Summary
Scientists in this field often joke, “If you don’t have a mechanism, say it’s ROS.” Seemingly connected to every biological process ever described, ROS has numerous pleiotropic roles in physiology and disease. In some contexts, ROS act as secondary messengers, controlling a variety of signaling cascades. In other scenarios, they initiate damage to macromolecules. Finally, in their worst form, ROS is deadly to cells and surrounding tissues. A set of molecules with detoxifying abilities, termed antioxidants, are the direct counterpart to ROS. Notably, antioxidants exist in the public domain, touted as a “cure-all” for diseases. Research has disproved many of these claims and, in some cases, shown the opposite. Of all the diseases, cancer stands out in its paradoxical relationship with antioxidants. While the field has made numerous strides in understanding the roles of antioxidants in cancer, many questions remain.
eTORC blurb
The paradoxical role of antioxidants in cancer has been a matter of intense debate in recent years. Here, we highlight the latest breakthroughs that shed light on the molecular mechanisms by which antioxidants are regulated in cancer to impact their initiation, progression, and metastatic capabilities.
Introduction
Antioxidants can be broadly defined as “substances that delay, prevent, or remove oxidative damage to specific target molecules by reacting with oxidants,” such as reactive oxygen species (ROS)1. Antioxidants and oxidants in equilibrium maintain redox homeostasis, and an imbalance in favor of oxidants can lead to oxidative stress, which has been linked to the pathogenesis of a myriad of diseases, including neurodegenerative and cardiovascular disorders, diabetes, and cancer2. Cancer tissues are believed to produce higher levels of ROS due to their altered metabolism, inflammation, hypoxic environment, and oncogene-driven upregulation of ROS-generating enzymes3, thus, suggesting the need for antioxidants to buffer excessive ROS. Here, we will discuss pertinent developments and knowledge gaps in the regulation of antioxidants in cancer. We will only minimally discuss, however, topics surrounding ferroptosis4–6 and ROS signaling7,8, for which details can be found in several excellent reviews.
Rather than prevent, antioxidant supplementation promotes cancer
Unchecked oxidative stress can cause DNA damage, mutations, and oncogenesis. However, an oversimplistic interpretation of these facts has led to the prevalent misconception that ROS are intrinsically deleterious while antioxidants would have beneficial effects on almost every aspect of human health. This premise potentially contributed to the widespread consumption of complementary and alternative medicine products (CAM), vitamins, minerals, and antioxidant dietary supplements, often without the knowledge of their physicians9, to prevent cancer, improve prognosis, or alleviate side effects of radio- and chemotherapy. The reported prevalence of supplement use varies considerably depending on the cancer type and patients’ demographics but is frequently reported to be higher than 50%10, and is positively associated with age, higher education levels, and female sex9–11. While at first some observational studies hinted at a possible benefit, large randomized controlled clinical trials conducted in the ‘90s and 2000s have not only consistently failed to demonstrate clinical benefits but also revealed that some antioxidants increased all-cause mortality12–21. Considering new evidence, recommendations against the use of antioxidants were published22. This subject has been revisited recently, with a new recommendation statement published by the US Preventive Services Task Force recommending against the use of β-carotene or vitamin E supplements for the prevention of cancer or cardiovascular disease23,24. Of note, the absence of positive outcomes in clinical studies could hypothetically be attributed to differences in the impact of the tested antioxidants when consumed as isolated chemicals versus when they are ingested through food, which contain intricate combinations of antioxidants, vitamins, and minerals. Thus, if antioxidants have any anti-tumor activity, we have not yet discovered when or how to dose, combine, and deliver these molecules to achieve clinical benefit. Certain antioxidants act as “pro-oxidants” at elevated levels (i.e., Vitamin C)25,26. Additionally, elucidation of the key pathways involved in the overabundance of reducing capabilities (i.e., reductive stress)27–30 will potentially assist with disentangling the contributions of antioxidant supplements. Separately, whether antioxidant supplements have systemic deleterious effects beyond cancer in humans, as seen with lower organism models31, remains to be answered.
Oxidative damage is believed to be an important part of the cytotoxicity of several anti-cancer agents as of radiotherapy32, raising the concern that antioxidants could potentially protect tumors by decreasing therapy efficacy, leading to higher recurrence and death rates. That could provide some mechanistic insight into why antioxidants have worsened prognostic when administered during therapy in some studies33,34. However, it doesn’t help explain why dietary antioxidants seem to increase cancer incidence, as seen in primary prevention trials14,20. Moving from “bedside-to-bench,” several cell line and animal studies have provided solid evidence that antioxidants increase tumor progression and reduce survival in mouse models of cancer through multiple mechanisms35–37. Developing a better mechanistic understanding of antioxidant supplements in cancer is a key step toward dispelling the current narrative of antioxidants as a cure-all and potentially finding coherent treatment avenues with them.
Antioxidants promote tumor initiation and progression
Besides dietary antioxidants, endogenously produced antioxidants govern pivotal roles within cancer cell biology. Glutathione reigns as the most abundant non-protein thiol in mammals (Fig. 1)38. This tripeptide is a key contributor to redox homeostasis as it is a substrate for antioxidant enzymes (e.g., glutathione peroxidases, glutathione-S-transferases, glyoxylases)39. These roles for GSH do not include its impact in modulating protein activity through S-glutathionylation40 and C-glutathionylation41. To a significantly lower extent, GSH spontaneously scavenges different ROS (e.g., hydroxyl radical (OH•), hypochlorous acid (HOCl), alkoxyl radical (RO•), nitric oxide radical (NO•), amongst others)42. To the best of our understanding, the synthesis of GSH is controlled by a two-step reaction. First, glutamate and cysteine are combined by the catalytic subunit of glutamate-cysteine ligase (GCLC) to produce gammaglutamylcysteine. Mice lacking GCLC undergo early embryonic lethality43, suggesting the need for GSH in developing tissues. This reaction is feedback inhibited by GSH; however, the modifying subunit of glutamate-cysteine ligase (GCLM) can significantly increase the threshold required for this inhibition44, allowing for an accumulation of intracellular GSH. GCLM is not required for animal survival; however, its loss prevents tumor initiation and progression45. A pharmacological inhibitor of GCLC, buthionine sulfoximine (BSO), can lower levels of GSH in vitro and in vivo46, although its anti-tumor effect is typically only found when combined with other therapies47–49. While BSO was first described nearly half a century ago50, improved compounds against GCLC have yet to be developed. The second step in GSH synthesis is the condensation of gamma-glutamylcysteine and glycine by glutathione synthetase (GSS) to produce GSH. Interestingly, targeting GSS genetically or pharmacologically in tumor models has largely remained unexplored, potentially due to its essential requirement for embryogenesis51. An alternative approach to lower GSH production is limiting the availability of the amino acid precursors glutamate, glycine, and cysteine (discussed in more detail later). And while these approaches have demonstrated anti-tumor effects, the pleiotropic, GSH-independent endpoints of these amino acids can confound some interpretations.
Figure 1. Dynamic regulation of antioxidants in cancer.
NRF2 is constantly degraded by the proteasome through ubiquitination when bound to KEAP1 and CUL3. The KEAP1/CUL3 complex disengages from NRF2 when oxidized by electrophiles or ROS, leading to NRF2 stabilization and increased transcription of gene targets (shown in yellow). These include enzymes in GSH synthesis (GCLM, GCLC, GSS) and regeneration (GSR), GSH-dependent detoxification (GPX), thioredoxin (TXN), and thioredoxin-related enzymes (TXNRD, PRX). Additionally, NRF2 promotes the transcription of genes involved in quinone reduction (NQO1), iron homeostasis (HMOX1), and NADPH production (G6PD, PGD, ME1, IDH1). Other NRF2 target genes (i.e., ALDH3A1) can induce liabilities, such as reductive stress. Further, the compartmentalization of antioxidants and related metabolites across organelles (i.e., nucleus, mitochondria, ER) plays critical roles in maintaining redox homeostasis in the cell.
Nuclear factor erythroid 2-related factor 2 (NRF2), the master transcriptional regulator antioxidant enzymes (including GCLC, GCLM, and many others), can also support tumorigenesis52 (Fig. 1). NRF2 level is controlled by the KEAP1-CUL3 E3 ubiquitin ligase complex, which constantly tags NRF2 to proteasomal degradation. Under oxidative stress, reactive cysteines in KEAP1 are oxidized, impeding its interaction with NRF2, thus, allowing for NRF2 to accumulate and engage in an antioxidant response. Somatic mutations in NRF2, KEAP1, or CUL3 that prevent NRF2 degradation have been documented in several types of cancer with varying frequencies, reaching up to 25% of lung squamous cell carcinoma, 15% of lung adenocarcinoma and 23% of esophagogastric squamous cell carcinoma53. In all cancers combined, it averages 1%. Human cancers with elevated levels of NRF2 are associated with poor prognosis54,55. It has been suggested that, in animal models, Nrf2 stabilization still requires an oncogenic driver (e.g., Kras) or loss of a tumor suppressor (e.g., Pten) to induce tumorigenesis, as Nrf2 or Keap1 mutations alone do not suffice, even in the absence of tumor suppressors like TP53 or LKB156. Interestingly, somatic alterations in the NRF2 pathway frequently co-occur with alterations in the PI3K pathway53, highlighting a synergistic effect between these two alterations that has also been observed in pre-clinical models57–61.
Nrf2 also rewires glucose metabolism towards the pentose phosphate pathway (PPP)57. This is achieved by transcriptionally upregulating several members of the PPP (e.g., G6PD, PGD), boosting NADPH production, and fueling purine synthesis, a major mechanism through which Nrf2 enables cell proliferation57 (Fig. 1). Interestingly, G6PD expression was not required to grow certain cancers driven by oncogenes (i.e., K-RasG12D)62. However, if G6PD expression is disrupted in cancers with the same oncogenes but also activated Nrf2, cells engage in alternative pathways to produce NADPH (e.g., IDH1, ME1) and drain intermediates from the TCA cycle, which can negatively impact tumor growth63. Overexpression of G6PD in immortalized human or murine cells, resulting in an augmented synthesis of NADPH and nucleosides, is sufficient to make them tumorigenic when xenografted into mice64. Remarkably, the sole addition of exogenous nucleosides or N-acetylcysteine can trigger the same tumorigenic effect. This relationship, however, is continually evolving. Recently, the inactivation of GADPH by oxidative stress was discovered to de-repress G6PD, facilitating flux through the PPP and NAPDH production65. Importantly, several other pathways support NADPH generation and are linked to tumor growth and survival66–69. Beyond Nrf2, further studies are required to deconvolute the impact of metabolic pathways on antioxidant abundance and tumors.
Nrf2’s antioxidant transcriptional program includes genes involved in GSH and thioredoxin (TXN) production/regeneration/utilization, labile iron handling, quinone reduction, and NADPH production; together, these programs can promote resistance to anti-cancer therapies, such as receptor tyrosine kinase inhibitors70,71. However, Nrf2’s antioxidant programs can deplete intracellular metabolites, creating targetable metabolic vulnerabilities72–74. One of these vulnerabilities, a reliance on de novo glutamate production through glutaminolysis, has been recently explored in a clinical trial (KEAPSAKE, NCT04265534) which tested the effect of the GLS inhibitor CB839 in KEAP1/NRF2-mutant non-small cell lung carcinoma (NSCLC). While this trial and others focused on NRF2-specific vulnerabilities have not advanced in the clinic, additional trials (i.e., BeGIN NCT03872427) are ongoing. Recently, more unbiased approaches (i.e., chemical and genetics screens) have been employed to identify new vulnerabilities in NRF2-driven cancers. It was shown that the survival of Keap1-mutated tumors depends on Slc33a1, a gene that encodes an ER-resident acetyl-CoA transporter involved in the unfolded protein response (UPR)75 (Fig. 1). Another screening campaign identified that a subset of NSCLC cell lines reduces proliferation upon Keap1 disruption due to Nrf2-mediated upregulation of the enzyme ALDH3A1, which overproduces NADH causing reductive stress76. Finally, Keap1 loss was identified to sensitize cancer cells to inhibition of the DNA repair protein ATM77. The vulnerabilities unveiled by these studies represent promising targets that warrant further research.
Antioxidants as metastasis-promoting agents
Loss of attachment to the extracellular matrix is a barrier to metastasis faced by epithelial cancer cells78. It was shown that metastatic cells experience elevated oxidative stress compared to primary tumors and cope with this stress by relying on NADPH production79–82. The folate pathway contributes to NADPH production, which potentially helps explain the role of folate in supporting metastasis and may help explain the detrimental effect of folate supplementation on cancer incidence and mortality83. Aside from NADPH production, it was shown that lymph nodes act as a safe haven from oxidative stress for metastatic cancer cells84. In contrast, increased oxidative stress in muscle tissue deters metastasis85. These findings suggest that local pools of antioxidants and ROS significantly impact the potential of some cancers to metastasize and progress.
Another protein that connects antioxidants and metastasis is BACH1, a pro-metastatic transcriptional factor86 that senses intracellular free heme levels. When free heme levels rise (e.g., released from hemoproteins during oxidative stress), BACH1 is marked for proteasomal degradation. Long-term treatment of Kras-TP53-mutated mice with dietary antioxidants (N-acetylcysteine or Vitamin E) reduces free heme levels, stabilizing BACH1 levels and promoting lung metastasis87. BACH1 levels are also stabilized in NRF2/Keap1-mutated tumors due to the upregulation of the classic NRF2 target heme oxygenase 1 (HMOX1), the enzyme responsible for the first and rate-limiting step in heme catabolism. Elevated BACH1 levels promote metastasis in animal models and are associated with increased metastatic burden, poor survival, and higher grade in human lung adenocarcinoma88. The extent to which NRF2 supports other transcription factors to impact tumor metastasis requires additional investigations.
When do antioxidants prevent or slow cancers?
In the previous sections, we discussed the compelling evidence showing how antioxidants support tumorigenesis and metastasis of different tumor types. However, the literature reveals a more complex scenario, as several studies found that activation of NRF2 can have a minimal effect on tumorigenesis89 and, in some cases, slow cancer progression and metastasis90,91. A key distinction is the context in which Nrf2 plays a role. Regarding tumor initiation driven by a carcinogen, NRF2 can act as a suppressor of initiation but a supporter of progression92. More recent studies have demonstrated that in terms of tumor initiation with a genetic driver, NRF2 can support initiation but prevent progression to high-grade tumors56. Further, NRF2 and other antioxidant-related proteins (i.e., TIGAR) can aid in preventing the development of metastasis69. These results emphasize that the role of antioxidants during carcinogenesis is complex and dynamic, changing over time as cells in different stages of carcinogenesis require different features to thrive. Notably, paradoxical functions depending on the threshold of oncogene activation are not uncommon in cancer biology. While oncogenic activation of cMYC and RAS strongly stimulate proliferation and are key drivers of several malignancies, overactivation of these oncogenes induces apoptosis and senescence, respectively93,94.
Aging is another context where antioxidants have demonstrated tumor-suppressive activity. Several mouse models of antioxidant depletion have shown increased cancer incidence, particularly in the later stages of an animal’s life95–97. However, some antioxidants, such as Prdx1, also promote cancer in fast-growing oncogenic-driven models98. It could be surmised that balancing antioxidant regulation is a knife-edge between cancer promotion and age-related damage, placing cancer on either side.
The dichotomy in the impact of antioxidants is observed in other scenarios, such as when a tumor suppressor becomes a tumor “supporter”. The best example is the tumor suppressor BRCA, which promotes the stabilization of NRF299,100. While BRCA is critical in preventing DNA damage101,102 and loss of BRCA leads to the development of a range of tumors103, BRCA doesn’t necessarily stay lost. The use of DNA damage-based therapies in BRCA1/2-mutated cancers has led to an interesting phenomenon in some cases where cancers become resistant to platinum and PARP inhibitors by undergoing reversion mutations that restore the functionality of BRCA104–106. Hypothetically, the cancer has already undergone the necessary mutations to become tumorigenic, and restoration of BRCA1/2 would now support tumor progression. While the most logical impact of BRCA re-expression is to support DNA repair in the face of DNA damage-inducing agents, stabilizing NRF2 is potentially an important factor in this therapy’s resistance and tumor progression. Indeed, NRF2 is critical in preventing DNA damage107. Further research is required to understand whether this patient group is potentially an ideal candidate for therapies that block NRF2 activity or its downstream antioxidant targets.
Cysteine in cancer
At the center of the battle between antioxidants and oxidative stress lies the tower of cysteine. Where cysteine comes from and where it goes is an ever-expanding list. Cells can acquire cysteine from the extracellular environment or synthesize it (from methionine) using the transsulfuration pathway. Uptake of extracellular cysteine (in the oxidized form of cystine) through the system xc− (xCT and 4F2hc) has been a major focus in cancer research108–110 (Fig. 2). Indeed, blocking cystine abundance, uptake, and availability genetically or pharmacologically can impair tumor growth111–114. The extent to which the transsulfuration pathway fuels cysteine production in cancer is less clear. Multiple reports have implicated a dependency of cancers on methionine and enzymes in the transsulfuration pathway115–117. Recent studies have shown that only select tissues (i.e., liver and pancreas) have this capacity to synthesize cysteine from methionine in vivo, and this capacity is diminished during tumorigenesis118. Nonetheless, limiting cystine and methionine in the diet is potentially a tractable approach through adopting a plant-based diet119; whether this approach is sufficient to impair tumor growth and progression remains to be determined.
Figure 2. Acquisition of cysteine from GSH catabolism conserves intracellular metabolites.
System xc− (xCT and 4F2hc) imports cystine at the expense of glutamate export and NADPH consumption. Loss of these metabolites places a burden on other enzymes (i.e., GLS, G6PD) to replenish them and maintain homeostasis. However, GGT-mediated catabolism of GSH costs nothing. Breakdown of GSH by GGT yields glutamate and cysteinylglycine, which upon import, can be further broken down by peptidases into cysteine and glycine. Since this is potentially a more economical route, cancer may favor acquiring cysteine through GSH breakdown.
There are ways for cells to acquire cysteine independent of xCT and transsulfuration pathways. The earliest evidence is from Harry Eagle, showing that supplementation with GSH can rescue cells grown without cystine120. At the time, it was not clear how this rescue occurred. While GSH can be exported from a cell121, the import of GSH is more complicated. A cellular transporter of GSH exists, termed HGT1122; however, this protein is not conserved beyond yeast. Interestingly, GCLM is conserved across all species except yeast. Whether a handoff occurred over time where cells lost the ability to import GSH through HGT1 but gained the ability to accumulate GSH through GCLM is unclear. Nonetheless, while a mechanism to import GSH into mammalian cells has yet to emerge, a family of proteins called gamma-glutamyltransferases can initiate the catabolism of extracellular GSH into its individual amino acids. First postulated more than half a century ago123, GSH is believed to be broken down into glutamate and cysteinylglycine, which can then fuel cysteine abundance in the cell, thus, explaining the observation by Eagle that GSH supplementation could rescue cystine deprivation.
GGT-mediated GSH breakdown might even be a more economical path for the cell to acquire cysteine compared to xCT-mediated cystine import. As previously mentioned, the xCT antiporter expels glutamate in exchange for importing cystine (Fig. 2). Further, upon entering the cell, cystine is reduced to cysteine in a NADPH-dependent reaction124. Thus, to use xCT to acquire cysteine, the cell loses a molecule of glutamate and NADPH125. In contrast, acquiring cysteine from GSH costs nothing. Upon being released from GSH, cysteinylglycine can enter the cell through PEPT1/2 transporters126; afterward, it is further broken down into cysteine and glycine by dipeptidases. The differential importance between GGT and xCT is clearly demonstrated in vivo, where the loss of GGT1 (the putative dominant isoform of GGT) in mice causes perinatal lethality and serum depletion of cysteine127. In contrast, loss of SLC7A11 in mice is well-tolerated and causes no differences in survival128. Further, the importance of GGT1 is found in humans, as patients with inborn errors in GGT1 suffer from glutathionuria and neurological disorders129. While these processes are poorly understood, further research is needed to understand the importance of GGT-mediated cysteine acquisition in tumors. If tumors rely on a mechanism that acquires cysteine while conserving glutamate, this could explain the lack of clinical impact glutaminase inhibitors (i.e., CB-839) have shown in the clinic.
Antioxidants in tumor microenvironments
The tumor microenvironment (TME) is a major determinant of the growth of any given tumor cell. Amino acid availability130, oxygen content131, fluid viscosity132, and other local features define the suitability of any cancerous growth. The milieu surrounding the individual cells of the tumor is the tumor interstitial fluid (TIF)133, a potentially critical source of metabolites (Fig. 3). In pancreatic tumors, the TIF is, surprisingly, low in antioxidant precursors, namely cystine134. While this could reflect a rapid uptake of cystine by tumors, an alternative hypothesis is that other molecules (i.e., GSH) could potentially serve as a source of antioxidant precursors to tumors. Interestingly, cancer cells may also acquire cysteine from surrounding stromal cells. Chronic lymphocytic leukemia (CLL) cells, which exhibit low xCT expression, are supported by marrow stromal cells where the latter consume cystine and subsequently release reduced cysteine135. Other precursors to GSH, such as glycine, are also abundant in the TME of certain malignancies and fuel GSH synthesis in cancer cells136. Together, the antioxidant content of the TME could provide a supportive and nutritive buffering zone for cancer cells to resist stressors.
Figure 3. The tumor microenvironment supplies resources to support antioxidant pools.
The tumor microenvironment (TME) is a heterogeneous composition, which includes cancer cells, stromal, and extracellular matrix. Tumor interstitial fluid (TIF) is the medium within the TME comprised of antioxidants, amino acids, and metabolites. Cancer cells acquire these extracellular molecules through transporters and scavenging processes (i.e., macropinocytosis). Once inside, these molecules (e.g., cysteine) feed antioxidant-dependent (i.e., GSH synthesis, persulfides) and -independent pathways (i.e., protein translation, CoA synthesis).
Tumor and stromal cells can compete for nutrients in the TME137. In this nutrient-scarce environment, extracellular protein scavenging, in the form of micropinocytosis and lysosome-mediated degradation, is necessary to survive138–143 (Fig. 3). Indeed, both stromal and cancer cells engage in macropinocytosis to break down extracellular molecules, such as albumin, to yield precursors for GSH synthesis, such as cysteine and glutamine144,145. Taken together, these observations strongly suggest that macropinocytosis may maintain antioxidant pools by contributing to nutrient uptake in cancers.
Compartmentalization of antioxidants in cancer cells
Subcellular compartmentalization of ROS and antioxidants is an important topic that is, to a large extent, poorly understood in tumors. Several innovative tools have informed the distribution of ROS and antioxidants across the cell146–150, but less is known about how antioxidants transit in and out of organelles or are produced locally within cellular compartments. Only recently was it discovered that mitochondria import GSH through SLC25A39 to maintain the iron-sulfur clusters in the ETC151,152. NADPH is also suggested to have fluxes within the cytosol and mitochondria that are distinct and independent153–155. More recently, using a combination of cysteine profiling and functional genomic screening, it was discovered that an accumulation of ROS in the nucleus could initiate a signaling cascade to dampen ROS production in the mitochondria156. The extent to which similar circuits exist between ROS production and antioxidant synthesis in other organelles warrants further investigation, with potentially exciting findings to be made.
While mitochondria are considered a central location for ROS production, the ER is unique in its elevated redox state, potentially due to the protein folding that occurs there157. Compared to GSH’s highly reduced state in other organelles, glutathione in the ER is predominantly found either in its oxidized state158 or bound to proteins159. Sec61 has been identified as a GSH importer into the ER160; however, whether this process is conserved in mammalian cells is unclear. Further, the production of NADPH through the pentose phosphate pathway can support GSH levels in the ER and maintain appropriate protein folding161. GSH-independent antioxidant enzymes, such as PRDX4, have also been implicated in maintaining the ER redox homeostasis in tumor cells162. Together, these findings implicate the ER as a unique location for redox buffering in the cell.
Conclusion
Along with the questions surrounding antioxidants and cancer posed throughout this review, numerous questions remain. Do tumors from distinct cell lineage (i.e., breast vs. intestinal) have a differential reliance on antioxidants? How does the persulfidation of antioxidant molecules impact tumor growth163–165? How are cofactors (i.e., NADPH) shared between enzymes that support antioxidants and those that drive biosynthetic processes, such as lipogenesis166? How does cysteine’s role in redox buffering balanced by the demand for it by other key tumor processes (i.e., protein translation, CoA synthesis, and iron-sulfur clusters)167–172? How is selenocysteine, a key amino acid in antioxidant enzymes, distributed amongst antioxidant proteins (i.e., TXNRD1/2, GPX4) in cancer cells173,174? Finally, what molecules remain to be discovered as “new” antioxidants? Similar to Sidney Farber’s initial tragic trials with folate175, our poor understanding of antioxidants has led to disappointing and sometimes dangerous outcomes in the clinic. Importantly, however, key aspects of standard-of-care treatments (i.e., radiation and chemotherapies) are linked to the depletion of antioxidants and the induction of oxidative stress. If we can elucidate the regulation of antioxidants in cancer, we can potentially reveal, like Farber, an entirely new world of therapies for patients.
Acknowledgments
I.S.H. is supported by the NIH (R01CA269813), American Cancer Society (RSG-23-971782-01-TBE), and pilot grant funding from the Wilmot Cancer Institute.
I.S.H. reports financial support from Kojin Therapeutics and consulting fees for Ono Pharma USA. Fundings and fees from these companies are outside the scope of the current work.
Footnotes
Declaration of Interests
The authors have no other disclosure to declare.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Murphy MP, Bayir H, Belousov V, Chang CJ, Davies KJA, Davies MJ, Dick TP, Finkel T, Forman HJ, Janssen-Heininger Y, et al. (2022). Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat Metab 4, 651–662. 10.1038/s42255-02200591-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Sies H, Belousov VV, Chandel NS, Davies MJ, Jones DP, Mann GE, Murphy MP, Yamamoto M, and Winterbourn C. (2022). Defining roles of specific reactive oxygen species (ROS) in cell biology and physiology. Nat Rev Mol Cell Biol. 10.1038/s41580-022-00456-z. [DOI] [PubMed] [Google Scholar]
- 3.Cheung EC, and Vousden KH (2022). The role of ROS in tumour development and progression. Nat Rev Cancer 22, 280–297. 10.1038/s41568-021-00435-0. [DOI] [PubMed] [Google Scholar]
- 4.Liang D, Minikes AM, and Jiang X. (2022). Ferroptosis at the intersection of lipid metabolism and cellular signaling. Mol Cell 82, 2215–2227. 10.1016/j.molcel.2022.03.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Rodriguez R, Schreiber SL, and Conrad M. (2022). Persister cancer cells: Iron addiction and vulnerability to ferroptosis. Mol Cell 82, 728–740. 10.1016/j.molcel.2021.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Dixon SJ, and Pratt DA (2023). Ferroptosis: A flexible constellation of related biochemical mechanisms. Mol Cell 83, 1030–1042. 10.1016/j.molcel.2023.03.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Sies H, and Jones DP (2020). Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat Rev Mol Cell Biol 21, 363–383. 10.1038/s41580-020-0230-3. [DOI] [PubMed] [Google Scholar]
- 8.Lennicke C, and Cocheme HM (2021). Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol Cell 81, 3691–3707. 10.1016/j.molcel.2021.08.018. [DOI] [PubMed] [Google Scholar]
- 9.Hyodo I, Amano N, Eguchi K, Narabayashi M, Imanishi J, Hirai M, Nakano T, and Takashima S. (2005). Nationwide survey on complementary and alternative medicine in cancer patients in Japan. J Clin Oncol 23, 2645–2654. 10.1200/JCO.2005.04.126. [DOI] [PubMed] [Google Scholar]
- 10.Velicer CM, and Ulrich CM (2008). Vitamin and mineral supplement use among US adults after cancer diagnosis: a systematic review. J Clin Oncol 26, 665–673. 10.1200/JCO.2007.13.5905. [DOI] [PubMed] [Google Scholar]
- 11.Cowan AE, Jun S, Gahche JJ, Tooze JA, Dwyer JT, Eicher-Miller HA, Bhadra A, Guenther PM, Potischman N, Dodd KW, and Bailey RL (2018). Dietary Supplement Use Differs by Socioeconomic and Health-Related Characteristics among U.S. Adults, NHANES 2011()2014. Nutrients 10. 10.3390/nu10081114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Alpha-Tocopherol, B.C.C.P.S.G. (1994). The effect of vitamin E and beta carotene on the incidence of lung cancer and other cancers in male smokers. N Engl J Med 330, 1029–1035. 10.1056/NEJM199404143301501. [DOI] [PubMed] [Google Scholar]
- 13.Greenberg ER, Baron JA, Tosteson TD, Freeman DH Jr., Beck GJ, Bond JH, Colacchio TA, Coller JA, Frankl HD, Haile RW, and et al. (1994). A clinical trial of antioxidant vitamins to prevent colorectal adenoma. Polyp Prevention Study Group. N Engl J Med 331, 141147. 10.1056/NEJM199407213310301. [DOI] [PubMed] [Google Scholar]
- 14.Omenn GS, Goodman GE, Thornquist MD, Balmes J, Cullen MR, Glass A, Keogh JP, Meyskens FL, Valanis B, Williams JH, et al. (1996). Effects of a combination of beta carotene and vitamin A on lung cancer and cardiovascular disease. N Engl J Med 334, 1150–1155. 10.1056/NEJM199605023341802. [DOI] [PubMed] [Google Scholar]
- 15.Hennekens CH, Buring JE, Manson JE, Stampfer M, Rosner B, Cook NR, Belanger C, LaMotte F, Gaziano JM, Ridker PM, et al. (1996). Lack of effect of long-term supplementation with beta carotene on the incidence of malignant neoplasms and cardiovascular disease. N Engl J Med 334, 1145–1149. 10.1056/NEJM199605023341801. [DOI] [PubMed] [Google Scholar]
- 16.Hercberg S, Galan P, Preziosi P, Bertrais S, Mennen L, Malvy D, Roussel AM, Favier A, and Briancon S. (2004). The SU.VI.MAX Study: a randomized, placebo-controlled trial of the health effects of antioxidant vitamins and minerals. Arch Intern Med 164, 2335–2342. 10.1001/archinte.164.21.2335. [DOI] [PubMed] [Google Scholar]
- 17.Bairati I, Meyer F, Gelinas M, Fortin A, Nabid A, Brochet F, Mercier JP, Tetu B, Harel F, Masse B, et al. (2005). A randomized trial of antioxidant vitamins to prevent second primary cancers in head and neck cancer patients. J Natl Cancer Inst 97, 481–488. 10.1093/jnci/dji095. [DOI] [PubMed] [Google Scholar]
- 18.Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, and Gluud C. (2007). Mortality in randomized trials of antioxidant supplements for primary and secondary prevention: systematic review and meta-analysis. JAMA 297, 842–857. 10.1001/jama.297.8.842. [DOI] [PubMed] [Google Scholar]
- 19.Lippman SM, Klein EA, Goodman PJ, Lucia MS, Thompson IM, Ford LG, Parnes HL, Minasian LM, Gaziano JM, Hartline JA, et al. (2009). Effect of selenium and vitamin E on risk of prostate cancer and other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 301, 39–51. 10.1001/jama.2008.864. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Klein EA, Thompson IM Jr., Tangen CM, Crowley JJ, Lucia MS, Goodman PJ, Minasian LM, Ford LG, Parnes HL, Gaziano JM, et al. (2011). Vitamin E and the risk of prostate cancer: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 306, 15491556. 10.1001/jama.2011.1437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bjelakovic G, Nikolova D, and Gluud C. (2013). Meta-regression analyses, meta-analyses, and trial sequential analyses of the effects of supplementation with beta-carotene, vitamin A, and vitamin E singly or in different combinations on all-cause mortality: do we have evidence for lack of harm? PLoS One 8, e74558. 10.1371/journal.pone.0074558. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Bjelakovic G, Nikolova D, Gluud LL, Simonetti RG, and Gluud C. (2012). Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst Rev 2012, CD007176. 10.1002/14651858.CD007176.pub2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Force USPST, Mangione CM, Barry MJ, Nicholson WK, Cabana M, Chelmow D, Coker TR, Davis EM, Donahue KE, Doubeni CA, et al. (2022). Vitamin, Mineral, and Multivitamin Supplementation to Prevent Cardiovascular Disease and Cancer: US Preventive Services Task Force Recommendation Statement. JAMA 327, 2326–2333. 10.1001/jama.2022.8970. [DOI] [PubMed] [Google Scholar]
- 24.O’Connor EA, Evans CV, Ivlev I, Rushkin MC, Thomas RG, Martin A, and Lin JS (2022). Vitamin and Mineral Supplements for the Primary Prevention of Cardiovascular Disease and Cancer: Updated Evidence Report and Systematic Review for the US Preventive Services Task Force. JAMA 327, 2334–2347. 10.1001/jama.2021.15650. [DOI] [PubMed] [Google Scholar]
- 25.Yun J, Mullarky E, Lu C, Bosch KN, Kavalier A, Rivera K, Roper J, Chio II, Giannopoulou EG, Rago C, et al. (2015). Vitamin C selectively kills KRAS and BRAF mutant colorectal cancer cells by targeting GAPDH. Science 350, 1391–1396. 10.1126/science.aaa5004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Schoenfeld JD, Sibenaller ZA, Mapuskar KA, Wagner BA, Cramer-Morales KL, Furqan M, Sandhu S, Carlisle TL, Smith MC, Abu Hejleh T, et al. (2017). O2(·-) and H2O2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate. Cancer Cell. 10.1016/j.ccell.2017.02.018. [DOI] [PubMed] [Google Scholar]
- 27.Manford AG, Mena EL, Shih KY, Gee CL, McMinimy R, Martinez-Gonzalez B, Sherriff R, Lew B, Zoltek M, Rodriguez-Perez F, et al. (2021). Structural basis and regulation of the reductive stress response. Cell 184, 5375–5390 e5316. 10.1016/j.cell.2021.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Manford AG, Rodriguez-Perez F, Shih KY, Shi Z, Berdan CA, Choe M, Titov DV, Nomura DK, and Rape M. (2020). A Cellular Mechanism to Detect and Alleviate Reductive Stress. Cell 183, 46–61 e21. 10.1016/j.cell.2020.08.034. [DOI] [PubMed] [Google Scholar]
- 29.Maity S, Rajkumar A, Matai L, Bhat A, Ghosh A, Agam G, Kaur S, Bhatt NR, Mukhopadhyay A, Sengupta S, and Chakraborty K. (2016). Oxidative Homeostasis Regulates the Response to Reductive Endoplasmic Reticulum Stress through Translation Control. Cell Rep. 10.1016/j.celrep.2016.06.025. [DOI] [PubMed] [Google Scholar]
- 30.Yang Y, Song Y, and Loscalzo J. (2007). Regulation of the protein disulfide proteome by mitochondria in mammalian cells. Proc Natl Acad Sci U S A 104, 10813–10817. 10.1073/pnas.0702027104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Gusarov I, Shamovsky I, Pani B, Gautier L, Eremina S, Katkova-Zhukotskaya O, Mironov A, Makarov Acapital A C, and Nudler E. (2021). Dietary thiols accelerate aging of C. elegans. Nat Commun 12, 4336. 10.1038/s41467-021-24634-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Hayes JD, Dinkova-Kostova AT, and Tew KD (2020). Oxidative Stress in Cancer. Cancer Cell 38, 167–197. 10.1016/j.ccell.2020.06.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Ambrosone CB, Zirpoli GR, Hutson AD, McCann WE, McCann SE, Barlow WE, Kelly KM, Cannioto R, Sucheston-Campbell LE, Hershman DL, et al. (2020). Dietary Supplement Use During Chemotherapy and Survival Outcomes of Patients With Breast Cancer Enrolled in a Cooperative Group Clinical Trial (SWOG S0221). J Clin Oncol 38, 804–814. 10.1200/JCO.19.01203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Bairati I, Meyer F, Jobin E, Gelinas M, Fortin A, Nabid A, Brochet F, and Tetu B. (2006). Antioxidant vitamins supplementation and mortality: a randomized trial in head and neck cancer patients. Int J Cancer 119, 2221–2224. 10.1002/ijc.22042. [DOI] [PubMed] [Google Scholar]
- 35.Zou ZV, Le Gal K, El Zowalaty AE, Pehlivanoglu LE, Garellick V, Gul N, Ibrahim MX, Bergh PO, Henricsson M, Wiel C, et al. (2021). Antioxidants Promote Intestinal Tumor Progression in Mice. Antioxidants (Basel) 10. 10.3390/antiox10020241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Le Gal K, Ibrahim MX, Wiel C, Sayin VI, Akula MK, Karlsson C, Dalin MG, Akyurek LM, Lindahl P, Nilsson J, and Bergo MO (2015). Antioxidants can increase melanoma metastasis in mice. Sci Transl Med 7, 308re308. 10.1126/scitranslmed.aad3740. [DOI] [PubMed] [Google Scholar]
- 37.Sayin VI, Ibrahim MX, Larsson E, Nilsson JA, Lindahl P, and Bergo MO (2014). Antioxidants accelerate lung cancer progression in mice. Sci. Transl. Med. 6, 221ra215. 10.1126/scitranslmed.3007653 [DOI] [PubMed] [Google Scholar]
- 38.Winterbourn CC, and Hampton MB (2008). Thiol chemistry and specificity in redox signaling. Free Radic. Biol. Med. 45, 549–561. 10.1016/j.freeradbiomed.2008.05.004. [DOI] [PubMed] [Google Scholar]
- 39.Deponte M. (2013). Glutathione catalysis and the reaction mechanisms of glutathione-dependent enzymes. Biochim Biophys Acta 1830, 3217–3266. 10.1016/j.bbagen.2012.09.018. [DOI] [PubMed] [Google Scholar]
- 40.Grek CL, Zhang J, Manevich Y, Townsend DM, and Tew KD (2013). Causes and consequences of cysteine S-glutathionylation. J Biol Chem 288, 26497–26504. 10.1074/jbc.R113.461368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Lai KY, Galan SRG, Zeng Y, Zhou TH, He C, Raj R, Riedl J, Liu S, Chooi KP, Garg N, et al. (2021). LanCLs add glutathione to dehydroamino acids generated at phosphorylated sites in the proteome. Cell 184, 2680–2695 e2626. 10.1016/j.cell.2021.04.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Flohe L. (2013). The fairytale of the GSSG/GSH redox potential. Biochim Biophys Acta 1830, 3139–3142. 10.1016/j.bbagen.2012.10.020. [DOI] [PubMed] [Google Scholar]
- 43.Dalton TP, Dieter MZ, Yang Y, Shertzer HG, and Nebert DW (2000). Knockout of the mouse glutamate cysteine ligase catalytic subunit (Gclc) gene: embryonic lethal when homozygous, and proposed model for moderate glutathione deficiency when heterozygous. Biochem. Biophys. Res. Commun 279, 324–329. 10.1006/bbrc.2000.3930S0006-291X(00)93930-5 [pii]. [DOI] [PubMed] [Google Scholar]
- 44.Chen Y, Shertzer HG, Schneider SN, Nebert DW, and Dalton TP (2005). Glutamate Cysteine Ligase Catalysis: DEPENDENCE ON ATP AND MODIFIER SUBUNIT FOR REGULATION OF TISSUE GLUTATHIONE LEVELS. J. Biol. Chem. 280, 33766–33774. 10.1074/jbc.M504604200. [DOI] [PubMed] [Google Scholar]
- 45.Harris IS, Treloar AE, Inoue S, Sasaki M, Gorrini C, Lee KC, Yung KY, Brenner D, Knobbe-Thomsen CB, Cox MA, et al. (2015). Glutathione and thioredoxin antioxidant pathways synergize to drive cancer initiation and progression. Cancer Cell 27, 211–222. 10.1016/j.ccell.2014.11.019. [DOI] [PubMed] [Google Scholar]
- 46.Watanabe T, Sagisaka H, Arakawa S, Shibaya Y, Watanabe M, Igarashi I, Tanaka K, Totsuka S, Takasaki W, and Manabe S. (2003). A novel model of continuous depletion of glutathione in mice treated with L-buthionine (S,R)-sulfoximine. J. Toxicol. Sci 28, 455–469. [DOI] [PubMed] [Google Scholar]
- 47.Vaziri-Gohar A, Hue JJ, Abbas A, Graor HJ, Hajihassani O, Zarei M, Titomihelakis G, Feczko J, Rathore M, Chelstowska S, et al. (2023). Increased glucose availability sensitizes pancreatic cancer to chemotherapy. Nat Commun 14, 3823. 10.1038/s41467-023-38921-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Harris IS, Endress JE, Coloff JL, Selfors LM, McBrayer SK, Rosenbluth JM, Takahashi N, Dhakal S, Koduri V, Oser MG, et al. (2019). Deubiquitinases Maintain Protein Homeostasis and Survival of Cancer Cells upon Glutathione Depletion. Cell Metab. 10.1016/j.cmet.2019.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lien EC, Lyssiotis CA, Juvekar A, Hu H, Asara JM, Cantley LC, and Toker A. (2016). Glutathione biosynthesis is a metabolic vulnerability in PI(3)K/Akt-driven breast cancer. Nat. Cell Biol 10.1038/ncb3341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Griffith OW, and Meister A. (1979). Potent and specific inhibition of glutathione synthesis by buthionine sulfoximine (S-n-butyl homocysteine sulfoximine). J. Biol. Chem. 254, 7558–7560. [PubMed] [Google Scholar]
- 51.Winkler A, Njalsson R, Carlsson K, Elgadi A, Rozell B, Abraham L, Ercal N, Shi ZZ, Lieberman MW, Larsson A, and Norgren S. (2011). Glutathione is essential for early embryogenesis--analysis of a glutathione synthetase knockout mouse. Biochem Biophys Res Commun 412, 121–126. 10.1016/j.bbrc.2011.07.056. [DOI] [PubMed] [Google Scholar]
- 52.DeNicola GM, Karreth FA, Humpton TJ, Gopinathan A, Wei C, Frese K, Mangal D, Yu KH, Yeo CJ, Calhoun ES, et al. (2011). Oncogene-induced Nrf2 transcription promotes ROS detoxification and tumorigenesis. Nature 475, 106–109. 10.1038/nature10189nature10189 [pii] 10.1038/nature10189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Sanchez-Vega F, Mina M, Armenia J, Chatila WK, Luna A, La KC, Dimitriadoy S, Liu DL, Kantheti HS, Saghafinia S, et al. (2018). Oncogenic Signaling Pathways in The Cancer Genome Atlas. Cell 173, 321–337 e310. 10.1016/j.cell.2018.03.035. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Shibata T, Ohta T, Tong KI, Kokubu A, Odogawa R, Tsuta K, Asamura H, Yamamoto M, and Hirohashi S. (2008). Cancer related mutations in NRF2 impair its recognition by Keap1-Cul3 E3 ligase and promote malignancy. Proc Natl Acad Sci U S A 105, 13568–13573. 10.1073/pnas.0806268105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Solis LM, Behrens C, Dong W, Suraokar M, Ozburn NC, Moran CA, Corvalan AH, Biswal S, Swisher SG, Bekele BN, et al. (2010). Nrf2 and Keap1 abnormalities in non-small cell lung carcinoma and association with clinicopathologic features. Clin Cancer Res 16, 3743–3753. 10.1158/1078-0432.CCR-09-3352. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.DeBlasi JM, Falzone A, Caldwell S, Prieto-Farigua N, Prigge JR, Schmidt EE, Chio IIC, Karreth FA, and DeNicola GM (2023). Distinct Nrf2 Signaling Thresholds Mediate Lung Tumor Initiation and Progression. Cancer Res 83, 1953–1967. 10.1158/0008-5472.CAN-22-3848. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Mitsuishi Y, Taguchi K, Kawatani Y, Shibata T, Nukiwa T, Aburatani H, Yamamoto M, and Motohashi H. (2012). Nrf2 redirects glucose and glutamine into anabolic pathways in metabolic reprogramming. Cancer Cell 22, 66–79. 10.1016/j.ccr.2012.05.016. [DOI] [PubMed] [Google Scholar]
- 58.Lien EC, Lyssiotis CA, Juvekar A, Hu H, Asara JM, Cantley LC, and Toker A. (2016). Glutathione biosynthesis is a metabolic vulnerability in PI(3)K/Akt-driven breast cancer. Nat Cell Biol 18, 572–578. 10.1038/ncb3341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Best SA, De Souza DP, Kersbergen A, Policheni AN, Dayalan S, Tull D, Rathi V, Gray DH, Ritchie ME, McConville MJ, and Sutherland KD (2018). Synergy between the KEAP1/NRF2 and PI3K Pathways Drives Non-Small-Cell Lung Cancer with an Altered Immune Microenvironment. Cell Metab 27, 935–943 e934. 10.1016/j.cmet.2018.02.006. [DOI] [PubMed] [Google Scholar]
- 60.Gwinn DM, Lee AG, Briones-Martin-Del-Campo M, Conn CS, Simpson DR, Scott AI, Le A, Cowan TM, Ruggero D, and Sweet-Cordero EA (2018). Oncogenic KRAS Regulates Amino Acid Homeostasis and Asparagine Biosynthesis via ATF4 and Alters Sensitivity to L-Asparaginase. Cancer Cell 33, 91–107.e106. 10.1016/j.ccell.2017.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cahuzac KM, Lubin A, Bosch K, Stokes N, Shoenfeld SM, Zhou R, Lemon H, Asara J, and Parsons RE (2023). AKT activation because of PTEN loss upregulates xCT via GSK3beta/NRF2, leading to inhibition of ferroptosis in PTEN-mutant tumor cells. Cell Rep 42, 112536. 10.1016/j.celrep.2023.112536. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Ghergurovich JM, Esposito M, Chen Z, Wang JZ, Bhatt V, Lan T, White E, Kang Y, Guo JY, and Rabinowitz JD (2020). Glucose-6-Phosphate Dehydrogenase Is Not Essential for K-Ras-Driven Tumor Growth or Metastasis. Cancer Res 80, 3820–3829. 10.1158/0008-5472.CAN-19-2486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Ding H, Chen Z, Wu K, Huang SM, Wu WL, LeBoeuf SE, Pillai RG, Rabinowitz JD, and Papagiannakopoulos T. (2021). Activation of the NRF2 antioxidant program sensitizes tumors to G6PD inhibition. Sci Adv 7, eabk1023. 10.1126/sciadv.abk1023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Zhang Y, Xu Y, Lu W, Ghergurovich JM, Guo L, Blair IA, Rabinowitz JD, and Yang X. (2021). Upregulation of Antioxidant Capacity and Nucleotide Precursor Availability Suffices for Oncogenic Transformation. Cell Metab 33, 94–109 e108. 10.1016/j.cmet.2020.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Talwar D, Miller CG, Grossmann J, Szyrwiel L, Schwecke T, Demichev V, Mikecin Drazic AM, Mayakonda A, Lutsik P, Veith C, et al. (2023). The GAPDH redox switch safeguards reductive capacity and enables survival of stressed tumour cells. Nat Metab 5, 660–676. 10.1038/s42255-023-00781-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bensaad K, Tsuruta A, Selak MA, Vidal MN, Nakano K, Bartrons R, Gottlieb E, and Vousden KH (2006). TIGAR, a p53-inducible regulator of glycolysis and apoptosis. Cell 126, 107–120. [DOI] [PubMed] [Google Scholar]
- 67.Kremer DM, Nelson BS, Lin L, Yarosz EL, Halbrook CJ, Kerk SA, Sajjakulnukit P, Myers A, Thurston G, Hou SW, et al. (2021). GOT1 inhibition promotes pancreatic cancer cell death by ferroptosis. Nat Commun 12, 4860. 10.1038/s41467-021-24859-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Dey P, Baddour J, Muller F, Wu CC, Wang H, Liao W-T, Lan Z, Chen A, Gutschner T, Kang Y, et al. (2017). Genomic deletion of malic enzyme 2 confers collateral lethality in pancreatic cancer. Nature 542, 119–123. 10.1038/nature21052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cheung EC, DeNicola GM, Nixon C, Blyth K, Labuschagne CF, Tuveson DA, and Vousden KH (2020). Dynamic ROS Control by TIGAR Regulates the Initiation and Progression of Pancreatic Cancer. Cancer Cell. 10.1016/j.ccell.2019.12.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Krall EB, Wang B, Munoz DM, Ilic N, Raghavan S, Niederst MJ, Yu K, Ruddy DA, Aguirre AJ, Kim JW, et al. (2017). KEAP1 loss modulates sensitivity to kinase targeted therapy in lung cancer. Elife 6. 10.7554/eLife.18970. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Foggetti G, Li C, Cai H, Hellyer JA, Lin WY, Ayeni D, Hastings K, Choi J, Wurtz A, Andrejka L, et al. (2021). Genetic Determinants of EGFR-Driven Lung Cancer Growth and Therapeutic Response In Vivo. Cancer Discov 11, 1736–1753. 10.1158/2159-8290.CD-20-1385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Romero R, Sayin VI, Davidson SM, Bauer MR, Singh SX, LeBoeuf SE, Karakousi TR, Ellis DC, Bhutkar A, Sanchez-Rivera FJ, et al. (2017). Keap1 loss promotes Kras-driven lung cancer and results in dependence on glutaminolysis. Nat Med 23, 1362–1368. 10.1038/nm.4407. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Sayin VI, LeBoeuf SE, Singh SX, Davidson SM, Biancur D, Guzelhan BS, Alvarez SW, Wu WL, Karakousi TR, Zavitsanou AM, et al. (2017). Activation of the NRF2 antioxidant program generates an imbalance in central carbon metabolism in cancer. Elife 6. 10.7554/eLife.28083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.LeBoeuf SE, Wu WL, Karakousi TR, Karadal B, Jackson SR, Davidson SM, Wong KK, Koralov SB, Sayin VI, and Papagiannakopoulos T. (2020). Activation of Oxidative Stress Response in Cancer Generates a Druggable Dependency on Exogenous Non-essential Amino Acids. Cell Metab 31, 339–350 e334. 10.1016/j.cmet.2019.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Romero R, Sanchez-Rivera FJ, Westcott PMK, Mercer KL, Bhutkar A, Muir A, Gonzalez Robles TJ, Lamboy Rodriguez S, Liao LZ, Ng SR, et al. (2020). Keap1 mutation renders lung adenocarcinomas dependent on Slc33a1. Nat Cancer 1, 589–602. 10.1038/s43018-0200071-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Weiss-Sadan T, Ge M, Hayashi M, Gohar M, Yao CH, de Groot A, Harry S, Carlin A, Fischer H, Shi L, et al. (2023). NRF2 activation induces NADH-reductive stress, providing a metabolic vulnerability in lung cancer. Cell Metab 35, 487–503 e487. 10.1016/j.cmet.2023.01.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Li H, Liu Y, Xiao Y, Wilson CN, Bai HJ, Jones MD, Wang S, DeVore JE, Maier EY, Durant ST, et al. (2023). CRISPR metabolic screen identifies ATM and KEAP1 as targetable genetic vulnerabilities in solid tumors. Proc Natl Acad Sci U S A 120, e2212072120. 10.1073/pnas.2212072120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Hawk MA, and Schafer ZT (2018). Mechanisms of redox metabolism and cancer cell survival during extracellular matrix detachment. J Biol Chem 293, 7531–7537. 10.1074/jbc.TM117.000260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Piskounova E, Agathocleous M, Murphy MM, Hu Z, Huddlestun SE, Zhao Z, Leitch AM, Johnson TM, DeBerardinis RJ, and Morrison SJ (2015). Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 527, 186–191. 10.1038/nature15726. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Aurora AB, Khivansara V, Leach A, Gill JG, Martin-Sandoval M, Yang C, Kasitinon SY, Bezwada D, Tasdogan A, Gu W, et al. (2022). Loss of glucose 6-phosphate dehydrogenase function increases oxidative stress and glutaminolysis in metastasizing melanoma cells. Proc Natl Acad Sci U S A 119. 10.1073/pnas.2120617119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Hawk MA, Gorsuch CL, Fagan P, Lee C, Kim SE, Hamann JC, Mason JA, Weigel KJ, Tsegaye MA, Shen L, et al. (2018). RIPK1-mediated induction of mitophagy compromises the viability of extracellular-matrix-detached cells. Nat Cell Biol 20, 272–284. 10.1038/s41556-018-0034-2. [DOI] [PubMed] [Google Scholar]
- 82.Tasdogan A, Faubert B, Ramesh V, Ubellacker JM, Shen B, Solmonson A, Murphy MM, Gu Z, Gu W, Martin M, et al. (2020). Metabolic heterogeneity confers differences in melanoma metastatic potential. Nature 577, 115–120. 10.1038/s41586-019-1847-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Ebbing M, Bonaa KH, Nygard O, Arnesen E, Ueland PM, Nordrehaug JE, Rasmussen K, Njolstad I, Refsum H, Nilsen DW, et al. (2009). Cancer incidence and mortality after treatment with folic acid and vitamin B12. JAMA 302, 2119–2126. 10.1001/jama.2009.1622. [DOI] [PubMed] [Google Scholar]
- 84.Ubellacker JM, Tasdogan A, Ramesh V, Shen B, Mitchell EC, Martin-Sandoval MS, Gu Z, McCormick ML, Durham AB, Spitz DR, et al. (2020). Lymph protects metastasizing melanoma cells from ferroptosis. Nature 585, 113–118. 10.1038/s41586-020-2623-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Crist SB, Nemkov T, Dumpit RF, Dai J, Tapscott SJ, True LD, Swarbrick A, Sullivan LB, Nelson PS, Hansen KC, and Ghajar CM (2022). Unchecked oxidative stress in skeletal muscle prevents outgrowth of disseminated tumour cells. Nature Cell Biology 24, 538–553. 10.1038/s41556-022-00881-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Liang Y, Wu H, Lei R, Chong RA, Wei Y, Lu X, Tagkopoulos I, Kung SY, Yang Q, Hu G, and Kang Y. (2012). Transcriptional network analysis identifies BACH1 as a master regulator of breast cancer bone metastasis. J Biol Chem 287, 33533–33544. 10.1074/jbc.M112.392332. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Wiel C, Le Gal K, Ibrahim MX, Jahangir CA, Kashif M, Yao H, Ziegler DV, Xu X, Ghosh T, Mondal T, et al. (2019). BACH1 Stabilization by Antioxidants Stimulates Lung Cancer Metastasis. Cell 178, 330–345 e322. 10.1016/j.cell.2019.06.005. [DOI] [PubMed] [Google Scholar]
- 88.Lignitto L, LeBoeuf SE, Homer H, Jiang S, Askenazi M, Karakousi TR, Pass HI, Bhutkar AJ, Tsirigos A, Ueberheide B, et al. (2019). Nrf2 Activation Promotes Lung Cancer Metastasis by Inhibiting the Degradation of Bach1. Cell 178, 316–329 e318. 10.1016/j.cell.2019.06.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Cai H, Chew SK, Li C, Tsai MK, Andrejka L, Murray CW, Hughes NW, Shuldiner EG, Ashkin EL, Tang R, et al. (2021). A Functional Taxonomy of Tumor Suppression in Oncogenic KRAS-Driven Lung Cancer. Cancer Discov 11, 1754–1773. 10.1158/2159-8290.CD-20-1325. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Khor TO, Huang MT, Prawan A, Liu Y, Hao X, Yu S, Cheung WK, Chan JY, Reddy BS, Yang CS, and Kong AN (2008). Increased susceptibility of Nrf2 knockout mice to colitis-associated colorectal cancer. Cancer Prev Res (Phila) 1, 187–191. 10.1158/1940-6207.CAPR-08-0028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Satoh H, Moriguchi T, Taguchi K, Takai J, Maher JM, Suzuki T, Winnard PT Jr., Raman V, Ebina M, Nukiwa T, and Yamamoto M. (2010). Nrf2-deficiency creates a responsive microenvironment for metastasis to the lung. Carcinogenesis 31, 1833–1843. 10.1093/carcin/bgq105. [DOI] [PubMed] [Google Scholar]
- 92.Satoh H, Moriguchi T, Takai J, Ebina M, and Yamamoto M. (2013). Nrf2 prevents initiation but accelerates progression through the Kras signaling pathway during lung carcinogenesis. Cancer Res. [DOI] [PubMed] [Google Scholar]
- 93.Murphy DJ, Junttila MR, Pouyet L, Karnezis A, Shchors K, Bui DA, Brown-Swigart L, Johnson L, and Evan GI (2008). Distinct thresholds govern Myc’s biological output in vivo. Cancer Cell 14, 447–457. 10.1016/j.ccr.2008.10.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Serrano M, Lin AW, McCurrach ME, Beach D, and Lowe SW (1997). Oncogenic ras provokes premature cell senescence associated with accumulation of p53 and p16INK4a. Cell 88, 593–602. 10.1016/s0092-8674(00)81902-9. [DOI] [PubMed] [Google Scholar]
- 95.Henderson CJ, Smith AG, Ure J, Brown K, Bacon EJ, and Wolf CR (1998). Increased skin tumorigenesis in mice lacking pi class glutathione S-transferases. Proc. Natl. Acad. Sci. U. S. A. 95, 5275–5280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Neumann CA, Krause DS, Carman CV, Das S, Dubey DP, Abraham JL, Bronson RT, Fujiwara Y, Orkin SH, and Van Etten RA (2003). Essential role for the peroxiredoxin Prdx1 in erythrocyte antioxidant defence and tumour suppression. Nature 424, 561–565. 10.1038/nature01819. [DOI] [PubMed] [Google Scholar]
- 97.Van Remmen H, Ikeno Y, Hamilton M, Pahlavani M, Wolf N, Thorpe SR, Alderson NL, Baynes JW, Epstein CJ, Huang TT, et al. (2003). Life-long reduction in MnSOD activity results in increased DNA damage and higher incidence of cancer but does not accelerate aging. Physiol. Genomics 16, 29–37. 10.1152/physiolgenomics.00122.2003 16/1/29 [pii]. [DOI] [PubMed] [Google Scholar]
- 98.Cao J, Schulte J, Knight A, Leslie NR, Zagozdzon A, Bronson R, Manevich Y, Beeson C, and Neumann CA (2009). Prdx1 inhibits tumorigenesis via regulating PTEN/AKT activity. EMBO J. 28, 1505–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Gorrini C, Baniasadi PS, Harris IS, Silvester J, Inoue S, Snow B, Joshi PA, Wakeham A, Molyneux SD, Martin B, et al. (2013). BRCA1 interacts with Nrf2 to regulate antioxidant signaling and cell survival. J. Exp. Med. 210, 1529–1544. 10.1084/jem.20121337 10.1084/jem.20121337. Epub 2013 Jul 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Gorrini C, Gang BP, Bassi C, Wakeham A, Baniasadi SP, Hao Z, Li WY, Cescon DW, Li Y-T, Molyneux S, et al. (2014). Estrogen controls the survival of BRCA1-deficient cells via a PI3K-NRF2-regulated pathway. Proc. Natl. Acad. Sci. U. S. A. 111, 4472–4477. 10.1073/pnas.1324136111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Scully R, Chen J, Plug A, Xiao Y, Weaver D, Feunteun J, Ashley T, and Livingston DM (1997). Association of BRCA1 with Rad51 in mitotic and meiotic cells. Cell 88, 265–275. 10.1016/s0092-8674(00)81847-4. [DOI] [PubMed] [Google Scholar]
- 102.Moynahan ME, Chiu JW, Koller BH, and Jasin M. (1999). Brca1 controls homology-directed DNA repair. Mol Cell 4, 511–518. 10.1016/s1097-2765(00)80202-6. [DOI] [PubMed] [Google Scholar]
- 103.Struewing JP, Hartge P, Wacholder S, Baker SM, Berlin M, McAdams M, Timmerman MM, Brody LC, and Tucker MA (1997). The risk of cancer associated with specific mutations of BRCA1 and BRCA2 among Ashkenazi Jews. N Engl J Med 336, 1401–1408. 10.1056/NEJM199705153362001. [DOI] [PubMed] [Google Scholar]
- 104.Ganesan S. (2018). Tumor Suppressor Tolerance: Reversion Mutations in BRCA1 and BRCA2 and Resistance to PARP Inhibitors and Platinum. JCO Precision Oncology, 1–4. 10.1200/PO.18.00001. [DOI] [PubMed] [Google Scholar]
- 105.Carneiro BA, Collier KA, Nagy RJ, Pamarthy S, Sagar V, Fairclough S, Odegaard J, Lanman RB, Costa R, Taxter T, et al. (2018). Acquired Resistance to Poly (ADP-ribose) Polymerase Inhibitor Olaparib in BRCA2-Associated Prostate Cancer Resulting From Biallelic BRCA2 Reversion Mutations Restores Both Germline and Somatic Loss-of-Function Mutations. JCO precision oncology 2, PO.17.00176. 10.1200/PO.17.00176. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Cheng HH, Salipante SJ, Nelson PS, Montgomery B, and Pritchard CC (2018). Polyclonal BRCA2 Reversion Mutations Detected in Circulating Tumor DNA After Platinum Chemotherapy in a Patient With Metastatic Prostate Cancer. JCO Precision Oncology 2, PO.17.00169. 10.1200/PO.17.00169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Sun X, Wang Y, Ji K, Liu Y, Kong Y, Nie S, Li N, Hao J, Xie Y, Xu C, et al. (2020). NRF2 preserves genomic integrity by facilitating ATR activation and G2 cell cycle arrest. Nucleic Acids Res 48, 9109–9123. 10.1093/nar/gkaa631. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Timmerman LA, Holton T, Yuneva M, Louie RJ, Padró M, Daemen A, Hu M, Chan DA, Ethier SP, van ‘t Veer LJ, et al. (2013). Glutamine Sensitivity Analysis Identifies the xCT Antiporter as a Common Triple-Negative Breast Tumor Therapeutic Target. Cancer Cell 24, 450465. 10.1016/j.ccr.2013.08.020 10.1016/j.ccr.2013.08.020. Epub 2013 Oct 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Liu X, Olszewski K, Zhang Y, Lim EW, Shi J, Zhang X, Zhang J, Lee H, Koppula P, Lei G, et al. (2020). Cystine transporter regulation of pentose phosphate pathway dependency and disulfide stress exposes a targetable metabolic vulnerability in cancer. Nat Cell Biol 22, 476–486. 10.1038/s41556-020-0496-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Guo W, Li K, Sun B, Xu D, Tong L, Yin H, Liao Y, Song H, Wang T, Jing B, et al. (2021). Dysregulated Glutamate Transporter SLC1A1 Propels Cystine Uptake via Xc(−) for Glutathione Synthesis in Lung Cancer. Cancer Res 81, 552–566. 10.1158/0008-5472.CAN-20-0617. [DOI] [PubMed] [Google Scholar]
- 111.Badgley MA, Kremer DM, Maurer HC, DelGiorno KE, Lee HJ, Purohit V, Sagalovskiy IR, Ma A, Kapilian J, Firl CEM, et al. (2020). Cysteine depletion induces pancreatic tumor ferroptosis in mice. Science 368, 85–89. 10.1126/science.aaw9872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Cramer SL, Saha A, Liu J, Tadi S, Tiziani S, Yan W, Triplett K, Lamb C, Alters SE, Rowlinson S, et al. (2016). Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat. Med 10.1038/nm.4232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Upadhyayula PS, Higgins DM, Mela A, Banu M, Dovas A, Zandkarimi F, Patel P, Mahajan A, Humala N, Nguyen TTT, et al. (2023). Dietary restriction of cysteine and methionine sensitizes gliomas to ferroptosis and induces alterations in energetic metabolism. Nat Commun 14, 1187. 10.1038/s41467-023-36630-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Daher B, Parks SK, Durivault J, Cormerais Y, Baidarjad H, Tambutte E, Pouyssegur J, and Vucetic M. (2019). Genetic Ablation of the Cystine Transporter xCT in PDAC Cells Inhibits mTORC1, Growth, Survival, and Tumor Formation via Nutrient and Oxidative Stresses. Cancer Res 79, 3877–3890. 10.1158/0008-5472.CAN-18-3855. [DOI] [PubMed] [Google Scholar]
- 115.Lien EC, Ghisolfi L, Geck RC, Asara JM, and Toker A. (2017). Oncogenic PI3K promotes methionine dependency in breast cancer cells through the cystine-glutamate antiporter xCT. Sci Signal 10. 10.1126/scisignal.aao6604. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Zhu J, Berisa M, Schworer S, Qin W, Cross JR, and Thompson CB (2019). Transsulfuration Activity Can Support Cell Growth upon Extracellular Cysteine Limitation. Cell Metab 30, 865–876 e865. 10.1016/j.cmet.2019.09.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Alborzinia H, Florez AF, Kreth S, Bruckner LM, Yildiz U, Gartlgruber M, Odoni DI, Poschet G, Garbowicz K, Shao C, et al. (2022). MYCN mediates cysteine addiction and sensitizes neuroblastoma to ferroptosis. Nat Cancer 3, 471–485. 10.1038/s43018-022-00355-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Yoon SJ, Combs JA, Falzone A, Prieto-Farigua N, Caldwell S, Ackerman HD, Flores ER, and DeNicola GM (2023). Comprehensive Metabolic Tracing Reveals the Origin and Catabolism of Cysteine in Mammalian Tissues and Tumors. Cancer Res 83, 1426–1442. 10.1158/0008-5472.CAN-22-3000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Sanderson SM, Gao X, Dai Z, and Locasale JW (2019). Methionine metabolism in health and cancer: a nexus of diet and precision medicine. Nat Rev Cancer 19, 625–637. 10.1038/s41568-019-0187-8. [DOI] [PubMed] [Google Scholar]
- 120.Eagle H. (1959). Amino acid metabolism in mammalian cell cultures. Science 130, 432–437. 10.1126/science.130.3373.432. [DOI] [PubMed] [Google Scholar]
- 121.Cao JY, Poddar A, Magtanong L, Lumb JH, Mileur TR, Reid MA, Dovey CM, Wang J, Locasale JW, Stone E, et al. (2019). A Genome-wide Haploid Genetic Screen Identifies Regulators of Glutathione Abundance and Ferroptosis Sensitivity. Cell Rep 26, 1544–1556 e1548. 10.1016/j.celrep.2019.01.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Bourbouloux A, Shahi P, Chakladar A, Delrot S, and Bachhawat AK (2000). Hgt1p, a high affinity glutathione transporter from the yeast Saccharomyces cerevisiae. J. Biol. Chem. 275, 13259–13265. [DOI] [PubMed] [Google Scholar]
- 123.Orlowski M, and Meister A. (1970). The gamma-glutamyl cycle: a possible transport system for amino acids. Proc Natl Acad Sci U S A 67, 1248–1255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Mandal PK, Seiler A, Perisic T, Kölle P, Banjac Canak A, Förster H, Weiss N, Kremmer E, Lieberman MW, Bannai S, et al. (2010). System x(c)- and thioredoxin reductase 1 cooperatively rescue glutathione deficiency. J. Biol. Chem 285, 22244–22253. 10.1074/jbc.M110.121327. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Muir A, Danai LV, Gui DY, Waingarten CY, Lewis CA, and Vander Heiden MG (2017). Environmental cystine drives glutamine anaplerosis and sensitizes cancer cells to glutaminase inhibition. Elife 6. 10.7554/eLife.27713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Frey IM, Rubio-Aliaga I, Siewert A, Sailer D, Drobyshev A, Beckers J, de Angelis MH, Aubert J, Bar Hen A, Fiehn O, et al. (2007). Profiling at mRNA, protein, and metabolite levels reveals alterations in renal amino acid handling and glutathione metabolism in kidney tissue of Pept2−/− mice. Physiol Genomics 28, 301–310. 10.1152/physiolgenomics.00193.2006. [DOI] [PubMed] [Google Scholar]
- 127.Lieberman MW, Wiseman AL, Shi ZZ, Carter BZ, Barrios R, Ou CN, Chévez-Barrios P, Wang Y, Habib GM, Goodman JC, et al. (1996). Growth retardation and cysteine deficiency in gamma-glutamyl transpeptidase-deficient mice. Proc. Natl. Acad. Sci. U. S. A 93, 7923–7926. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Sato H, Shiiya A, Kimata M, Maebara K, Tamba M, Sakakura Y, Makino N, Sugiyama F, Yagami K, Moriguchi T, et al. (2005). Redox imbalance in cystine/glutamate transporter-deficient mice. J. Biol. Chem 280, 37423–37429. 10.1074/jbc.M506439200. [DOI] [PubMed] [Google Scholar]
- 129.Darin N, Leckstrom K, Sikora P, Lindgren J, Almen G, and Asin-Cayuela J. (2018). gammaglutamyl transpeptidase deficiency caused by a large homozygous intragenic deletion in GGT1. Eur J Hum Genet 26, 808–817. 10.1038/s41431-018-0122-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Reinfeld BI, Madden MZ, Wolf MM, Chytil A, Bader JE, Patterson AR, Sugiura A, Cohen AS, Ali A, Do BT, et al. (2021). Cell-programmed nutrient partitioning in the tumour microenvironment. Nature 593, 282–288. 10.1038/s41586-021-03442-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ackerman D, Tumanov S, Qiu B, Michalopoulou E, Spata M, Azzam A, Xie H, Simon MC, and Kamphorst JJ (2018). Triglycerides Promote Lipid Homeostasis during Hypoxic Stress by Balancing Fatty Acid Saturation. Cell Reports 24, 2596–2605.e2595. 10.1016/j.celrep.2018.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Bera K, Kiepas A, Godet I, Li Y, Mehta P, Ifemembi B, Paul CD, Sen A, Serra SA, Stoletov K, et al. (2022). Extracellular fluid viscosity enhances cell migration and cancer dissemination. Nature 611, 365–373. 10.1038/s41586-022-05394-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Wiig H, and Swartz MA (2012). Interstitial fluid and lymph formation and transport: physiological regulation and roles in inflammation and cancer. Physiol Rev 92, 1005–1060. 10.1152/physrev.00037.2011. [DOI] [PubMed] [Google Scholar]
- 134.Sullivan MR, Danai LV, Lewis CA, Chan SH, Gui DY, Kunchok T, Dennstedt EA, Vander Heiden MG, and Muir A. (2019). Quantification of microenvironmental metabolites in murine cancers reveals determinants of tumor nutrient availability. Elife 8. 10.7554/eLife.44235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Zhang W, Trachootham D, Liu J, Chen G, Pelicano H, Garcia-Prieto C, Lu W, Burger JA, Croce CM, Plunkett W, et al. (2012). Stromal control of cystine metabolism promotes cancer cell survival in chronic lymphocytic leukaemia. Nat. Cell Biol 10.1038/ncb2432 ncb2432 [pii] 10.1038/ncb2432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Xia J, Zhang J, Wu X, Du W, Zhu Y, Liu X, Liu Z, Meng B, Guo J, Yang Q, et al. (2022). Blocking glycine utilization inhibits multiple myeloma progression by disrupting glutathione balance. Nat Commun 13, 4007. 10.1038/s41467-022-31248-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Apiz Saab JJ, Dzierozynski LN, Jonker PB, AminiTabrizi R, Shah H, Menjivar RE, Scott AJ, Nwosu ZC, Zhu Z, Chen RN, et al. (2023). Pancreatic tumors exhibit myeloid-driven amino acid stress and upregulate arginine biosynthesis. Elife 12. 10.7554/eLife.81289. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Davidson SM, Jonas O, Keibler MA, Hou HW, Luengo A, Mayers JR, Wyckoff J, Del Rosario AM, Whitman M, Chin CR, et al. (2016). Direct evidence for cancer-cell-autonomous extracellular protein catabolism in pancreatic tumors. Nat. Med 10.1038/nm.4256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Palm W, Park Y, Wright K, Pavlova NN, Tuveson DA, and Thompson CB (2015). The Utilization of Extracellular Proteins as Nutrients Is Suppressed by mTORC1. Cell 162, 259–270. 10.1016/j.cell.2015.06.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Zhang Y, Recouvreux MV, Jung M, Galenkamp KMO, Li Y, Zagnitko O, Scott DA, Lowy AM, and Commisso C. (2021). Macropinocytosis in Cancer-Associated Fibroblasts Is Dependent on CaMKK2/ARHGEF2 Signaling and Functions to Support Tumor and Stromal Cell Fitness. Cancer Discov 11, 1808–1825. 10.1158/2159-8290.CD-20-0119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Zhang MS, Cui JD, Lee D, Yuen VW, Chiu DK, Goh CC, Cheu JW, Tse AP, Bao MH, Wong BPY, et al. (2022). Hypoxia-induced macropinocytosis represents a metabolic route for liver cancer. Nat Commun 13, 954. 10.1038/s41467-022-28618-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Commisso C, Davidson SM, Soydaner-Azeloglu RG, Parker SJ, Kamphorst JJ, Hackett S, Grabocka E, Nofal M, Drebin JA, Thompson CB, et al. (2013). Macropinocytosis of protein is an amino acid supply route in Ras-transformed cells. Nature 497, 633–637. 10.1038/nature12138. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Kamphorst JJ, Nofal M, Commisso C, Hackett SR, Lu W, Grabocka E, Vander Heiden MG, Miller G, Drebin JA, Bar-Sagi D, et al. (2015). Human pancreatic cancer tumors are nutrient poor and tumor cells actively scavenge extracellular protein. Cancer Res 75, 544–553. 10.1158/0008-5472.CAN-14-2211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Armenta DA, Laqtom NN, Alchemy G, Dong W, Morrow D, Poltorack CD, Nathanson DA, Abu-Remalieh M, and Dixon SJ (2022). Ferroptosis inhibition by lysosome-dependent catabolism of extracellular protein. Cell Chem Biol 29, 1588–1600 e1587. 10.1016/j.chembiol.2022.10.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Byun JK, Lee S, Kang GW, Lee YR, Park SY, Song IS, Yun JW, Lee J, Choi YK, and Park KG (2022). Macropinocytosis is an alternative pathway of cysteine acquisition and mitigates sorafenib-induced ferroptosis in hepatocellular carcinoma. J Exp Clin Cancer Res 41, 98. 10.1186/s13046-022-02296-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Mishina NM, Bogdanova YA, Ermakova YG, Panova AS, Kotova DA, Bilan DS, Steinhorn B, Arner ESJ, Michel T, and Belousov VV (2019). Which Antioxidant System Shapes Intracellular H2O2 Gradients? Antioxid Redox Signal 31, 664–670. 10.1089/ars.2018.7697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Gutscher M, Pauleau A-L, Marty L, Brach T, Wabnitz GH, Samstag Y, Meyer AJ, and Dick TP (2008). Real-time imaging of the intracellular glutathione redox potential. Nat. Methods 5, 553–559. 10.1038/nmeth.1212. [DOI] [PubMed] [Google Scholar]
- 148.Gutscher M, Sobotta MC, Wabnitz GH, Ballikaya S, Meyer AJ, Samstag Y, and Dick TP (2009). Proximity-based protein thiol oxidation by H2O2-scavenging peroxidases. J. Biol. Chem 284, 31532–31540. 10.1074/jbc.M109.059246 10.1074/jbc.M109.059246. Epub 2009 Sep 15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Albrecht SC, Barata AG, Grosshans J, Teleman AA, and Dick TP (2011). In Vivo Mapping of Hydrogen Peroxide and Oxidized Glutathione Reveals Chemical and Regional Specificity of Redox Homeostasis. Cell Metab. [DOI] [PubMed] [Google Scholar]
- 150.Morgan B, Van Laer K, Owusu TNE, Ezeriņa D, Pastor-Flores D, Amponsah PS, Tursch A, and Dick TP (2016). Real-time monitoring of basal H2O2 levels with peroxiredoxin-based probes. Nat. Chem. Biol 10.1038/nchembio.2067. [DOI] [PubMed] [Google Scholar]
- 151.Wang Y, Yen FS, Zhu XG, Timson RC, Weber R, Xing C, Liu Y, Allwein B, Luo H, Yeh H-W, et al. (2021). SLC25A39 is necessary for mitochondrial glutathione import in mammalian cells. Nature 599, 136–140. 10.1038/s41586-021-04025-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Shi X, Reinstadler B, Shah H, To TL, Byrne K, Summer L, Calvo SE, Goldberger O, Doench JG, Mootha VK, and Shen H. (2022). Combinatorial GxGxE CRISPR screen identifies SLC25A39 in mitochondrial glutathione transport linking iron homeostasis to OXPHOS. Nat Commun 13, 2483. 10.1038/s41467-022-30126-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Niu X, Stancliffe E, Gelman SJ, Wang L, Schwaiger-Haber M, Rowles JL 3rd, Shriver LP, and Patti GJ (2023). Cytosolic and mitochondrial NADPH fluxes are independently regulated. Nat Chem Biol 19, 837–845. 10.1038/s41589-023-01283-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Lewis CA, Parker SJ, Fiske BP, McCloskey D, Gui DY, Green CR, Vokes NI, Feist AM, Vander Heiden MG, and Metallo CM (2014). Tracing compartmentalized NADPH metabolism in the cytosol and mitochondria of mammalian cells. Mol Cell 55, 253–263. 10.1016/j.molcel.2014.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Fan J, Ye J, Kamphorst JJ, Shlomi T, Thompson CB, and Rabinowitz JD (2014). Quantitative flux analysis reveals folate-dependent NADPH production. Nature 510, 298–302. 10.1038/nature13236 10.1038/nature13236. Epub 2014 May 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Zhang J, Simpson CM, Berner J, Chong HB, Fang J, Ordulu Z, Weiss-Sadan T, Possemato AP, Harry S, Takahashi M, et al. (2023). Systematic identification of anticancer drug targets reveals a nucleus-to-mitochondria ROS-sensing pathway. Cell 186, 2361–2379 e2325. 10.1016/j.cell.2023.04.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Tu BP, and Weissman JS (2004). Oxidative protein folding in eukaryotes: mechanisms and consequences. J. Cell Biol. 164, 341–346. 10.1083/jcb.200311055. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hwang C, Sinskey AJ, and Lodish HF (1992). Oxidized redox state of glutathione in the endoplasmic reticulum. Science 257, 1496–1502. [DOI] [PubMed] [Google Scholar]
- 159.Bass R, Ruddock LW, Klappa P, and Freedman RB (2004). A major fraction of endoplasmic reticulum-located glutathione is present as mixed disulfides with protein. J Biol Chem 279, 5257–5262. 10.1074/jbc.M304951200. [DOI] [PubMed] [Google Scholar]
- 160.Ponsero AJ, Igbaria A, Darch MA, Miled S, Outten CE, Winther JR, Palais G, D’Autréaux B, Delaunay-Moisan A, and Toledano MB (2017). Endoplasmic Reticulum Transport of Glutathione by Sec61 Is Regulated by Ero1 and Bip. Mol. Cell 67, 962–973.e965. 10.1016/j.molcel.2017.08.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161.Li H, Ericsson M, Rabasha B, Budnik B, Chan SH, Freinkman E, Lewis CA, Doench JG, Wagner BK, Garraway LA, and Schreiber SL (2019). 6-Phosphogluconate Dehydrogenase Links Cytosolic Carbohydrate Metabolism to Protein Secretion via Modulation of Glutathione Levels. Cell Chem Biol 26, 1306–1314 e1305. 10.1016/j.chembiol.2019.05.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Jain P, Dvorkin-Gheva A, Mollen E, Malbeteau L, Xie M, Jessa F, Dhavarasa P, Chung S, Brown KR, Jang GH, et al. (2021). NOX4 links metabolic regulation in pancreatic cancer to endoplasmic reticulum redox vulnerability and dependence on PRDX4. Sci Adv 7. 10.1126/sciadv.abf7114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Barayeu U, Schilling D, Eid M, Xavier da Silva TN, Schlicker L, Mitreska N, Zapp C, Grater F, Miller AK, Kappl R, et al. (2023). Hydropersulfides inhibit lipid peroxidation and ferroptosis by scavenging radicals. Nat Chem Biol 19, 28–37. 10.1038/s41589-022-01145-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Benchoam D, Semelak JA, Cuevasanta E, Mastrogiovanni M, Grassano JS, Ferrer-Sueta G, Zeida A, Trujillo M, Moller MN, Estrin DA, and Alvarez B. (2020). Acidity and nucleophilic reactivity of glutathione persulfide. J Biol Chem 295, 15466–15481. 10.1074/jbc.RA120.014728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wu Z, Khodade VS, Chauvin JR, Rodriguez D, Toscano JP, and Pratt DA (2022). Hydropersulfides Inhibit Lipid Peroxidation and Protect Cells from Ferroptosis. J Am Chem Soc 144, 15825–15837. 10.1021/jacs.2c06804. [DOI] [PubMed] [Google Scholar]
- 166.Nelson ME, Lahiri S, Chow JD, Byrne FL, Hargett SR, Breen DS, Olzomer EM, Wu LE, Cooney GJ, Turner N, et al. (2017). Inhibition of hepatic lipogenesis enhances liver tumorigenesis by increasing antioxidant defence and promoting cell survival. Nat Commun 8, 14689. 10.1038/ncomms14689. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Kelly B, Carrizo GE, Edwards-Hicks J, Sanin DE, Stanczak MA, Priesnitz C, Flachsmann LJ, Curtis JD, Mittler G, Musa Y, et al. (2021). Sulfur sequestration promotes multicellularity during nutrient limitation. Nature 591, 471–476. 10.1038/s41586-021-03270-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Dibble CC, Barritt SA, Perry GE, Lien EC, Geck RC, DuBois-Coyne SE, Bartee D, Zengeya TT, Cohen EB, Yuan M, et al. (2022). PI3K drives the de novo synthesis of coenzyme A from vitamin B5. Nature 608, 192–198. 10.1038/s41586-022-04984-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Jouandin P, Marelja Z, Shih YH, Parkhitko AA, Dambowsky M, Asara JM, Nemazanyy I, Dibble CC, Simons M, and Perrimon N. (2022). Lysosomal cystine mobilization shapes the response of TORC1 and tissue growth to fasting. Science 375, eabc4203. 10.1126/science.abc4203. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Alvarez SW, Sviderskiy VO, Terzi EM, Papagiannakopoulos T, Moreira AL, Adams S, Sabatini DM, Birsoy K, and Possemato R. (2017). NFS1 undergoes positive selection in lung tumours and protects cells from ferroptosis. Nature. 10.1038/nature24637. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Fujihara KM, Zhang BZ, Jackson TD, Ogunkola MO, Nijagal B, Milne JV, Sallman DA, Ang CS, Nikolic I, Kearney CJ, et al. (2022). Eprenetapopt triggers ferroptosis, inhibits NFS1 cysteine desulfurase, and synergizes with serine and glycine dietary restriction. Sci Adv 8, eabm9427. 10.1126/sciadv.abm9427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Matye D, Gunewardena S, Chen J, Wang H, Wang Y, Hasan MN, Gu L, Clayton YD, Du Y, Chen C, et al. (2022). TFEB regulates sulfur amino acid and coenzyme A metabolism to support hepatic metabolic adaptation and redox homeostasis. Nat Commun 13, 5696. 10.1038/s41467-022-33465-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Eaton JK, Furst L, Ruberto RA, Moosmayer D, Hilpmann A, Ryan MJ, Zimmermann K, Cai LL, Niehues M, Badock V, et al. (2020). Selective covalent targeting of GPX4 using masked nitrile-oxide electrophiles. Nat Chem Biol 16, 497–506. 10.1038/s41589-020-0501-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Takahashi N, Cho P, Selfors LM, Kuiken HJ, Kaul R, Fujiwara T, Harris IS, Zhang T, Gygi SP, and Brugge JS (2020). 3D Culture Models with CRISPR Screens Reveal Hyperactive NRF2 as a Prerequisite for Spheroid Formation via Regulation of Proliferation and Ferroptosis. Mol Cell 80, 828–844 e826. 10.1016/j.molcel.2020.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Farber S, and Diamond LK (1948). Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid. N Engl J Med 238, 787–793. 10.1056/NEJM194806032382301. [DOI] [PubMed] [Google Scholar]