S, Dong D, Lv H and Shang P: Alterations in cellular iron metabolism provide more therapeutic opportunities for cancer. Int J Mol Sci. 19:15452018. View Article : Google Scholar : PubMed/NCBI

58 

Basak T and Kanwar RK: Iron imbalance in cancer: Intersection of deficiency and overload. Cancer Med. 11:3837–3853. 2022. View Article : Google Scholar : PubMed/NCBI

59 

Chifman J, Laubenbacher R and Torti SV: A systems biology approach to iron metabolism. Adv Exp Med Biol. 844:201–225. 2014. View Article : Google Scholar : PubMed/NCBI

60 

Han C, Liu Y, Dai R, Ismail N, Su W and Li B: Ferroptosis and its potential role in human diseases. Front Pharmacol. 11:2392020. View Article : Google Scholar : PubMed/NCBI

61 

Manz DH, Blanchette NL, Paul BT, Torti FM and Torti SV: Iron and cancer: Recent insights. Ann N Y Acad Sci. 1368:149–161. 2016. View Article : Google Scholar : PubMed/NCBI

62 

Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G and Tang D: Ferroptosis is a type of Autophagy-dependent cell death. Semin Cancer Biol. 66:89–100. 2020. View Article : Google Scholar : PubMed/NCBI

63 

Mancias JD, Wang X, Gygi SP, Harper JW and Kimmelman AC: Quantitative proteomics identifies NCOA4 as the cargo receptor mediating ferritinophagy. Nature. 509:105–109. 2014. View Article : Google Scholar : PubMed/NCBI

64 

Hou W, Xie Y, Song X, Sun X, Lotze MT, Zeh HJ III, Kang R and Tang D: Autophagy promotes ferroptosis by degradation of ferritin. Autophagy. 12:1425–1428. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, Kang R and Tang D: Ferroptosis: Process and function. Cell Death Differ. 23:369–379. 2016. View Article : Google Scholar : PubMed/NCBI

66 

Yang J, Mo J, Dai J, Ye C, Cen W, Zheng X, Jiang L and Ye L: Cetuximab promotes RSL3-induced ferroptosis by suppressing the Nrf2/HO-1 signalling pathway in KRAS mutant colorectal cancer. Cell Death Dis. 12:10792021. View Article : Google Scholar : PubMed/NCBI

67 

Tian X, LI S and Ge G: Apatinib promotes ferroptosis in colorectal cancer cells by targeting ELOVL6/ACSL4 Signaling. Cancer Manag Res. 13:1333–1342. 2021. View Article : Google Scholar : PubMed/NCBI

68 

Zhu JF, Liu Y, Li WT, Li MH, Zhen CH, Sun PW, Chen JX, Wu WH and Zeng W: Ibrutinib facilitates the sensitivity of colorectal cancer cells to ferroptosis through BTK/NRF2 pathway. Cell Death Dis. 14:1512023. View Article : Google Scholar : PubMed/NCBI

69 

Zhao X and Chen F: Propofol induces the ferroptosis of colorectal cancer cells by downregulating STAT3 expression. Oncol Lett. 22:7672021. View Article : Google Scholar : PubMed/NCBI

70 

Chen H, Qi Q, Wu N, Wang Y, Feng Q, Jin R and Jiang L: Aspirin promotes RSL3-induced ferroptosis by suppressing mTOR/SREBP-1/SCD1-mediated lipogenesis in PIK3CA-mutant colorectal cancer. Redox Biol. 55:1024262022. View Article : Google Scholar : PubMed/NCBI

71 

Shen Z, Zhao L, Yoo SA, Lin Z, Zhang Y, Yang W and Piao J: Emodin induces ferroptosis in colorectal cancer through NCOA4-mediated ferritinophagy and NF-κb pathway inactivation. Apoptosis. May 5–2024.doi: 10.1007/s10495-024-01973-2 (Epub ahead of print). View Article : Google Scholar

72 

Wu Y, Pi D, Zhou S, Yi Z, Dong Y, Wang W, Ye H, Chen Y, Zuo Q and Ouyang M: Ginsenoside Rh3 induces pyroptosis and ferroptosis through the Stat3/p53/NRF2 axis in colorectal cancer cells. Acta Biochim Biophys Sin (Shanghai). 55:587–600. 2023. View Article : Google Scholar : PubMed/NCBI

73 

Ming T, Lei J, Peng Y, Wang M, Liang Y, Tang S, Tao Q, Wang M, Tang X, He Z, et al: Curcumin suppresses colorectal cancer by induction of ferroptosis via regulation of p53 and solute carrier family 7 member 11/glutathione/glutathione peroxidase 4 signaling axis. Phytother Res. 38:3954–3972. 2024. View Article : Google Scholar : PubMed/NCBI

74 

Miyazaki K, Xu C, Shimada M and Goel A: Curcumin and andrographis exhibit Anti-tumor effects in colorectal cancer via activation of ferroptosis and dual suppression of glutathione Peroxidase-4 and ferroptosis suppressor Protein-1. Pharmaceuticals (Basel). 16:3832023. View Article : Google Scholar : PubMed/NCBI

75 

Ji X, Chen Z, Lin W, Wu Q, Wu Y, Hong Y, Tong H, Wang C and Zhang Y: Esculin induces endoplasmic reticulum stress and drives apoptosis and ferroptosis in colorectal cancer via PERK regulating eIF2α/CHOP and Nrf2/HO-1 cascades. J Ethnopharmacol. 328:1181392024. View Article : Google Scholar : PubMed/NCBI

76 

Lai JQ, Zhao LL, Hong C, Zou QM, Su JX, Li SJ, Zhou XF, Li ZS, Deng B, Cao J and Qi Q: Baicalein triggers ferroptosis in colorectal cancer cells via blocking the JAK2/STAT3/GPX4 axis. Acta Pharmacol Sin. 45:1715–1726. 2024. View Article : Google Scholar : PubMed/NCBI

77 

Li J, Jiang JL, Chen YM and Lu WQ: KLF2 inhibits colorectal cancer progression and metastasis by inducing ferroptosis via the PI3K/AKT signaling pathway. J Pathol Clin Res. 9:423–435. 2023. View Article : Google Scholar : PubMed/NCBI

78 

Park S, Oh J, Kim M and Jin EJ: Bromelain effectively suppresses Kras-mutant colorectal cancer by stimulating ferroptosis. Anim Cells Syst (Seoul). 22:334–340. 2018. View Article : Google Scholar : PubMed/NCBI

79 

Wei R, Zhao Y, Wang J, Yang X, Li S, Wang Y, Yang X, Fei J, Hao X, Zhao Y, et al: Tagitinin C induces ferroptosis through PERK-Nrf2-HO-1 signaling pathway in colorectal cancer cells. Int J Biol Sci. 17:2703–2717. 2021. View Article : Google Scholar : PubMed/NCBI

80 

Ko YH, Domingo-Vidal M, Roche M, Lin Z, Whitaker-Menezes D, Seifert E, Capparelli C, Tuluc M, Birbe RC, Tassone P, et al: TP53-inducible glycolysis and apoptosis regulator (TIGAR) metabolically reprograms carcinoma and stromal cells in breast cancer. J Biol Chem. 291:26291–26303. 2016. View Article : Google Scholar : PubMed/NCBI

81 

Liu MY, Li HM, Wang XY, Xia R, Li X, Ma YJ, Wang M and Zhang HS: TIGAR drives colorectal cancer ferroptosis resistance through ROS/AMPK/SCD1 pathway. Free Radic Biol Med. 182:219–231. 2022. View Article : Google Scholar : PubMed/NCBI

82 

Chaudhary N, Choudhary BS, Shah SG, Khapare N, Dwivedi N, Gaikwad A, Joshi N, Raichanna J, Basu S, Gurjar M, et al: Lipocalin 2 expression promotes tumor progression and therapy resistance by inhibiting ferroptosis in colorectal cancer. Int J Cancer. 149:1495–1511. 2021. View Article : Google Scholar : PubMed/NCBI

83 

Liu X, Yan C, Chang C, Meng F, Shen W, Wang S and Zhang Y: FOXA2 suppression by TRIM36 exerts Anti-tumor role in colorectal cancer via inducing NRF2/GPX4-Regulated ferroptosis. Adv Sci (Weinh). 10:e23045212023. View Article : Google Scholar : PubMed/NCBI

84 

Wu W, Zhao Y, Qin B, Jiang X, Wang C, Hu R, Ma R, Lee MH, Liu H, Li K and Yuan P: Non-canonical role of UCKL1 on ferroptosis defence in colorectal cancer. EBioMedicine. 93:1046502023. View Article : Google Scholar : PubMed/NCBI

85 

Martino E, Balestrieri A, Aragona F, Bifulco G, Mele L, Campanile G, Balestrieri ML and D'Onofrio N: MiR-148a-3p promotes colorectal cancer cell ferroptosis by targeting SLC7A11. Cancers (Basel). 15:43422023. View Article : Google Scholar : PubMed/NCBI

86 

Elrebehy MA, Abdelghany TM, Elshafey MM, Gomaa MH and Doghish AS: miR-509-5p promotes colorectal cancer cell ferroptosis by targeting SLC7A11. Pathol Res Pract. 247:1545572023. View Article : Google Scholar : PubMed/NCBI

87 

Liu L, Yao H, Zhou X, Chen J, Chen G, Shi X, Wu G, Zhou G and He S: MiR-15a-3p regulates ferroptosis via targeting glutathione peroxidase GPX4 in colorectal cancer. Mol Carcinog. 61:301–310. 2022. View Article : Google Scholar : PubMed/NCBI

88 

Fan H, Ai R, Mu S, Niu X, Guo Z and Liu L: MiR-19a suppresses ferroptosis of colorectal cancer cells by targeting IREB2. Bioengineered. 13:12021–12029. 2022. View Article : Google Scholar : PubMed/NCBI

89 

Zheng S, Hu L, Song Q, Shan Y, Yin G, Zhu H, Kong W and Zhou C: miR-545 promotes colorectal cancer by inhibiting transferring in the non-normal ferroptosis signaling. Aging. 13:26137–26147. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Han Y, Gao X, Wu N, Jin Y, Zhou H, Wang W, Liu H, Chu Y, Cao J, Jiang M, et al: Long noncoding RNA LINC00239 inhibits ferroptosis in colorectal cancer by binding to Keap1 to stabilize Nrf2. Cell Death Dis. 13:7422022. View Article : Google Scholar : PubMed/NCBI

91 

Binnewies M, Roberts EW, Kersten K, Chan V, Fearon DF, Merad M, Coussens LM, Gabrilovich DI, Ostrand-Rosenberg S, Hedrick CC, et al: Understanding the tumor immune microenvironment (TIME) for effective therapy. Nat Med. 24:541–550. 2018. View Article : Google Scholar : PubMed/NCBI

92 

Chen F, Zhuang X, Lin L, Yu P, Wang Y, Shi Y, Hu G and Sun Y: New horizons in tumor microenvironment biology: Challenges and opportunities. BMC Med. 13:452015. View Article : Google Scholar : PubMed/NCBI

93 

Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, Zeh HJ, Kang R, Wang J and Tang D: Autophagy-dependent ferroptosis drives tumor-associated macrophage polarization via release and uptake of oncogenic KRAS protein. Autophagy. 16:2069–2083. 2020. View Article : Google Scholar : PubMed/NCBI

94 

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 : PubMed/NCBI

95 

Yao L, Hou J, Wu X, Lu Y, Jin Z, Yu Z, Yu B, Li J, Yang Z, Li C, et al: Cancer-associated fibroblasts impair the cytotoxic function of NK cells in gastric cancer by inducing ferroptosis via iron regulation. Redox Biol. 67:1029232023. View Article : Google Scholar : PubMed/NCBI

96 

St Paul M and Ohashi PS: The roles of CD8+ T cell subsets in antitumor immunity. Trends Cell Biol. 30:695–704. 2020. View Article : Google Scholar : PubMed/NCBI

97 

Tang R, Xu J, Zhang B, Liu J, Liang C, Hua J, Meng Q, Yu X and Shi S: Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol. 13:1102020. View Article : Google Scholar : PubMed/NCBI

98 

Han J, Khatwani N, Searles TG, Turk MJ and Angeles CV: Memory CD8+ T cell responses to cancer. Semin Immunol. 49:1014352020. View Article : Google Scholar : PubMed/NCBI

99 

Lin Z, Zou S and Wen K: The crosstalk of CD8+ T cells and ferroptosis in cancer. Front Immunol. 14:12554432024. View Article : Google Scholar : PubMed/NCBI

100 

Yang C, Zhang Y, Lin S, Liu Y and Li W: Suppressing the KIF20A/NUAK1/Nrf2/GPX4 signaling pathway induces ferroptosis and enhances the sensitivity of colorectal cancer to oxaliplatin. Aging (Albany NY). 13:13515–13534. 2021. View Article : Google Scholar : PubMed/NCBI

101 

Lin JF, Hu PS, Wang YY, Tan YT, Yu K, Liao K, Wu QN, Li T, Meng Q, Lin JZ, et al: Phosphorylated NFS1 weakens oxaliplatin-based chemosensitivity of colorectal cancer by preventing PANoptosis. Signal Transduct Target Ther. 7:542022. View Article : Google Scholar : PubMed/NCBI

102 

Xu Y, Hao J, Chen Q, Qin Y, Qin H, Ren S, Sun C, Zhu Y, Shao B, Zhang J and Wang H: Inhibition of the RBMS1/PRNP axis improves ferroptosis resistance-mediated oxaliplatin chemoresistance in colorectal cancer. Mol Carcinog. 63:224–237. 2024. View Article : Google Scholar : PubMed/NCBI

103 

Li B, Wei Z, Wang Z, Xu F, Yang J, Lin B, Chen Y, Wenren H, Wu L, Guo X, et al: Fusobacterium nucleatum induces oxaliplatin resistance by inhibiting ferroptosis through E-cadherin/β-catenin/GPX4 axis in colorectal cancer. Free Radic Biol Med. 220:125–138. 2024. View Article : Google Scholar : PubMed/NCBI

104 

Zeng K, Li W, Wang Y, Zhang Z, Zhang L, Zhang W, Xing Y and Zhou C: Inhibition of CDK1 Overcomes oxaliplatin resistance by regulating ACSL4-mediated ferroptosis in colorectal cancer. Adv Sci (Weinh). 10:23010882023. View Article : Google Scholar : PubMed/NCBI

105 

Liu W, Liu C, Xiao J, Qian C, Chen Z, Lin W, Zhang Y, Wu J, Zhou R and Zhao L: HTRA1 interacts with SLC7A11 to modulate colorectal cancer chemosensitivity by inhibiting ferroptosis. Cell Death Discovery. 10:2282024. View Article : Google Scholar : PubMed/NCBI

106 

Mu M, Zhang Q, Zhao C, Li X, Chen Z, Sun X and Yu J: 3-Bromopyruvate overcomes cetuximab resistance in human colorectal cancer cells by inducing Autophagy-dependent ferroptosis. Cancer Gene Therapy. 30:1414–1425. 2023. View Article : Google Scholar : PubMed/NCBI

107 

Chen P, Li X, Zhang R, Liu S, Xiang Y, Zhang M, Chen X, Pan T, Yan L, Feng J, et al: Combinative treatment of β-elemene and cetuximab is sensitive to KRAS mutant colorectal cancer cells by inducing ferroptosis and inhibiting Epithelial-mesenchymal transformation. Theranostics. 10:5107–5119. 2020. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2024
Volume 28 Issue 6

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo Z, Zhuang H and Shi X: Therapeutic efficacy of ferroptosis in the treatment of colorectal cancer (Review). Oncol Lett 28: 563, 2024.
APA
Guo, Z., Zhuang, H., & Shi, X. (2024). Therapeutic efficacy of ferroptosis in the treatment of colorectal cancer (Review). Oncology Letters, 28, 563. https://doi.org/10.3892/ol.2024.14697
MLA
Guo, Z., Zhuang, H., Shi, X."Therapeutic efficacy of ferroptosis in the treatment of colorectal cancer (Review)". Oncology Letters 28.6 (2024): 563.
Chicago
Guo, Z., Zhuang, H., Shi, X."Therapeutic efficacy of ferroptosis in the treatment of colorectal cancer (Review)". Oncology Letters 28, no. 6 (2024): 563. https://doi.org/10.3892/ol.2024.14697