Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 15.
Published in final edited form as: Mol Aspects Med. 2020 Apr 25;75:100860. doi: 10.1016/j.mam.2020.100860

Iron: The Cancer Connection

Suzy V Torti a,*, Frank M Torti b
PMCID: PMC9107937  NIHMSID: NIHMS1801683  PMID: 32340745

Abstract

Iron plays an essential role in normal biological processes: The generation of cellular energy, oxygen transport, DNA synthesis and repair are all processes that require iron-coordinated proteins, either as elemental iron, heme or iron-sulfur clusters. As a transition metal with two major biological oxidation states, iron is also a critical intermediate in the generation of reactive oxygen species that can damage cellular structures and contribute to both aging and cancer. In this review, we focus on experimental and epidemiologic evidence that links iron and cancer, as well as strategies that have been proposed to either reduce or increase cellular iron for cancer therapy.

Keywords: transferrin receptor, ferroportin, metastasis, iron chelator, ferroptosis

1. Introduction

Pathways that govern iron metabolism in non-malignant cells are perturbed in cancer cells. Here, we provide an overview of the links between iron and cancer, specifically the evidence that iron can contribute to the initiation, growth and metastatic spread of tumors. We also discuss targeting iron as a potential anti-cancer strategy. For a more detailed description of iron regulatory pathways in normal cells, the reader is referred to other chapters in this book. We also refer the reader to recent reviews that delve more extensively into particular facets of the relationship between iron and cancer at the cellular and systemic level, including the role of iron in immune metabolism1 and roles of heme iron2.

2. Iron as a carcinogen

Iron is an essential nutrient that is required for functions ranging from oxygen transport to respiration to DNA synthesis. However, due to its ability to redox cycle (gain and lose electrons), iron also has the potential to participate in deleterious reactions that produce free radicals. In particular, iron catalyzes the formation of hydroxyl and possibly ferryl radicals3(Fenton chemistry), two species that can damage DNA, lipids and proteins, and are major contributors to oxidative stress48.

The ability of iron to catalyze the formation of DNA strand breaks and oxidize DNA bases9 renders it a potential mutagen. When mutations occur in critical genes that regulate cell growth and proliferation, cancer ensues. In most cancers, it is the accumulation of multiple mutations over time that is thought to drive cancer -- single oncogenic events such as the (9;22)(q34;q11.2) translocation “Philadelphia chromosome” that gives rise to the BCR-ABL1 fusion protein and results in CML10 are the exception rather than the rule. Thus, we believe that iron can be considered a driver in an oncogenic program: as cancer cells acquire more iron, more radicals are formed, and more mutations are acquired.

3. Experimental links between iron and cancer

What is the evidence for a mutagenic activity of iron? The link between excess iron and cancer was observed decades ago, in studies demonstrating that exposure to iron oxide dust triples the incidence of pulmonary tumors in mice11, and that intramuscular injection of iron dextran induces sarcoma in rats12. Work begun in the 80’s13 demonstrated that a single injection of iron (ferric nitrilotriacetate) induces oxidative DNA damage in rat kidneys and gives rise to tumors that exhibit a mutational spectrum similar (within limits) to that seen in human kidney cancer14,15. Another link between iron and carcinogenic mutations is seen in patients with hemochromatosis, a genetic disease that can lead to iron overload and an increased risk of developing hepatocellular carcinoma16. Such patients exhibit an accumulation of p53 mutations17 and epigenetic changes in the liver18 that are characteristic of hepatocellular carcinoma, even in non-malignant tissue. Such findings suggest that the link between iron, excess mutation and tumor formation is pertinent to human disease. Although all these examples involve exposure to supra-physiological levels of iron, they demonstrate the potential of iron to foster carcinogenic events in vivo.

As an essential nutrient, iron is also required for tumor growth. Early experimental studies demonstrated the ability of excess dietary iron to enhance tumor growth in mice1922. Tumor incidence in mice that spontaneously develop intestinal tumors due to heterozygosity in the APC tumor suppressor is similarly increased in mice fed a high iron diet and decreased in mice fed an iron-restricted diet23. Conversely, iron withdrawal, produced by treatment with small molecule iron chelators or by dietary manipulation, inhibits tumor growth both in spontaneous and xenograft models of cancer24,25.

A genetically engineered mouse model with tissue-specific deletion of FBXL5 in the mouse liver was recently used to more explicitly demonstrate the effects of hepatic iron overload on cancer26. FBXL5 is an ubiquitin ligase that degrades IRP2 (iron regulatory protein 2), a protein that post-translationally controls expression of transferrin receptor (TFR1), ferritin, and ferroportin. In the absence of FBXL5, IRP2 is constitutively expressed, resulting in TFR1 stability (enhanced iron uptake) and repression of ferroportin and ferritin (decreased iron efflux and storage, respectively). Collectively, these molecular events lead to hepatic iron overload27. Consistent with the pro-tumorigenic activity of iron, FBXL5-deficient mice exhibit oxidative stress and develop liver tumors late in life. Moreover they exhibit a high hepatic tumor incidence and increased tumor size following exposure to chemical (diethylnitrosamine; DEN) or viral carcinogens (hepatitis C viral transgene26).

These and other observations illustrating the pro-tumorigenic effects of iron and the anti-tumorigenic effects of iron depletion form the conceptual foundation for the strategy to use iron chelation in anti-cancer therapy (discussed in Section 8 below).

4. Epidemiology of iron and cancer

Population-based studies have examined the relationships between iron intake and/or stores of iron and human cancer risk. Many, but not all studies have observed a link between high iron and cancer.

Early nutritional surveys demonstrated a significant increase in transferrin saturation (the fraction of transferrin in the blood that is bound to iron) in men who subsequently developed cancer relative to those who remained cancer-free2831. More recent meta-analyses that examine the association between biomarkers of iron status and risk of specific cancers indicate that the association between iron and cancer may be cancer-type specific: in a meta-analysis of 27 studies of women with breast cancer there was an increase in relative risk of breast cancer between highest and lowest levels of iron (RR 1.22; 95% CI 1.01–1.47)32. A meta-analysis of hepatocellular carcinoma similarly observed an association between both high serum ferritin and high serum iron and primary liver cancer risk (6 studies, HR 1.49, 95% CI 1.13–1.96 for ferritin; 3 studies, HR 2.47; 95% CI 1.31– 4.63 for serum iron)33, and approximately three-quarters of 33 studies of colorectal neoplasia supported an association between increased iron intake or increased iron stores with increased risk of colorectal neoplasia34. However a meta-analysis of 13 studies found no association between serum iron and lung cancer risk35. The association between dietary iron intake and cancer risk has also been examined using food frequency questionnaires, with similarly mixed results. For example, in a large study of over 500,000 participants with 8 years follow-up, individuals in the highest quintile of red meat intake exhibited a significantly increased risk of several malignancies when compared to those in the lowest quintile, including esophageal cancer, colorectal cancer, liver cancer and lung cancer36. However red meat intake was not associated with gastric cancer, bladder cancer, leukemia, lymphoma, melanoma, breast or prostate cancer36. Overall, the most consistent associations have been between consumption of red meat and colorectal cancer, although there has been a call for better-designed studies to more definitively test this association37.

There are both technical and biological reasons for the variance in the strength of the association between iron intake or biochemical indices of body iron and cancer. As examples, (1) assessments of iron intake are often derived from questionnaires that depend on recall which is sometimes incomplete or inaccurate; (2) there is a well-established variability in iron absorption after intake; and (3) single short periods of assessment are generally used to infer intake over a long period of time. In addition, although the association between ingestion of specific foods, such as red meat, with increased cancer risk has been attributed to the high iron (heme) content of red meat, it may also be due to other constituents of this complex food. Further, when blood tests are used to assess body iron stores in humans, the tests themselves are subject to factors independent of body iron content that affect results. For example, serum ferritin, a widely used marker of iron status in humans, can be markedly altered by inflammatory conditions that are independent of the subject’s iron status 38.

In addition to assessing the risk associated with increased stores or intake of iron, another approach to evaluate the connection between iron and cancer has been to examine the consequences of diseases that result in systemic iron overload in tissues such as the liver. As discussed above in Section 3, one such disease is hereditary hemochromatosis, which results in iron retention in the liver and other organs. Although hereditary hemochromatosis can result from at least five distinct genetic abnormalities, the most common mutation is in the HFE gene; patients with homozygous mutations have a 20–200 fold increased risk of liver cancer as well as increased cancer risk at other sites39,40. In an interesting example of gene/environment interaction, in certain African populations the risk of hepatocellular carcinoma and esophageal carcinoma may be exacerbated by the intake of home-brewed alcoholic drinks contaminated with iron41,42.

Another link between iron body stores and cancer risk comes from studies in which iron is depleted by blood donation (which reduces body iron by removing red blood cells, a major reservoir of iron) or increased by blood transfusion. A study involving examination of over 800,000 computerized files of Scandinavian individuals reported that cancer risk was increased in recipients of blood transfusions, with elevated incidence ratios of the transfused population versus the general population for cancers of the tongue, mouth, pharynx, esophagus, liver, and respiratory and urinary tracts and for squamous cell skin carcinoma43. However, the authors could not rule out the presence of undiagnosed occult cancers that may have produced symptoms necessitating the blood transfusion; possible immunosuppressive effects of transfusions (i.e. transfusion-related immunomodulation (TRIM)) may also have contributed to the observed elevation in cancer risk44. Conversely in a study of individuals who were frequent blood donors, cancer risk was significantly decreased (RR) of 0.79 (p <0.001) 45. A subsequent study of blood donors reported a more limited association: there was a significant trend of decreasing risk for cancers of the liver, lung, colon, stomach, and esophagus (P(trend) < .00146), but not other malignancies, between the lowest and highest categories of estimated iron loss from blood donation. Perhaps most compellingly, in a clinical study in which phlebotomy was performed to attempt to reduce peripheral arterial disease, phlebotomy decreased overall cancer risk and cancer-specific mortality47.

5. Mechanisms of dysregulation of iron in cancer

Once a cancer develops, there is substantial and growing evidence of increased metabolically available iron in the cancer cells themselves. This form of iron is frequently termed the labile iron pool, or LIP, and is believed to represent a small but biologically important fraction of total cellular iron48. In addition to the further acquisition of mutations, such excess iron, both in the form of labile iron and heme, supports cancer growth. How does the cancer cell acquire this additional iron?

Upregulation of transferrin receptor 1 (TFR1) is one way. Increases in TFR1, the receptor for transferrin-bound iron that is responsible for cellular iron uptake, have been repeatedly observed in numerous cancers, including breast, lung and bladder cancer, CLL, non-Hodgkin’s lymphoma, and others49. The increase in TFR1 can be directly oncogene driven; for example c-myc transcriptionally upregulates TFR1; alternatively, the increase in TFR1 can occur indirectly, for example as a function of FBXL5 reduction27 or IRP2 upregulation50,51. Another mechanism for increasing iron acquisition is upregulation of DMT1 in colorectal cancer52. CD44 is a cell surface protein whose expression is increased in cells undergoing the epithelial to mesenchymal transition (EMT) and is frequently used as a cancer stem cell marker. A recent study demonstrated that CD44 is responsible for glycosaminoglycan-mediated endocytosis of iron, suggesting not only that CD44 may play a role in iron acquisition in cancer stem cells, but directly tying iron to regulation of EMT53.

NGAL (lipocalin 2; gene name LCN2), is a 24 kd secreted glycoprotein and member of the lipocalin family of proteins that transport small hydrophobic ligands. NGAL binds iron complexed to a siderophore, and can be internalized by the cell surface receptor 24p3R. NGAL produced by tumor-associated macrophages can deliver iron and promote growth of tumor cells54. In addition, NGAL is upregulated in some cancers, and uptake of iron via this pathway has been linked to breast and renal cancer, among others55,56.

An additional mechanism by which cancer cells acquire excess iron is by inhibiting its efflux. In 2010, our laboratory reported that ferroportin (FPN), a cellular iron efflux pump, is downregulated in breast cancer57. We subsequently reported similar results in prostate58 and ovarian59 cancer; others have confirmed and expanded these results in other malignancies, including lung cancer60, multiple myeloma61 and a subset of patients with AML62. Multiple mechanisms for the decrease in ferroportin in cancer cells have been described, including upregulation of IRP250,51, which inhibits translation of FPN mRNA; upregulation of hepcidin, which triggers the degradation of FPN protein58, microRNAs such as miR-20a60 and miR-485–3p 63, and promoter methylation64.

Not all cancers use the same pathways to enhance iron acquisition and retention. Even within a single cancer, alternative mechanisms can be engaged. In work in breast cancer, for example, our laboratory showed that two distinct patterns of gene expression could result in the same phenotype of reduced patient survival: (1) Decreased iron export through reduction of ferroportin and overexpression of hepcidin; or (2) increased iron uptake through high expression of the transferrin receptor and reduction of HFE, a negative regulator of TFFR165. This work further demonstrated that an “iron regulatory gene signature” or IRGS, effectively predicts clinical outcome in a cohort of breast cancer patients treated with the same therapy. The IRGS includes ferroportin and transferrin receptor, as well as 14 additional genes of iron metabolism65. Clearly, the constellation of modifications in genes that govern iron metabolism in cancer involves much more than TFR1 and ferroportin. Systems biology approaches may be useful in parsing out these differences and identifying key signatures and drivers among them6668.

6. Iron addiction

There is evidence that the enhanced uptake and retention of iron in cancer constitutes a metabolic addiction – i.e., that cancer cells develop a dependence on iron that exceeds that of their non-malignant counterparts. Thus cancer cells are more sensitive to the anti-proliferative effects of a range of chemically distinct iron chelators, including desferoxamine, tachpyridine, the di-2-pyridylketone class of thiosemicarbazones (e.g. Dp44mT and DpC), than non-cancer cells69,70. Consistent with these findings, forced expression of ferroportin, which reduces intracellular iron by genetic rather than chemical means, is sufficient to reduce the rate of proliferation of breast, prostate and ovarian cancer cells57,59,71. Since the anti-proliferative effect of iron restriction on cancer cells tends to be greater than that on non-cancer cells59, we have termed this phenomenon “iron addiction”59.

7. Iron and metastasis

Although data are limited, there is evidence that iron may affect cancer metastasis as well as tumor growth. NDRG1, a metastasis suppressor that is downregulated in numerous cancers72, is upregulated following treatment with iron chelators73. Accordingly, treatment with the iron chelator Dp44mT induced NDRG1 and reduced formation of bone metastasis of human bone metastatic breast cancer cells (MDA-MB231-BoM) in intracardially injected mice72. Dp44mT also inhibited the formation of lung tumors in a metastatic model created by injection of osteosarcoma cells in nude mice74. Using induction of ferroportin to deplete iron, our group observed a decrease in the number of tumors in a mouse model of metastasis59. In this model, ovarian cancer cells were injected directly into the peritoneum (a major site of ovarian cancer metastases). Further, the FPN-mediated decrease in tumor number was associated with a decrease in invasion, IL6, and phosphorylated STAT359, a result consistent with results implicating activation of STAT3 in iron-dependent promotion of colorectal tumorigenesis52. Finally, the observation that expression of ferroportin and TFR1 predicts survival of breast cancer patients65 and that disrupted regulation of these pathways is associated with poorer survival of patients with prostate cancer58 implies a role for iron in human metastasis, since mortality from these cancers is primarily due to metastatic disease.

8. Iron and signaling in cancer

How does iron transmit a signal to induce tumor cell proliferation and metastasis? Our knowledge in this area is incomplete. However, it is clear that iron both regulates and is regulated by signaling pathways in cancer. Generally, the role of iron in tumor cell proliferation and metastasis has been linked to its function as a cofactor, required for the function of proteins essential to these processes, as well as to the ability of iron to impact signaling pathways directly. In turn, oncogenes, tumor suppressors, and growth factor and cytokine-mediated signaling pathways regulate critical proteins in iron metabolism. We provide a few important examples illustrating these principles.

8.1. Regulation of cell signaling by iron.

Iron is essential for the catalytic function of ribonucleotide reductase, the enzyme that converts ribonucleotides to deoxyribonucleotides, a critical step in DNA synthesis and therefore an obligate step in cell replication. Iron in the form of iron-sulfur clusters is also required for the activity of essentially all replicative DNA polymerases as well as proteins involved in genome maintenance and repair75. These include helicases such as FancJ and XPD; glycosylases involved in DNA repair; primase, Chir1 and DNA2, involved in DNA replication; RTEL1 with roles in homologous recombination, and others75. In addition, iron depletion by IRP2 knockdown or iron chelation induces the cell cycle regulators p15, p21 and p27, causing accumulation of cells in G0/G1, implying that iron is permissive for cell cycle progression.50 Mechanisms are still unclear; however, iron-dependent control of p21 by the transcription factor SP1 and its interacting partners ER-α and c-Jun in SK-MEL-28 melanoma cells has recently been described76, whereas in prostate cancer cells, p53 and KLF6 appear to play a role in iron-dependent regulation of p2171. By affecting these processes, iron directly impacts cell proliferation as well as DNA integrity.

Iron also influences the activity of the p53 tumor suppressor, arguably one of the most important barriers to tumor formation in humans. It was demonstrated that iron in the form of heme directly binds to p53, stimulating its degradation and interfering with its interaction with DNA, thus reducing p53’s anti-tumor effects77,78. However, it has also been reported that supplementation of cell cultures with exogenous inorganic iron reduces levels of MDM2, the ubiquitin ligase responsible for p53 degradation79,80, which would be expected to increase levels of p53, and in fact did so in one report80. The iron-mediated decrease in MDM2 was shown to occur by an IRP2-dependent mechanism, in which stabilization of MDM2 mRNA by IRP2 binding to its 3’ UTR was disrupted by excess iron79. It is possible that whether iron increases or decreases p53 depends on the form of iron (inorganic iron versus heme), the relative rates of these apparently competing pathways, or the particular cell types involved; however, this requires additional investigation. There is evidence that p53 in turn regulates iron: p53 translationally increases ferritin, reducing the availability of intracellular iron during p53-mediated cell cycle arrest81. More recent results suggest that this may occur through direct transcriptional induction of ISCU2 (iron sulfur cluster assembly enzyme) by p53. Upregulation of ISCU2 favors the formation of iron-sulfur clusters in IRP1, diminishing the activity of IRP1 as an RNA binding molecule (i.e. reducing its activity as a translational repressor of ferritin and stabilizer of TFR1 mRNA) and favoring its activity as a cytosolic aconitase82.

In addition to iron itself, proteins of iron metabolism may also act as signaling molecules. For example, ferritin exists both in intracellular and extracellular compartments38. The origin of extracellular serum ferritin has been unclear for decades, but it now appears that macrophages are at least one likely source of secreted ferritin 83,84 In conjunction with the unexpected observation that ferritin can induce NFKB85, a transcription factor that induces multiple tumor-promoting cytokines including IL6, this suggests a potential role for ferritin in sustaining a pro-tumorigenic microenvironment. Further, since NFKB can also induce ferritin86, the potential exists for the participation of ferritin in a tumor-promoting feed-forward loop. Such a model would be consistent with the link between elevated ferritin and poor outcome observed in some malignancies, such as AML87.

8.2. Regulation of iron by oncogenic signaling pathways.

Early evidence of a connection between oncogenic pathways and iron was provided by the observation that adenoviral E1A, a viral oncogene with immortalizing properties similar to those of c-myc, represses ferritin88. Subsequently, it was demonstrated that the c-myc oncogene directly and coordinately represses transcription of ferritin while transcriptionally upregulating IRP2 in EBV-immortalized B cells89. These effects increased intracellular iron and were required for cell transformation by c-myc. Consistent with these results, c-myc was later shown to activate TFR1 to enhance proliferation and tumorigenesis in in vitro and in vivo models of B cell lymphoma90.

WNT and STAT3 signaling have both been implicated in iron signaling in colorectal cancer. Iron loading increased WNT signaling in colorectal cells with mutant APC91, providing a mechanistic underpinning for the subsequent observation that iron exacerbates tumorigenesis in the background of APC loss23. The iron dependence of this pathway was also revealed by a WNT inhibitor screen92. Separately, iron-mediated induction of JAK1-STAT3 signaling by CDK1 was recently shown to promote colorectal tumorigenesis52. Whether these pathways are differentially triggered in tumors from different individuals and/or are the result of different upstream activation events remains to be determined.

Hepcidin is a peptide hormone produced by the liver that is a master regulator of body iron homeostasis. Cancer cells also produce hepcidin, which binds to ferroportin on the surface of cancer cells and thereby downregulates iron efflux, acting as a mechanism of iron retention57. Multiple signaling pathways impinge on the synthesis of hepcidin in cancer. These include BMPs and downstream SMADs, as well as the JAK/STAT3 pathway, which controls hepcidin synthesis in response to inflammatory cytokines. Our laboratory recently demonstrated that hepcidin regulatory pathways are also influenced by the microenvironment: in breast cancer cells grown as spheroids in three-dimensional culture, GDF15 emerged as a potent regulator of hepcidin. Further, inclusion of tumor-associated fibroblasts in the spheroids potentiated hepcidin synthesis through secretion of IL693. These results suggest that it will be necessary to construct new paradigms for tumor-mediated control of iron that takes into account tumor architecture and the tumor microenvironment.

9. Iron: therapeutic approaches

Observations of iron dysregulation in cancer have given rise to several strategies to target this metabolic vulnerability for cancer therapy. These approaches can loosely be grouped into four categories: (1) depleting iron, based on the concept that iron is a metabolite upon which cancers are particularly dependent; (2) using iron mimetics to disrupt iron metabolism; (3) using proteins of iron metabolism that are over-expressed in cancer as targets for the delivery of anti-cancer agents; (4) providing additional iron, based on the concept that the excess iron present in cancer cells renders them oxidatively fragile and increases their susceptibility to iron-mediated oxidative stress.

9.1. Strategies to deplete cancer cells of iron.

This is historically one of the earliest approaches to leverage iron for cancer therapy. Soon after the discovery of transferrin receptor as the mechanism of iron delivery into cells and the demonstration that this receptor is upregulated in multiple cancers, anti-TfR antibodies were developed and shown to inhibit tumor growth94. Anti-TfR1 antibodies administered in a phase I clinical trial exhibited some activity, particularly in hematopoetic malignancies, and modest systemic toxicity95, encouraging the search for humanized antibodies with improved activity96,97.

Several different small molecule iron chelators used in the clinical treatment of iron overload, such as desferrioxamine and more recently deferiprone98,99 and deferasirox100, were shown to preferentially reduce cancer cell growth in cell culture and reduce tumor burden in animal models when administered either alone or in combination with other anti-cancer drugs. A few small clinical trials, mostly of desferrioxamine, produced encouraging but limited responses101,102.

9.2. Using iron mimetics to disrupt iron metabolism.

Gallium(III) shares certain chemical characteristics with iron(III) that enables it to interact with iron-binding proteins and disrupt iron-dependent tumor cell growth103. This includes inhibition of ribonucleotide reductase and mitochondrial function. Newer gallium ligands such as Tris(8-quinolinolato)gallium(III) (KP46) and gallium maltolate have replaced simple gallium salts such as gallium nitrate, and are undergoing clinical evaluation.

9.3. Exploiting proteins of iron metabolism for targeted delivery.

This strategy has focused largely on TFR1, a cell surface receptor frequently over-expressed in cancer cells that can be co-opted to serve as a delivery vehicle for anti-cancer drugs. Examples include transferrin conjugated to chemotherapeutic drugs such as doxorubicin and cisplatin, toxins such as diptheria toxin and ricin, and anti-neoplastic nucleic acids49. Toxicity was an issue in early trials, but the development of improved antibodies holds promise, and this is still an area of active investigation96,104.

9.4. Leveraging iron to generate cytotoxic oxidative stress in cancer cells.

Although iron chelators are generally used to deplete iron, iron chelators of the appropriate chemical structure3 can also bind iron (and copper) in a fashion that promotes redox cycling and consequently the generation of toxic free radicals. Examples of iron chelators of this class, such as the thiosemicarbazones DpC and Dp44mT, have shown substantial promise as anti-cancer and anti-metastatic agents in preclinical studies and progressed to evaluation in clinical trials105. In selected cancer cells, iron itself may have pro-oxidant cytotoxic effects: a recent finding that leukemic cells from a subset of patients with AML exhibit low levels of ferroportin led to the observation that ferumoxytol (Feraheme), an FDA-approved iron oxide nanoparticle used in treatment of iron deficiency, can induce oxidative stress and reduce tumor burden in a murine leukemia model62.

Finally, ferroptosis is a recently-described mechanism of cell death notable for its iron-dependence106. Ferroptosis is triggered by an accumulation of peroxidized membrane phospholipids, which distinguishes ferroptosis mechanistically from other forms of regulated cell death. Since the formation of lipid radicals is fostered by iron, cancer cells with excess iron may be particularly susceptible to drugs that induce ferroptosis. In line with this prediction, we observed that ovarian cancer stem cells could be effectively targeted in vitro and in vivo by erastin, a preclinical drug that induces ferroptosis59. Salinomycin, a drug with efficacy against cancer stem cells, may also act by inducing ferroptosis: salinomycin sequesters lysosomal iron, resulting in an iron depletion response that triggers lysosomal degradation of ferritin, production of ROS, and a death pathway resembling ferroptosis107. Conversely, prominin2 protects cells from ferroptosis by stimulating efflux of ferritin and its associated iron in endosomes and multivesicular bodies108. The ongoing effort to discover new ferroptosis-inducing agents to treat cancer109, and the entry of these agents into early stage clinical trials in cancer patients (ClinicalTrials.gov NCT01695590), suggests that this may be a promising venue for further study.

Figure:

Figure:

Examples of iron trafficking proteins altered in cancer. Small red circles represent iron. Proteins colored in orange are increased in most cancer cells when compared to non-malignant cells; proteins colored in blue are decreased. Upregulation of proteins involved in iron import, such as TFR1, DMT1 and 24p3R increase the labile iron pool; downregulation of ferritin shifts more iron to the labile iron pool; reduction of ferroportin increases iron retention by decreasing iron efflux. Hepcidin degrades ferroportin – more hepcidin is associated with decreased iron efflux. Upregulation of IRP2 is one of several mechanisms underlying these changes: IRP2 stabilizes TFR1 mRNA by binding to the 3’UTR of its mRNA; IRP2 also binds to the 5’ UTR of the ferritin and ferroportin mRNA to repress their translation. (IRP1 also functions as a translational regulator of TFR1, ferritin, and ferroportin; however it does not appear to exert a similar pro-tumorigenic effect). Ferritin consists of H and L subunits that are similarly regulated by IRP2; for simplicity these subunits are both represented as “ferritin”. Note that these changes are not observed in all cancers and are not necessarily present simultaneously.

Acknowledgements:

We thank all laboratory members, past and present, who contributed to the work from our laboratories cited in this review.

Funding:

this work was supported by National Cancer Institute grant numbers R01CA188025 (SVT) and R01CA233636 (FMT).

References

  • 1.Cronin SJF, Woolf CJ, Weiss G, Penninger JM. The Role of Iron Regulation in Immunometabolism and Immune-Related Disease. Front Mol Biosci. 2019;6:116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Fiorito V, Chiabrando D, Petrillo S, Bertino F, Tolosano E. The Multifaceted Role of Heme in Cancer. Front Oncol. 2019;9:1540. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Koppenol WH, Hider RH. Iron and redox cycling. Do’s and don’ts. Free Radic Biol Med. 2019;133:3–10. [DOI] [PubMed] [Google Scholar]
  • 4.Inoue S, Kawanishi S. Hydroxyl radical production and human DNA damage induced by ferric nitrilotriacetate and hydrogen peroxide. Cancer Res. 1987;47(24 Pt 1):6522–6527. [PubMed] [Google Scholar]
  • 5.Nackerdien Z, Rao G, Cacciuttolo MA, Gajewski E, Dizdaroglu M. Chemical nature of DNA-protein cross-links produced in mammalian chromatin by hydrogen peroxide in the presence of iron or copper ions. Biochemistry. 1991;30(20):4873–4879. [DOI] [PubMed] [Google Scholar]
  • 6.Dizdaroglu M, Nackerdien Z, Chao BC, Gajewski E, Rao G. Chemical nature of in vivo DNA base damage in hydrogen peroxide-treated mammalian cells. Arch Biochem Biophys. 1991;285(2):388–390. [DOI] [PubMed] [Google Scholar]
  • 7.Dizdaroglu M, Rao G, Halliwell B, Gajewski E. Damage to the DNA bases in mammalian chromatin by hydrogen peroxide in the presence of ferric and cupric ions. Arch Biochem Biophys. 1991;285(2):317–324. [DOI] [PubMed] [Google Scholar]
  • 8.Dizdaroglu M, Jaruga P. Mechanisms of free radical-induced damage to DNA. Free Radic Res. 2012;46(4):382–419. [DOI] [PubMed] [Google Scholar]
  • 9.Linn S DNA damage by iron and hydrogen peroxide in vitro and in vivo. Drug Metab Rev. 1998;30(2):313–326. [DOI] [PubMed] [Google Scholar]
  • 10.Jabbour E, Kantarjian H. Chronic myeloid leukemia: 2018 update on diagnosis, therapy and monitoring. Am J Hematol. 2018;93(3):442–459. [DOI] [PubMed] [Google Scholar]
  • 11.Campbell JA. Effects of Precipitated Silica and of Iron Oxide on the Incidence of Primary Lung Tumours in Mice. Br Med J. 1940;2(4156):275–280. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Richmond HG. Induction of sarcoma in the rat by iron-dextran complex. Br Med J. 1959;1(5127):947–949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Ebina Y, Okada S, Hamazaki S, Ogino F, Li JL, Midorikawa O. Nephrotoxicity and renal cell carcinoma after use of iron- and aluminum-nitrilotriacetate complexes in rats. J Natl Cancer Inst. 1986;76(1):107–113. [PubMed] [Google Scholar]
  • 14.Akatsuka S, Yamashita Y, Ohara H, et al. Fenton reaction induced cancer in wild type rats recapitulates genomic alterations observed in human cancer. PLoS One. 2012;7(8):e43403. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Akatsuka S, Li GH, Toyokuni S. Superiority of rat over murine model for studies on the evolution of cancer genome. Free Radic Res. 2018;52(11–12):1323–1327. [DOI] [PubMed] [Google Scholar]
  • 16.Niederau C, Fischer R, Sonnenberg A, Stremmel W, Trampisch HJ, Strohmeyer G. Survival and causes of death in cirrhotic and in noncirrhotic patients with primary hemochromatosis. N Engl J Med. 1985;313(20):1256–1262. [DOI] [PubMed] [Google Scholar]
  • 17.Hussain SP, Raja K, Amstad PA, et al. Increased p53 mutation load in nontumorous human liver of wilson disease and hemochromatosis: oxyradical overload diseases. Proc Natl Acad Sci U S A. 2000;97(23):12770–12775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lehmann U, Wingen LU, Brakensiek K, et al. Epigenetic defects of hepatocellular carcinoma are already found in non-neoplastic liver cells from patients with hereditary haemochromatosis. Hum Mol Genet. 2007;16(11):1335–1342. [DOI] [PubMed] [Google Scholar]
  • 19.Hann HW, Stahlhut MW, Menduke H. Iron enhances tumor growth. Observation on spontaneous mammary tumors in mice. Cancer. 1991;68(11):2407–2410. [DOI] [PubMed] [Google Scholar]
  • 20.Thompson HJ, Kennedy K, Witt M, Juzefyk J. Effect of dietary iron deficiency or excess on the induction of mammary carcinogenesis by 1-methyl-1-nitrosourea. Carcinogenesis. 1991;12(1):111–114. [DOI] [PubMed] [Google Scholar]
  • 21.Singh M, Lu J, Briggs SP, McGinley JN, Haegele AD, Thompson HJ. Effect of excess dietary iron on the promotion stage of 1-methyl-1-nitrosourea-induced mammary carcinogenesis: pathogenetic characteristics and distribution of iron. Carcinogenesis. 1994;15(8):1567–1570. [DOI] [PubMed] [Google Scholar]
  • 22.Diwan BA, Kasprzak KS, Anderson LM. Promotion of dimethylbenz[a]anthracene-initiated mammary carcinogenesis by iron in female Sprague-Dawley rats. Carcinogenesis. 1997;18(9):1757–1762. [DOI] [PubMed] [Google Scholar]
  • 23.Radulescu S, Brookes MJ, Salgueiro P, et al. Luminal iron levels govern intestinal tumorigenesis after Apc loss in vivo. Cell Rep. 2012;2(2):270–282. [DOI] [PubMed] [Google Scholar]
  • 24.Hann HW, Stahlhut MW, Blumberg BS. Iron nutrition and tumor growth: decreased tumor growth in iron-deficient mice. Cancer Res. 1988;48(15):4168–4170. [PubMed] [Google Scholar]
  • 25.Hann HW, Stahlhut MW, Rubin R, Maddrey WC. Antitumor effect of deferoxamine on human hepatocellular carcinoma growing in athymic nude mice. Cancer. 1992;70(8):2051–2056. [DOI] [PubMed] [Google Scholar]
  • 26.Muto Y, Moroishi T, Ichihara K, et al. Disruption of FBXL5-mediated cellular iron homeostasis promotes liver carcinogenesis. J Exp Med. 2019;216(4):950–965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Moroishi T, Nishiyama M, Takeda Y, Iwai K, Nakayama KI. The FBXL5-IRP2 axis is integral to control of iron metabolism in vivo. Cell Metab. 2011;14(3):339–351. [DOI] [PubMed] [Google Scholar]
  • 28.Stevens RG, Graubard BI, Micozzi MS, Neriishi K, Blumberg BS. Moderate elevation of body iron level and increased risk of cancer occurrence and death. Int J Cancer. 1994;56(3):364–369. [DOI] [PubMed] [Google Scholar]
  • 29.Stevens RG, Jones DY, Micozzi MS, Taylor PR. Body iron stores and the risk of cancer. N Engl J Med. 1988;319(16):1047–1052. [DOI] [PubMed] [Google Scholar]
  • 30.Knekt P, Reunanen A, Takkunen H, Aromaa A, Heliovaara M, Hakulinen T. Body iron stores and risk of cancer. Int J Cancer. 1994;56(3):379–382. [DOI] [PubMed] [Google Scholar]
  • 31.Wu T, Sempos CT, Freudenheim JL, Muti P, Smit E. Serum iron, copper and zinc concentrations and risk of cancer mortality in US adults. Ann Epidemiol. 2004;14(3):195–201. [DOI] [PubMed] [Google Scholar]
  • 32.Chang VC, Cotterchio M, Khoo E. Iron intake, body iron status, and risk of breast cancer: a systematic review and meta-analysis. BMC Cancer. 2019;19(1):543. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Tran KT, Coleman HG, McCain RS, Cardwell CR. Serum Biomarkers of Iron Status and Risk of Primary Liver Cancer: A Systematic Review and Meta-Analysis. Nutr Cancer. 2019;71(8):1365–1373. [DOI] [PubMed] [Google Scholar]
  • 34.Nelson RL. Iron and colorectal cancer risk: human studies. Nutr Rev. 2001;59(5):140–148. [DOI] [PubMed] [Google Scholar]
  • 35.Chen HF, Wu LX, Li XF, et al. A meta-analysis of association between serum iron levels and lung cancer risk. Cell Mol Biol (Noisy-le-grand). 2018;64(13):33–37. [PubMed] [Google Scholar]
  • 36.Cross AJ, Leitzmann MF, Gail MH, Hollenbeck AR, Schatzkin A, Sinha R. A prospective study of red and processed meat intake in relation to cancer risk. PLoS Med. 2007;4(12):e325. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Turner ND, Lloyd SK. Association between red meat consumption and colon cancer: A systematic review of experimental results. Exp Biol Med (Maywood). 2017;242(8):813–839. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Wang W, Knovich MA, Coffman LG, Torti FM, Torti SV. Serum ferritin: Past, present and future. Biochim Biophys Acta. 2010;1800(8):760–769. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Elmberg M, Hultcrantz R, Ekbom A, et al. Cancer risk in patients with hereditary hemochromatosis and in their first-degree relatives. Gastroenterology. 2003;125(6):1733–1741. [DOI] [PubMed] [Google Scholar]
  • 40.Fargion S, Valenti L, Fracanzani AL. Hemochromatosis gene (HFE) mutations and cancer risk: expanding the clinical manifestations of hereditary iron overload. Hepatology. 2010;51(4):1119–1121. [DOI] [PubMed] [Google Scholar]
  • 41.Isaacson C, Bothwell TH, MacPhail AP, Simon M. The iron status of urban black subjects with carcinoma of the oesophagus. S Afr Med J. 1985;67(15):591–593. [PubMed] [Google Scholar]
  • 42.Kew MC. Hepatic iron overload and hepatocellular carcinoma. Liver Cancer. 2014;3(1):31–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Hjalgrim H, Edgren G, Rostgaard K, et al. Cancer incidence in blood transfusion recipients. J Natl Cancer Inst. 2007;99(24):1864–1874. [DOI] [PubMed] [Google Scholar]
  • 44.Youssef LA, Spitalnik SL. Transfusion-related immunomodulation: a reappraisal. Curr Opin Hematol. 2017;24(6):551–557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Merk K, Mattsson B, Mattsson A, Holm G, Gullbring B, Bjorkholm M. The incidence of cancer among blood donors. Int J Epidemiol. 1990;19(3):505–509. [DOI] [PubMed] [Google Scholar]
  • 46.Edgren G, Reilly M, Hjalgrim H, et al. Donation frequency, iron loss, and risk of cancer among blood donors. J Natl Cancer Inst. 2008;100(8):572–579. [DOI] [PubMed] [Google Scholar]
  • 47.Zacharski LR, Chow BK, Howes PS, et al. Decreased cancer risk after iron reduction in patients with peripheral arterial disease: results from a randomized trial. J Natl Cancer Inst. 2008;100(14):996–1002. [DOI] [PubMed] [Google Scholar]
  • 48.Kakhlon O, Cabantchik ZI. The labile iron pool: characterization, measurement, and participation in cellular processes(1). Free Radic Biol Med. 2002;33(8):1037–1046. [DOI] [PubMed] [Google Scholar]
  • 49.Daniels TR, Bernabeu E, Rodriguez JA, et al. The transferrin receptor and the targeted delivery of therapeutic agents against cancer. Biochim Biophys Acta. 2012;1820(3):291–317. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Deng Z, Manz DH, Torti SV, Torti FM. Iron-responsive element-binding protein 2 plays an essential role in regulating prostate cancer cell growth. Oncotarget. 2017;8(47):82231–82243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Wang W, Deng Z, Hatcher H, et al. IRP2 regulates breast tumor growth. Cancer Res. 2014;74(2):497–507. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Xue X, Ramakrishnan SK, Weisz K, et al. Iron Uptake via DMT1 Integrates Cell Cycle with JAK-STAT3 Signaling to Promote Colorectal Tumorigenesis. Cell Metab. 2016;24(3):447–461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Muller S SF, Caneque T, Lafon A, Versini A, Wu T, Lombard B, Loew D, Durand A, Vallot C, Bauland S, Servant N, Rodriguez R. CD44 regulates epigenetic plasticity by mediating iron endocytosis. bioRxiv. 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Duan X, He K, Li J, et al. Tumor associated macrophages deliver iron to tumor cells via Lcn2. Int J Physiol Pathophysiol Pharmacol. 2018;10(2):105–114. [PMC free article] [PubMed] [Google Scholar]
  • 55.Chakraborty S, Kaur S, Guha S, Batra SK. The multifaceted roles of neutrophil gelatinase associated lipocalin (NGAL) in inflammation and cancer. Biochim Biophys Acta. 2012;1826(1):129–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Rehwald C, Schnetz M, Urbschat A, et al. The iron load of lipocalin-2 (LCN-2) defines its pro-tumour function in clear-cell renal cell carcinoma. Br J Cancer. 2020;122(3):421–433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Pinnix ZK, Miller LD, Wang W, et al. Ferroportin and iron regulation in breast cancer progression and prognosis. Sci Transl Med. 2010;2(43):43ra56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Tesfay L, Clausen KA, Kim JW, et al. Hepcidin regulation in prostate and its disruption in prostate cancer. Cancer Res. 2015;75(11):2254–2263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Basuli D, Tesfay L, Deng Z, et al. Iron addiction: a novel therapeutic target in ovarian cancer. Oncogene. 2017;36(29):4089–4099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Babu KR, Muckenthaler MU. miR-20a regulates expression of the iron exporter ferroportin in lung cancer. J Mol Med (Berl). 2016;94(3):347–359. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Kong Y, Hu L, Lu K, et al. Ferroportin downregulation promotes cell proliferation by modulating the Nrf2-miR-17–5p axis in multiple myeloma. Cell Death Dis. 2019;10(9):624. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Trujillo-Alonso V, Pratt EC, Zong H, et al. FDA-approved ferumoxytol displays anti-leukaemia efficacy against cells with low ferroportin levels. Nat Nanotechnol. 2019;14(6):616–622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Sangokoya C, Doss JF, Chi JT. Iron-responsive miR-485–3p regulates cellular iron homeostasis by targeting ferroportin. PLoS Genet. 2013;9(4):e1003408. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Chen Y, Zhang S, Wang X, et al. Disordered signaling governing ferroportin transcription favors breast cancer growth. Cell Signal. 2015;27(1):168–176. [DOI] [PubMed] [Google Scholar]
  • 65.Miller LD, Coffman LG, Chou JW, et al. An iron regulatory gene signature predicts outcome in breast cancer. Cancer Res. 2011;71(21):6728–6737. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Konstorum A, Lynch ML, Torti SV, Torti FM, Laubenbacher RC. A Systems Biology Approach to Understanding the Pathophysiology of High-Grade Serous Ovarian Cancer: Focus on Iron and Fatty Acid Metabolism. OMICS. 2018;22(7):502–513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Chifman J, Arat S, Deng Z, et al. Activated Oncogenic Pathway Modifies Iron Network in Breast Epithelial Cells: A Dynamic Modeling Perspective. PLoS Comput Biol. 2017;13(2):e1005352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Chifman J, Laubenbacher R, Torti SV. A systems biology approach to iron metabolism. Adv Exp Med Biol. 2014;844:201–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Abeysinghe RD, Greene BT, Haynes R, et al. p53-independent apoptosis mediated by tachpyridine, an anti-cancer iron chelator. Carcinogenesis. 2001;22(10):1607–1614. [DOI] [PubMed] [Google Scholar]
  • 70.Yuan J, Lovejoy DB, Richardson DR. Novel di-2-pyridyl-derived iron chelators with marked and selective antitumor activity: in vitro and in vivo assessment. Blood. 2004;104(5):1450–1458. [DOI] [PubMed] [Google Scholar]
  • 71.Deng Z, Manz DH, Torti SV, Torti FM. Effects of Ferroportin-Mediated Iron Depletion in Cells Representative of Different Histological Subtypes of Prostate Cancer. Antioxid Redox Signal. 2019;30(8):1043–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Liu W, Xing F, Iiizumi-Gairani M, et al. N-myc downstream regulated gene 1 modulates Wnt-beta-catenin signalling and pleiotropically suppresses metastasis. EMBO Mol Med. 2012;4(2):93–108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Le NT, Richardson DR. Iron chelators with high antiproliferative activity up-regulate the expression of a growth inhibitory and metastasis suppressor gene: a link between iron metabolism and proliferation. Blood. 2004;104(9):2967–2975. [DOI] [PubMed] [Google Scholar]
  • 74.Li P, Zheng X, Shou K, et al. The iron chelator Dp44mT suppresses osteosarcoma’s proliferation, invasion and migration: in vitro and in vivo. Am J Transl Res. 2016;8(12):5370–5385. [PMC free article] [PubMed] [Google Scholar]
  • 75.Fuss JO, Tsai CL, Ishida JP, Tainer JA. Emerging critical roles of Fe-S clusters in DNA replication and repair. Biochim Biophys Acta. 2015;1853(6):1253–1271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Moussa RS, Kovacevic Z, Bae DH, Lane DJR, Richardson DR. Transcriptional regulation of the cyclin-dependent kinase inhibitor, p21(CIP1/WAF1), by the chelator, Dp44mT. Biochim Biophys Acta Gen Subj. 2018;1862(3):761–774. [DOI] [PubMed] [Google Scholar]
  • 77.Shen J, Sheng X, Chang Z, et al. Iron metabolism regulates p53 signaling through direct heme-p53 interaction and modulation of p53 localization, stability, and function. Cell Rep. 2014;7(1):180–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Shen J, Sheng X, Chang Z, et al. The heme-p53 interaction: Linking iron metabolism to p53 signaling and tumorigenesis. Mol Cell Oncol. 2016;3(1):e965642. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Zhang J, Kong X, Zhang Y, Sun W, Xu E, Chen X. Mdm2 is a target and mediator of IRP2 in cell growth control. FASEB J. 2020;34(2):2301–2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Dongiovanni P, Fracanzani AL, Cairo G, et al. Iron-dependent regulation of MDM2 influences p53 activity and hepatic carcinogenesis. Am J Pathol. 2010;176(2):1006–1017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhang F, Wang W, Tsuji Y, Torti SV, Torti FM. Post-transcriptional modulation of iron homeostasis during p53-dependent growth arrest. J Biol Chem. 2008;283(49):33911–33918. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Funauchi Y, Tanikawa C, Yi Lo PH, et al. Regulation of iron homeostasis by the p53-ISCU pathway. Sci Rep. 2015;5:16497. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Cohen LA, Gutierrez L, Weiss A, et al. Serum ferritin is derived primarily from macrophages through a nonclassical secretory pathway. Blood. 2010;116(9):1574–1584. [DOI] [PubMed] [Google Scholar]
  • 84.Truman-Rosentsvit M, Berenbaum D, Spektor L, et al. Ferritin is secreted via 2 distinct nonclassical vesicular pathways. Blood. 2018;131(3):342–352. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ruddell RG, Hoang-Le D, Barwood JM, et al. Ferritin functions as a proinflammatory cytokine via iron-independent protein kinase C zeta/nuclear factor kappaB-regulated signaling in rat hepatic stellate cells. Hepatology. 2009;49(3):887–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Kwak EL, Larochelle DA, Beaumont C, Torti SV, Torti FM. Role for NF-kappa B in the regulation of ferritin H by tumor necrosis factor-alpha. J Biol Chem. 1995;270(25):15285–15293. [DOI] [PubMed] [Google Scholar]
  • 87.Bertoli S, Paubelle E, Berard E, et al. Ferritin heavy/light chain (FTH1/FTL) expression, serum ferritin levels, and their functional as well as prognostic roles in acute myeloid leukemia. Eur J Haematol. 2019;102(2):131–142. [DOI] [PubMed] [Google Scholar]
  • 88.Tsuji Y, Kwak E, Saika T, Torti SV, Torti FM. Preferential repression of the H subunit of ferritin by adenovirus E1A in NIH-3T3 mouse fibroblasts. J Biol Chem. 1993;268(10):7270–7275. [PubMed] [Google Scholar]
  • 89.Wu KJ, Polack A, Dalla-Favera R. Coordinated regulation of iron-controlling genes, H-ferritin and IRP2, by c-MYC. Science. 1999;283(5402):676–679. [DOI] [PubMed] [Google Scholar]
  • 90.O’Donnell KA, Yu D, Zeller KI, et al. Activation of transferrin receptor 1 by c-Myc enhances cellular proliferation and tumorigenesis. Mol Cell Biol. 2006;26(6):2373–2386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Brookes MJ, Boult J, Roberts K, et al. A role for iron in Wnt signalling. Oncogene. 2008;27(7):966–975. [DOI] [PubMed] [Google Scholar]
  • 92.Song S, Christova T, Perusini S, et al. Wnt inhibitor screen reveals iron dependence of beta-catenin signaling in cancers. Cancer Res. 2011;71(24):7628–7639. [DOI] [PubMed] [Google Scholar]
  • 93.Blanchette-Farra N, Kita D, Konstorum A, et al. Contribution of three-dimensional architecture and tumor-associated fibroblasts to hepcidin regulation in breast cancer. Oncogene. 2018;37(29):4013–4032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Trowbridge IS, Lopez F. Monoclonal antibody to transferrin receptor blocks transferrin binding and inhibits human tumor cell growth in vitro. Proc Natl Acad Sci U S A. 1982;79(4):1175–1179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Brooks D, Taylor C, Dos Santos B, et al. Phase Ia trial of murine immunoglobulin A antitransferrin receptor antibody 42/6. Clin Cancer Res. 1995;1(11):1259–1265. [PubMed] [Google Scholar]
  • 96.Daniels-Wells TR, Penichet ML. Transferrin receptor 1: a target for antibody-mediated cancer therapy. Immunotherapy. 2016;8(9):991–994. [DOI] [PubMed] [Google Scholar]
  • 97.Daniels-Wells TR, Candelaria PV, Leoh LS, Nava M, Martinez-Maza O, Penichet ML. An IgG1 Version of the Anti-transferrin Receptor 1 Antibody ch128.1 Shows Significant Antitumor Activity Against Different Xenograft Models of Multiple Myeloma: A Brief Communication. J Immunother. 2020;43(2):48–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kast RE, Skuli N, Sardi I, et al. Augmentation of 5-Aminolevulinic Acid Treatment of Glioblastoma by Adding Ciprofloxacin, Deferiprone, 5-Fluorouracil and Febuxostat: The CAALA Regimen. Brain Sci. 2018;8(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Simoes RV, Veeraperumal S, Serganova IS, et al. Inhibition of prostate cancer proliferation by Deferiprone. NMR Biomed. 2017;30(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Bedford MR, Ford SJ, Horniblow RD, Iqbal TH, Tselepis C. Iron chelation in the treatment of cancer: a new role for deferasirox? J Clin Pharmacol. 2013;53(9):885–891. [DOI] [PubMed] [Google Scholar]
  • 101.Donfrancesco A, Deb G, Dominici C, Pileggi D, Castello MA, Helson L. Effects of a single course of deferoxamine in neuroblastoma patients. Cancer Res. 1990;50(16):4929–4930. [PubMed] [Google Scholar]
  • 102.Yamasaki T, Terai S, Sakaida I. Deferoxamine for advanced hepatocellular carcinoma. N Engl J Med. 2011;365(6):576–578. [DOI] [PubMed] [Google Scholar]
  • 103.Chitambar CR. The therapeutic potential of iron-targeting gallium compounds in human disease: From basic research to clinical application. Pharmacol Res. 2017;115:56–64. [DOI] [PubMed] [Google Scholar]
  • 104.Kim SS, Harford JB, Moghe M, Rait A, Pirollo KF, Chang EH. Targeted nanocomplex carrying siRNA against MALAT1 sensitizes glioblastoma to temozolomide. Nucleic Acids Res. 2018;46(3):1424–1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Merlot AM, Kalinowski DS, Kovacevic Z, et al. Exploiting Cancer Metal Metabolism using Anti-Cancer Metal- Binding Agents. Curr Med Chem. 2019;26(2):302–322. [DOI] [PubMed] [Google Scholar]
  • 106.Stockwell BR, Friedmann Angeli JP, Bayir H, et al. Ferroptosis: A Regulated Cell Death Nexus Linking Metabolism, Redox Biology, and Disease. Cell. 2017;171(2):273–285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Mai TT, Hamai A, Hienzsch A, et al. Salinomycin kills cancer stem cells by sequestering iron in lysosomes. Nat Chem. 2017;9(10):1025–1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Brown CW, Amante JJ, Chhoy P, et al. Prominin2 Drives Ferroptosis Resistance by Stimulating Iron Export. Dev Cell. 2019;51(5):575–586 e574. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lu B, Chen XB, Ying MD, He QJ, Cao J, Yang B. The Role of Ferroptosis in Cancer Development and Treatment Response. Front Pharmacol. 2017;8:992. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES