Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2025 May 1.
Published in final edited form as: Trends Endocrinol Metab. 2024 Feb 20;35(5):400–412. doi: 10.1016/j.tem.2024.01.009

Dietary methionine restriction on cancer development and antitumor immunity

Ming Ji 1,2, Qing Xu 1,2, Xiaoling Li 1,*
PMCID: PMC11096033  NIHMSID: NIHMS1961485  PMID: 38383161

Abstract

Methionine restriction has been shown to suppress tumor growth and improve the responses to various anti-cancer therapies. However, methionine itself is required for proliferation, activation, and differentiation of T cells that are critical for antitumor immunity. The dual impact of methionine, influencing both tumor and immune cells, has generated concerns regarding the potential consequences of methionine restriction on T-cell immunity and its possible role in promoting cancer. In this review, we systemically examined current literature on the interactions between dietary methionine, cancer cells, and immune cells. Based on recent findings in immunocompetent animals with methionine restriction, we further discussed how tumor stage-specific methionine dependence of immune cells and cancer cells in the tumor microenvironment could ultimately dictate the response of tumors to methionine restriction.

Keywords: sulfur metabolism, methylation, redox homeostasis, antitumor immunity, gut microbiota

Interactions between dietary methionine, cancer cells, immune cells, and gut microbiota

Cancer cells demand abundant nutrients for their rapid growth, epigenetic manipulation, and redox balance [1]. Restricting nutrients like glucose and amino acids, especially methionine (see Glossary), has been explored as a less toxic alternative to traditional cancer treatments [2-4]. As the only sulfur-containing essential amino acid, methionine initiates protein translation and fuels vital biological processes such as methylation and redox homeostasis [5], which are closely linked to cancer development and tumor progression. The discovery of cancer methionine dependency has opened the avenue to exploiting methionine restriction (MR) as a promising nutritional strategy in cancer therapy [5-7], complementing its established benefits in aging and metabolic health [8,9].

The importance of methionine in fundamental cellular processes makes sustained methionine supply also essential for the proliferation and activation of immune cells. As an integral part of antitumor immunity, immune cells like T cells, Natural killer cells and tumor associated macrophages are responsible for identifying and eliminating abnormal cancerous growth [10]. Recent studies have shown that T cells require methionine for their activation and differentiation [11-13]. Since methionine is an essential amino acid that can only be obtained from the diet, competition between cancer cells and immune cells for dietary methionine has presented a challenge for effective cancer treatment with MR.

Another key player in dietary methionine intervention-based cancer therapy is the human microbiome, whose composition and function are significantly influenced by different diets [14]. In turn, the microbiota and its metabolites could also modulate host metabolism and immune system [15,16]. Therefore, MR as a dietary approach involves complex interactions between diets, cancer cells, immune cells, and microbiota. Consequently, despite promising findings that MR inhibits tumor growth and improves cancer therapy, factors such as the timing, duration, and degree of MR, the stage and the type of cancer, as well as the immune status of patients, could all confound the tumor response to MR-based treatments [17-20]. This review will focus on recent research on the precise mechanisms and clinical implications of these interactions.

Methionine metabolism

Methionine metabolism is regulated through several interconnected pathways (Figure 1). In the methionine cycle, methionine transforms into the universal methyl donor S-Adenosyl methionine (SAM). SAM is also crucial for polyamine synthesis, producing byproducts that can regenerate methionine via the methionine salvage pathway. SAM can be converted to homocysteine (Hcy), which, through remethylation (via the folate cycle) or transsulfuration pathways, contributes to the synthesis of methionine, cysteine, and other key sulfur-containing molecules such as hydrogen sulfide (H2S) and glutathione (GSH). These pathways, integral to one-carbon metabolism, sustain the synthesis of purine and pyrimidine (the folate cycle), methylation (the methionine cycle), and redox balance (the transsulfuration pathway), all of which are vital for cell growth and function [5].

Figure 1. One-carbon metabolic pathways and key enzymes.

Figure 1.

MAT1A: Methionine Adenosyltransferase 1A; MAT2A and 2B: Methionine Adenosyltransferase 2A and 2B; BHMT: Betaine-Homocysteine S-Methyltransferase; CBS: Cystathionine Beta-Synthase; CTH: Cystathionine Gamma-Lyase; CDO1: Cysteine Dioxygenase Type 1.

Cell-autonomous impact of methionine metabolism on tumor growth

The reliance of cancer cells on methionine was initially revealed when tumor cells were found to be more dependent on exogenous methionine than normal cells [7]. Although the degree of methionine dependence varies among human cancer cells [21], this finding led to a hypothesis that limiting methionine intake could cell-autonomously hinder cancer growth. Indeed, various studies have demonstrated that MR inhibits tumor growth and enhances the effectiveness of cytotoxic agents in both cultured cells and animal models [19,22-26]. Cancer cell-autonomous mechanisms, such as cell cycle arrest, apoptosis, and alterations in nucleotide and redox metabolism, have been reported to underlie the antitumor effect of MR [5]. In-depth metabolic analyses further revealed that MR directly impacts metabolic flux in one-carbon metabolism in tumor cells, leading to inhibition of tumor growth and sensitization of tumors to chemotherapy and radiotherapy [5,26]. In this section, we summarize some key cell-autonomous mechanisms linking MR-induced metabolic changes to phenotypic outcomes in cancer cells (Figure 2).

Figure 2. Cell-autonomous impact of methionine metabolism on cancer cells.

Figure 2.

Cancer cells are highly dependent on exogenous methionine for their proliferation, survival, and stress resistance. Firstly, SAM-dependent methylation of DNA and histones epigenetically regulates the expression of key factors involved in mitophagy, apoptosis, and oncogenesis, thereby inhibiting cell death and promoting tumorigenesis. SAM-dependent methylation of RNA and non-histone proteins involved in regulation of immune inhibition and type I interferon response in cancer cells hinders their response to immunotherapy. Secondly, sulfur-containing antioxidative molecules derived through the transsulfuration pathway, including cysteine, GSH, and H2S, are important for cancer cells survival and stress resistance. GSH protects cancer cells from ferroptosis and apoptosis induced by ROS and lipid peroxides. It also promotes oxidative phosphorylation (OXPHOS) and ATP production through glutathionylation (GS) of mitochondrial enzymes. H2S, when at a low physiological concentration, acts as a cytoprotective agent by suppressing ROS, promoting energy metabolism, and enhancing angiogenesis. Many of these actions are mediated, at least partially, by protein sulfhydration (SH). Consequently, MR, cysteine restriction, and/or elevated polyamine flux have been reported to inhibit tumor growth, induce cell death, and sensitize tumor to different anti-cancer therapies. Figure was created with BioRender.com.

SAM-dependent methylation

As the primary methyl donor for the methylation of DNA, histone, RNA, and other proteins, SAM is important for epigenetic regulation of gene expression, DNA repair, and protein functions [27,28]. MR has been reported to increase cancer cell death through suppression of SAM-dependent DNA methylation. For instance, in gastric cancer cells, the promoter of a mitochondrial mitophagy receptor protein BNIP3 is hypermethylated. MR induces demethylation of this promoter, enhancing BNIP3 expression and promoting mitophagic cell death [29].

MR and subsequent SAM depletion also induce rapid change of histone methylation [30], which alters gene expression and may mediate the effect of MR on cancer [31]. For example, in acute myeloid leukemia, methionine starvation reduces H3K36me3 and induces cell death [32]. In tumor-initiating cancer cells (TIC), MR causes drastic decrease of both SAM levels and overall histone methylation, disrupting the tumorigenic potential of TIC [33]

MR also affects RNA methylation, which takes place mainly at the N6 position of an adenine base (m6A) [34]. MR has been shown to reduce the m6A methylation thereby affecting the translation of immune checkpoint transcripts PD-L1 and V-domain Ig suppressor of T cell activation (VISTA) in tumor cells, enhancing their response to antitumor immunity [35].

Additionally, MR can suppress tumor growth through reduced methylation of non-histone proteins. CyclicGMP-AMPsynthase (cGAS), a cytosolic DNA sensor that produces cGAMP to activate STING mediated type I interferon signaling upon binding of cytosolic DNA, has been shown to be methylated by methyltransferase SUV39H1 in cancer cells [36]. This modification leads to chromatin sequestration of cGAS. MR reduces the methylation of cGAS, decreasing its chromatin tethering and promoting its DNA binding, which in turn activates the type I interferon pathway in cultured cancer cells. Interestingly, targeting the SUV39H1-cGAS axis in either cancer cells or host cells increases immune cell infiltration, reduces tumor growth, and sensitizes tumors to radiotherapy and immunotherapy [36].

SAM-dependent polyamine biosynthesis

SAM is important for synthesis of polyamines. Elevated polyamine flux can compete with other methionine/SAM-dependent pathways for SAM in cancer cells, making them sensitive to interventions that block methionine recycling or disrupt redox balance. For example, enhanced polyamine flux synergizes with inhibition of methionine salvage pathway to induce apoptosis in prostate cancer cells [37]. Increased polyamine pathway activity also makes cancer cells more dependent on de novo-synthesized cysteine from methionine, thereby synergizing with cysteine starvation to induce their death [38]. Therefore, therapeutic interventions that deplete methionine/SAM through activating the polyamine pathway have the potential to enhance the efficacy of MR-mediated cancer treatment.

Cysteine, GSH, and redox homeostasis

Cysteine obtained from diet or synthesized from methionine through the transsulfuration pathway is rate-limiting for synthesis of GSH, an important thiol-containing antioxidant that maintains redox homeostasis and protects cells from oxidative stress induced by reactive oxidative species (ROS) and lipid peroxides [39,40]. Due to their increased ROS production, many cancer cells have high levels of GSH, which contributes to their resistance to anticancer therapy [41]. MR-induced GSH depletion sensitizes cancer cells to anticancer therapy. For instance, in triple-negative breast cancer (TNBC) cells, MR-induced GSH depletion leads to increased accumulation of ROS, which activates NRF2 to induce thioredoxin reductase (TXNRD), rendering tumors more dependent on the thioredoxin pathway for redox balance. Consequently, MR synergizes with TXNRD inhibitor auranofin to inhibit TNBC tumor growth [42].

Depletion of cysteine or cystine, which is often removed in the MR diets in animal studies, mimics the impact of MR on GSH and antioxidative defense in cancer cells. In leukemia stem cells (LSCs), cysteine depletion impairs GSH synthesis and reduces glutathionylation of succinate dehydrogenase A (SDHA), a key regulator of electron transport chain complex (ETC) II. Loss of SDHA glutathionylation inhibits ETC II activity and oxidative phosphorylation, diminishing ATP production and inducing LSC death [43]. In multiple cancer allograft/xenograft models, systemic depletion of cysteine or cystine with an engineered human cyst(e)inase diminishes GSH, resulting in cell cycle arrest and death in cancer cells [44].

Cysteine depletion-induced GSH reduction can also trigger ferroptosis. For example, cysteine depletion through inhibition of cystine import induces ferroptosis in pancreatic cancer [45]. In glycolytic ovarian clear cell carcinoma, cysteine depletion suppresses tumor growth primarily by oxidative stress-dependent necrosis and ferroptosis [46]. In esophageal squamous cell carcinoma, dietary methionine/cystine restriction downregulates the expression of methionine transporter SLC43A2, GPX4a, and the NFκB signaling pathway. The blocked NFκB pathway further decreases the expression of SLC43A2 and GPX4, inducing ferroptosis and apoptosis [47]. In glioma, methionine/cysteine deprivation can synergize with the GPX4 inhibitor RSL3 to increase lipid peroxidation and ferroptotic cell death [48]. Interestingly, a recent report showed that cyst(e)ine deprivation-induced shortage of lysosomal cystine, but not cytosolic cysteine, sensitizes cancer cells to ferroptosis through the aryl hydrocarbon receptor-ATF4 axis [49].

Cysteine depletion may play a large role in MR-mediated antitumor effects, as cysteine supplementation reverses the effects of MR on adiposity and ROS production [50,51] as well as on tumor inhibition [20]. In line with this notion, the cystine deprivation-induced defect in antioxidative defense in cancer cells can be affected by the manipulation of upstream methionine metabolism. Short-term methionine starvation prompts transcription of cation transport regulator homolog 1 (CHAC1), a protein essential for GSH degradation, enhancing tumor ferroptosis [18]. Intermittent methionine deprivation also induces tumor ferroptosis, synergizing with PD-1 blockade-activated CD8+ T cells to improve the tumor response to immunotherapy. However, prolonged methionine deprivation suppresses ferroptosis initiation by impeding the translation of CHAC1 [18]. Conversely, activation of transsulfuration pathway that generates cysteine and GSH attenuates ferroptosis induced by cysteine deprivation [52,53].

H2S

The transsulfuration pathway also generates H2S, a gasotransmitter that regulates vasodilation, cellular bioenergetics, and anti-inflammation. H2S also modulates cell signaling through protein sulfhydration [54,55]. The role of H2S in cancer appears to be complex with a bimodal pharmacological character [54]. Generally, H2S maintains normal physiology and acts as a cytoprotective and antioxidant agent at a low physiological concentration. These actions affect the proliferation and migration of cancer cells in a tumor-cell-type dependent manner. After reaching a certain threshold, H2S may promote tumorigenesis. On the other hand, at a higher concentration, H2S exhibits tumor inhibition effects through various mechanisms, including mitochondrial inhibition, activation of cell death signaling, intracellular acidification, and activation of apoptosis. Consequently, H2S donors have been considered as anti-cancer drugs [54,56].

Enzymes involved in metabolism of sulfur-containing amino acids (SAAs)

Given the significant impact of methionine and sulfur metabolic irregularities on tumor development, it is not surprising that SAA metabolic enzymes are frequently dysregulated in cancers [57-59]. For instance, in human liver tumors, many SAA enzymes are downregulated. This reduction correlates with aggressive tumors and a poor prognosis [60-63]. Recently, we discovered that HNF4α, the master regulator of hepatic genes, transcriptionally regulates key enzymes in SAA metabolism in the liver transsulfuration pathway, such as BHMT, CBS, CTH, CDO1, and the liver-specific MAT1A that converts methionine into SAM. This regulation dictates the sensitivity of liver cancer to MR-induced cell death. Knocking down HNF4α or SAA enzymes in HNF4α-positive epithelial liver cancer lines impairs SAA metabolism and promotes epithelial-mesenchymal transition, increasing resistance to MR [19].

Several studies have targeted MAT2A, the rate-limiting enzyme that produces SAM from methionine in extrahepatic tissues, for MR-mediated cancer treatment [64-66]. In breast cancer stem cells (CSC), the combination of MR and MAT2A inhibition is more effective than either alone for tumor inhibition [64]. In diffuse midline gliomas, low levels of MAT2A increase their dependence on methionine. MR therefore reduces the tumor growth and extends the survival of tumor-bearing mice [65]. MAT2A is upregulated in cisplatin resistant bladder cancer cells, and its knockdown synergizes with MR to restore cisplatin sensitivity in immunodeficient mice but not in an immunocompetent model. Instead, inhibition of SLC7A6, a methionine transporter that is highly expressed in cisplatin resistant cancer cells but minimally expressed in CD8+ T cells, is additionally required for MAT2A inhibition to overcome cisplatin resistance in immunocompetent mice [66].

Non-cell-autonomous impact of methionine metabolism on cancer through antitumor immunity

Methionine is also critical for the growth and survival of non-cancer cells. For instance, a sustained supply of exogenous methionine is required for the proliferation and activation of T cells [13], a critical component of antitumor immunity [11]. Therefore, methionine metabolism could also non-cell-autonomously impact cancer growth and progression through regulation of immune cell-mediated antitumor immunity. Below are a few mechanisms underscoring these non-cell-autonomous impacts (Figure 3).

Figure 3. The impact of methionine metabolism on immune cells and antitumor immunity.

Figure 3.

First, methionine (Met) is critical for the proliferation and activation of effector T cells, a vital component of antitumor immunity. SAM-dependent methylation of H3K79me2 increases the expression STAT5 thereby activating CD8+ T cells. This modification also increases the expression of AMPK in conventional CD4+ T cells, leading to suppression of PD1. During tumor progression, tumor cells can outcompete T cells for methionine in the tumor microenvironment by increasing the expression of SLC43A2, impairing T cell-mediated antitumor immunity. Moreover, the methionine uptake of tumor-infiltrating T cells can be inhibited by the acidic metabolic waste products within the tumor microenvironment, which reduces H3K27me3 at the promoters of critical T cell memory genes, keeping them in a 'stem-like memory' state with reduced effector functions. Furthermore, dietary methionine and cysteine (Cys) are important precursors for gut microbial production of H2S, which can enhance T cell survival and activity by increasing GAPDH sulhydration and glycolysis. MR can therefore impair effector T cell-mediated antitumor immunity. Finally, methionine and sulfur metabolites are also important for the survival and function of regulatory immune cells in vitro. SLC43A2-mediated methionine uptake is essential for the survival of activated Treg cells, and H2S-mediated sulfhydration of NFYB promotes the differentiation of Treg through TETs and FOXP3. System Xc- mediated import of cystine in MDSCs sequesters extracellular cysteine and cystine to block full T cell activation. Figure was created with BioRender.com.

SAM-dependent histone methylation in regulation of T cell immunity

Methionine can boost T cell activation and differentiation via SAM-mediated enhancement of histone activation marks. In CD4+ helper T cells, MR decreases H3K4me3 on the promoters of genes involved in cytokine production and cell cycle progression, suppressing pathogenic Th17 cell expansion and mitigating T cell-mediated neuroinflammation and autoimmune diseases in mice [67]. Bian et al. recently showed that tumor cells can surpass CD8+ T cells in the competition for methionine via high expression of SLC43A2 in the tumor microenvironment. This competition decreases intracellular methionine and SAM in T cells, leading to reduced H3K79me2 on the promoter of STAT5, a key player in T cell development, and hindering T cell-mediated antitumor immunity [12]. Reduced methionine in the tumor microenvironment also affects CD4+ helper T cells. Low methionine in CD4+ T cells decreased H3K79me2, leading to downregulation of AMPK. Reduced AMPK then increases the expression of PD-1, the T cell checkpoint inhibitor, and impairs antitumor immunity of CD4+ T cells [68].

The methionine uptake of tumor-infiltrating T cells could also be inhibited by the acidic metabolic waste products within the tumor microenvironment [69]. This metabolic rewiring reduces intracellular SAM and the deposition of H3K27me3, a repressive histone mark, at the promoters of critical T cell memory-associated genes, such as CCR7, KLF2, LEF1, and TCF7. T cells maintained in an acidic environment, therefore, are kept in a 'stem-like memory' state with inhibited effector functions. Intriguingly, when adoptively transferred into recipient mice, these T cells display a long-term in vivo persistence and antitumor activity [69].

H2S, gut microbiota, and immune system

Sulfur-containing metabolites produced in the transsulfuration pathway, including GSH and H2S, can also modulate the function and activity of immune cells involved in innate and adaptive immunity [70,71]. For instance, GSH is required for T-cell proliferation and effector functions [72]. H2S-mediated immune regulation, like what is observed in cancer cells, is context- and dose-dependent (bell-shaped or bimodal), with an optimal H2S “zone” for physiological functions and therapeutic improvements of various immune cells in health and disease. Mechanistically, an optimal H2S level protects immune cells from ROS-induced toxicity, increases their viability, and stimulates their mobility, thereby enhancing their function [70]. Additionally, physiological levels of H2S enhance activation of T cells by promoting cell proliferation and the expression of markers characteristic of the activation state [73]. Therefore, H2S could regulate cancer development and progression non-cell-autonomously through modulation of antitumor immunity.

Interestingly, the gut is a major H2S-producing organ, with a large portion of H2S produced from sulfate-producing gut microbiota that metabolize inorganic or organic sulfur compounds in the diet [74]. Given that gut microbiota is known to interact with diets to regulate host metabolism and immune system, and that the modulation of gut microbiota can enhance immunotherapy for cancer [15,16,75], H2S could be an important mediator of MR on cancer development and treatment. Indeed, a recent study from our group has found that MR reduces H2S produced from the microbiota, and this reduction contributes at least partially to MR-induced impairment of antitumor immunity [20]. Mechanistically, H2S enhances immune cell survival and activity by increasing GAPDH sulhydration, boosting glycolysis.

Methionine metabolism in regulatory immune cells

Methionine and sulfur metabolites are also important for the survival and function of regulatory immune cells. Activated T-regulatory (Treg) cells) have high methionine uptake via SLC43A2, which is essential for their survival upon IL-2 deprivation in vitro [76]. Moreover, the differentiation of Treg cells can be driven by H2S-mediated sulfhydration of NFYB, which promotes the expression of methylcytosine dioxygenases Tet1 and Tet2, leading to demethylation of the promoter of Foxp3 [77]. Myeloid-derived suppressor cells (MDSCs), known for their strong inhibition of T-cell-mediated antitumor immunity, express the system xc- transporter that imports cystine but lacks the cysteine export transporter [78]. This expression pattern allows MDSCs to import extracellular cystine without releasing cysteine back into their microenvironment, leading to sequestration of extracellular cysteine and cystine that blocks full T cell activation in vitro [78].

The impact of tumor methionine metabolism on T cell immunity

Dysregulated methionine metabolism in tumor cells has been linked to impaired T cell immunity. For instance, in hepatocellular carcinoma (HCC), elevated SAM and 5-methylthioadenosine (MTA), two key metabolites in the methionine salvage pathway, induce T cell exhaustion [79]. As discussed previously, increased methionine uptake of tumor cells during tumor progression via SLC43A2 can outcompete CD8+ T cells for methionine, leading to impaired T cell survival and function [12]; SAM-mediated m6A methylation of PD-L1 and VISTA in tumor cells inhibits T cell immunity [35]; and SAM-mediated methylation of cGAS reduces type I interferon signaling in cultured cancer cells, and reduced methylation of cGAS is linked to increased immune cell infiltration and sensitivity to immunotherapy in vivo [36]. These studies suggest that elevated methionine metabolism in tumor cells may blunt the response of tumors to antitumor immunotherapy.

The impact of MR on antitumor immunotherapy

The importance of methionine and its metabolites in the metabolic and epigenetic regulation of immune cells, particularly T cells, strongly suggests that MR could impair immune cell-mediated antitumor immunity. In support of this notion, intratumoral supplementation of methionine have been reported to increase CD8+ T cell immunity in tumor-bearing mice and patients with colon cancer [12] and reduce PD-1 expression in CD4+ T cells and suppress tumor growth [68]. Dietary supplementation of methionine, cystine, or a hydrogen sulfide donor stimulates anti-tumor immunity and suppresses tumor progression [20]. However, recent studies have revealed the dichotomy of MR on the response of tumors to antitumor immunotherapy in immunocompetent mice [18,20,35,80,81], highlighting the multifaceted effects of MR on the interaction between cancer cells and immune cells in the tumor microenvironment (Figure 4). Careful comparison of the experimental design and therapeutic outcome of each of above studies suggests that the impact of MR on the response of tumors to antitumor immunotherapy is likely dependent on the cancer stage. In studies where MR was initiated after tumors were established, MR synergizes with antitumor immunotherapy to suppress tumor growth [18,35,80,81]. However, in the study where MR was started before tumors were initiated/established, MR impairs the efficacy of antitumor immunotherapy [20].

Figure 4, Key Figure. The impact of MR on tumor growth and therapeutic response is dependent on the relative methionine dependence of T cells vs cancer cells in the tumor microenvironment.

Figure 4, Key Figure.

MR is anti-cancer in immunodeficient individuals with tumors that are sensitive to MR. In healthy people or immunocompetent patients at early cancer stages, MR may become pro-cancer by impairing T cell activation, which could lead to uncontrolled tumor growth or resistance to immunotherapy. In cancer patients at advanced stages, tumor cells may exhibit a higher dependency on methionine in comparison to T cells. MR could synergize with non-immune-mediated therapeutic regimens to suppress tumor progression. Therefore, the impact of MR on tumor growth and therapeutic response is affected by the immune status and tumor stage. Figure was created with BioRender.com.

For established tumors in immunocompetent mice, MR enhances their response to antitumor immunotherapy through multiple mechanisms. These tumor-cell-autonomous actions include decreasing m6A RNA methylation on the transcripts of immune checkpoint genes PD-L1 and VISTA thereby reducing their expression and their inhibition of T cells [35], stimulating the transcription of a GSH degradation protein CHAC1 thereby sensitizing tumor cells to ferroptosis [18], or promoting cGAS-STING pathway and type I interferon signaling in cultured cancer cells in vitro [36,81]. In these studies, MR increased the response of established tumors to antitumor immunotherapy indirectly by either reducing the immune-inhibiting activity of tumor cells or increasing their sensitivity to redox stress. Therefore, with the exception of one study showing that MR skews innate tumor-associated macrophages toward a more tumoricidal M1-type phenotype [80], currently, there is a lack of direct evidence confirming that MR could genuinely augment the efficacy of immunotherapy by boosting immune cell function and their antitumor immunity in these experimental settings.

The experimental setting in which the dietary methionine intervention was started prior to the onset of tumor growth directly investigates the influence of dietary intervention on the intrinsic antitumor immunity derived from immune cells [20,82]. In this setting, dietary MR exacerbates cancer progression and impairs the outcome of antitumor immunotherapy in multiple cancer models in immunocompetent mice by diminishing the survival and activity of immune cells, notably T cells [20]. This finding aligns with prior studies showing that methionine metabolism is indispensable for the immune responses, especially T cell-mediated antitumor immunity [12,13,67,68]. Moreover, in addition to previously revealed SAM-mediated epigenetic regulation [12,67,68], dietary MR hinders the proliferation and activation of T cells primarily through impairment of redox homeostasis downstream of SAM, as supplementation of cysteine (a H2S precursor), a H2S donor, or H2S-producing microbes, counteracts the MR-induced resistance to anti-tumor immunotherapy [20]. Therefore, methionine and its downstream metabolites play crucial roles in regulating gene expression, redox balance, and cellular proliferation in T cells, ultimately impacting their ability to mount effective immune responses against cancer.

In conclusion, the complex relationship between dietary methionine intervention and antitumor immunity involves the direct modulation of immune cell function as well as indirect immune regulation from gut microbiota and cancer cells, and this relationship is sensitive to tumor stage. Thus, the timing of MR initiation and tumor stage will determine the MR-induced cancer outcomes.

Concluding remarks and future perspectives

Despite extensive research into the effects of MR on tumors, currently, there are no ongoing cancer clinical trials involving the combination of dietary methionine intervention and immunotherapy. However, a recent study in healthy adults indicates that outcomes of dietary sulfur amino acid restriction in animal models are applicable to humans [83]. As previously noted, methionine and its downstream products are important for overall functions of both tumor cells and immune cells. Thus, whether dietary MR exerts pro-cancer or anti-cancer effects is contextually dependent on the relative requirement for methionine by tumor cells compared to immune cells (Figure 4). Preclinical mouse studies suggest that MR might be advantageous for immunocompromised cancer patients due to its direct inhibitory effects on tumor growth without involvement of the immune system. On the other hand, for healthy people or immunocompetent patients at early cancer stages, MR may impair immune cell activation, which could lead to uncontrolled tumor growth or resistance to immunotherapy. In advanced cancer cases, tumor cells may exhibit a higher dependency on methionine in comparison to immune cells within the tumor microenvironment. At this stage, the synergistic integration of MR with non-immune-mediated therapeutic regimens, such as chemotherapy or radiation therapy, represents a promising therapeutic strategy (Figure 4, key figure).

Of important note, dietary MR exerts profound impacts on metabolism, immunity, and overall health [83]. For example, MR in young rats stunts growth and impairs bone health [84]. Several studies that examine the feasibility of MR treatment in humans have suggested that short-term MR treatment is well tolerated, but the long-term safety and potential side effects of sustained MR remain to be further understood [26,85-88]. Thorough research is therefore imperative to assess the safety of dietary methionine intervention for diverse patient populations. Importantly, substantial challenges persist before clinical implementation (see Outstanding questions). In the future, a multidisciplinary approach is needed to address these challenges and to further test the personalized and tumor stage-specific strategies for cancer treatment with methionine intervention.

Outstanding questions.

  1. Which tumor types are more sensitive to methionine intervention?

  2. Could biosensors/tracers evaluating tumor methionine dependence be developed to guide methionine intervention for cancer treatment?

  3. Which specific probiotic strains have the potential to amplify the therapeutic effects of methionine supplements or H2S donor?

  4. What is the impact of MR on immune cells other than the T-cells, such as B cells, natural killer cells, neutrophils, monocytes, and dendritic cells?

Highlights.

  1. As a sulfur-containing essential amino acid important for a broad array of fundamental cellular processes, methionine is critical for the proliferation, stress resistance, and overall functions of both cancer cells and immune cells.

  2. Dietary methionine regulates the antitumor immunity by directly influencing immune cell function and indirectly modulating the immune system through interactions with gut microbiota and cancer cells.

  3. The ultimate effect of dietary methionine restriction on tumor growth and therapeutic response depends on the relative reliance of T cells versus cancer cells on methionine in the tumor microenvironment.

  4. Any possible anti-cancer benefits of dietary methionine restriction require careful consideration of the immune system, microbiota, and tumor stage.

Acknowledgements

We thank members of the Li laboratory, and Drs. Lih-Wen Deng and Zefeng Wang for critical reading of the manuscript. The work related to this article was supported by the Intramural Research Program of the NIH, National Institute of Environmental Health Sciences to X.L. (Z01 ES102205). We apologize to those colleagues whose work has not been cited due to space limitations.

Glossary

Antitumor immunity

refers to the body's immune responses against the development and progression of cancer. T cells play a central role in antitumor immunity. Cytotoxic (CD8+) T cells recognize and eliminate tumor cells. Helper (CD4+) T cells promote the response of cytotoxic T cells, while T-regulatory cells (Tregs) expressing the transcription factor FoxP3 suppress the activity of cytotoxic T cells. Myeloid-derived suppressor cells (MDSCs) are known to inhibit the activation of CD8+ and CD4+ T cells.

Ferroptosis

a form of cell death characterized by iron-dependent lipid peroxide accumulation, GSH is required to detoxify peroxides through glutathione peroxidase 4 (GPX4).

Methionine and cysteine

the only two sulfur-containing proteinogenic amino acids. Methionine is essential and is acquired solely through diet, while cysteine is semi-essential and can be obtained from diet or synthesized from methionine. Cystine is the more stable dimeric form of cysteine.

Oxidative stress

a condition caused by an imbalance in redox status, characterized by an excess of reactive oxygen species (ROS) or inadequate antioxidants that scavenge and neutralize ROS. This condition can cause damage to cellular components and contribute to the development of various diseases, including cancer.

PD-1/PD-L1 Targeting Immunotherapy

a type of cancer treatment that focuses on blocking the interaction between the programmed cell death protein 1 (PD-1) on T cells and its ligand, programmed death-ligand 1 (PD-L1) on cancer cells. This interaction is a key immune checkpoint that can suppress the immune response and allow cancer cells to evade elimination by the immune system. Antibody-based inhibitors of PD1/PDL1 disrupt the immune checkpoint pathway and boost T cell-mediated antitumor immunity, exhibiting substantial effectiveness against many tumors.

Polyamine synthesis

the cellular process that generates polyamines such as putrescine, spermidine, and spermine. Polyamines are small positively molecules that interact with negatively charged macromolecules, such as DNA, RNA, protein, and phospholipids, and are essential for cell growth and function.

Protein sulfhydration

a post-translational modification that adds a thiol group (-SH) to cysteine residues in proteins, forming a persulfide bond (-SSH).

Redox balance

the equilibrium between oxidation and reduction reactions that involve the transfer of electrons between molecules. Maintaining the balance between the oxidized and reduced molecules in the body is crucial for cellular function and overall health.

SAM-dependent methylation

a biochemical process in which a methyl group is transferred from SAM to various substrates including the cytosine base of DNA, the lysine (K) and arginine (R) residues of histone proteins, the N6 position of the adenine base (m6A) of RNA, the lysine (K) residue of non-histone proteins.

Thioredoxin reductase (TXNRD)

the rate-limiting enzyme in the thioredoxin pathway that reduces thioredoxin using NADPH. The thioredoxin system is a key antioxidant system in defense against oxidative stress by regulating protein dithiol/disulfide balance.

Footnotes

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.

Declaration of interests

The authors declare no competing interests.

References

  • 1.Faubert B. et al. (2020) Metabolic reprogramming and cancer progression. Science 368, 152-+. ARTN eaaw5473 10.1126/science.aaw5473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Wang L. et al. (2018) Autophagy mediates glucose starvation-induced glioblastoma cell quiescence and chemoresistance through coordinating cell metabolism, cell cycle, and survival. Cell Death & Disease 9. ARTN 213 10.1038/s41419-017-0242-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Kang M. et al. (2023) Glucose Deprivation Induces Cancer Cell Death through Failure of ROS Regulation. International Journal of Molecular Sciences 24. ARTN 11969 10.3390/ijms241511969 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Butler M. et al. (2021) Amino Acid Depletion Therapies: Starving Cancer Cells to Death. Trends Endocrinol Metab 32, 367–381. 10.1016/j.tem.2021.03.003 [DOI] [PubMed] [Google Scholar]
  • 5.Sanderson SM et al. (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]
  • 6.Sugimura T. et al. (1959) Quantitative nutritional studies with water-soluble, chemically defined diets. VIII. The forced feeding of diets each lacking in one essential amino acid. Arch Biochem Biophys 81, 448–455. 10.1016/0003-9861(59)90225-5 [DOI] [PubMed] [Google Scholar]
  • 7.Mecham JO et al. (1983) The Metabolic Defect of Methionine Dependence Occurs Frequently in Human-Tumor Cell-Lines. Biochemical and Biophysical Research Communications 117, 429–434. Doi 10.1016/0006-291x(83)91218-4 [DOI] [PubMed] [Google Scholar]
  • 8.Orentreich N. et al. (1993) Low methionine ingestion by rats extends life span. J Nutr 123, 269–274. 10.1093/jn/123.2.269 [DOI] [PubMed] [Google Scholar]
  • 9.Miller RA et al. (2005) Methionine-deficient diet extends mouse lifespan, slows immune and lens aging, alters glucose, T4, IGF-I and insulin levels, and increases hepatocyte MIF levels and stress resistance. Aging Cell 4, 119–125. 10.1111/j.1474-9726.2005.00152.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Yang YL et al. (2023) T cells, NK cells, and tumor-associated macrophages in cancer immunotherapy and the current state of the art of drug delivery systems. Frontiers in Immunology 14. ARTN 1199173 10.3389/fimmu.2023.1199173 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Zhao T and Lum JJ (2022) Methionine cycle-dependent regulation of T cells in cancer immunity. Frontiers in Oncology 12. ARTN 969563 10.3389/fonc.2022.969563 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Bian Y. et al. (2020) Cancer SLC43A2 alters T cell methionine metabolism and histone methylation. Nature 585, 277–282. 10.1038/s41586-020-2682-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Sinclair LV et al. (2019) Antigen receptor control of methionine metabolism in T cells. Elife 8. 10.7554/eLife.44210 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Frame LA et al. (2020) Current explorations of nutrition and the gut microbiome: a comprehensive evaluation of the review literature. Nutr Rev 78, 798–812. 10.1093/nutrit/nuz106 [DOI] [PubMed] [Google Scholar]
  • 15.Tremaroli V and Backhed F. (2012) Functional interactions between the gut microbiota and host metabolism. Nature 489, 242–249. 10.1038/nature11552 [DOI] [PubMed] [Google Scholar]
  • 16.Zheng D. et al. (2020) Interaction between microbiota and immunity in health and disease. Cell Res 30, 492–506. 10.1038/s41422-020-0332-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Wanders D. et al. (2020) Methionine Restriction and Cancer Biology. Nutrients 12. ARTN 684 10.3390/nu12030684 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Xue Y. et al. (2023) Intermittent dietary methionine deprivation facilitates tumoral ferroptosis and synergizes with checkpoint blockade. Nature Communications 14. 10.1038/S41467-023-40518-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Xu Q. et al. (2020) HNF4alpha regulates sulfur amino acid metabolism and confers sensitivity to methionine restriction in liver cancer. Nat Commun 11, 3978. 10.1038/S41467-020-17818-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ji M. et al. (2023) Methionine restriction-induced sulfur deficiency impairs antitumour immunity partially through gut microbiota. Nat Metab. 10.1038/s42255-023-00854-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Stern PH et al. (1984) Altered methionine metabolism occurs in all members of a set of diverse human tumor cell lines. J Cell Physiol 119, 29–34. 10.1002/jcp.1041190106 [DOI] [PubMed] [Google Scholar]
  • 22.Guo H. et al. (1993) Therapeutic tumor-specific cell cycle block induced by methionine starvation in vivo. Cancer Res 53, 5676–5679 [PubMed] [Google Scholar]
  • 23.Poirson-Bichat F. et al. (1997) Growth of methionine-dependent human prostate cancer (PC-3) is inhibited by ethionine combined with methionine starvation. Br J Cancer 75, 1605–1612. 10.1038/bjc.1997.274 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Sinha R. et al. (2014) Dietary methionine restriction inhibits prostatic intraepithelial neoplasia in TRAMP mice. Prostate 74, 1663–1673. 10.1002/pros.22884 [DOI] [PubMed] [Google Scholar]
  • 25.Hens JR et al. (2016) Methionine-restricted diet inhibits growth of MCF10AT1-derived mammary tumors by increasing cell cycle inhibitors in athymic nude mice (vol 16, 349, 2016). Bmc Cancer 16. ARTN 474 10.1186/s12885-016-2404-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Gao X. et al. (2019) Dietary methionine influences therapy in mouse cancer models and alters human metabolism. Nature 572, 397–401. 10.1038/s41586-019-1437-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Mato JM et al. (2013) S-adenosylmethionine metabolism and liver disease. Annals of Hepatology 12, 183–189. Doi 10.1016/S1665-2681(19)31355-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Bauerle MR et al. (2015) Mechanistic Diversity of Radical-Adenosylmethionine (SAM)-dependent Methylation. Journal of Biological Chemistry 290, 3995–4002. 10.1074/jbc.R114.607044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Xin L. et al. (2021) Methionine deficiency promoted mitophagy via lncRNA PVT1-mediated promoter demethylation of BNIP3 in gastric cancer. International Journal of Biochemistry & Cell Biology 141. ARTN 106100 10.1016/j.biocel.2021.106100 [DOI] [PubMed] [Google Scholar]
  • 30.Mentch SJ and Locasale JW (2016) One-carbon metabolism and epigenetics: understanding the specificity. Ann N Y Acad Sci 1363, 91–98. 10.1111/nyas.12956 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Mentch SJ et al. (2015) Histone Methylation Dynamics and Gene Regulation Occur through the Sensing of One-Carbon Metabolism. Cell Metab 22, 861–873. 10.1016/j.cmet.2015.08.024 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cunningham A. et al. (2022) Dietary methionine starvation impairs acute myeloid leukemia progression. Blood 140, 2037–2052. 10.1182/blood.2022017575 [DOI] [PubMed] [Google Scholar]
  • 33.Wang Z. et al. (2019) Methionine is a metabolic dependency of tumor-initiating cells. Nat Med 25, 825–837. 10.1038/s41591-019-0423-5 [DOI] [PubMed] [Google Scholar]
  • 34.Boulias K and Greer EL (2023) Biological roles of adenine methylation in RNA. Nature Reviews Genetics 24, 143–160. 10.1038/s41576-022-00534-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Li T. et al. (2023) Methionine deficiency facilitates antitumour immunity by altering m(6)A methylation of immune checkpoint transcripts. Gut 72, 501–511. 10.1136/gutjnl-2022-326928 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Fang L. et al. (2023) Methionine restriction promotes cGAS activation and chromatin untethering through demethylation to enhance antitumor immunity. Cancer Cell 41, 1118-+. 10.1016/j.ccell.2023.05.005 [DOI] [PubMed] [Google Scholar]
  • 37.Affronti HC et al. (2020) Pharmacological polyamine catabolism upregulation with methionine salvage pathway inhibition as an effective prostate cancer therapy. Nature Communications 11. ARTN 52 10.1038/s41467-019-13950-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zhang T. et al. (2020) Polyamine pathway activity promotes cysteine essentiality in cancer cells. Nature Metabolism 2, 1062-+. 10.1038/s42255-020-0253-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Zhang HF et al. (2022) Transsulfuration, minor player or crucial for cysteine homeostasis in cancer. Trends in Cell Biology 32, 800–814. 10.1016/j.tcb.2022.02.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Nin DS et al. (2021) Cysteine Metabolism in Cancer Progression and Therapy Resistance. Oxidative Stress: Human Diseases and Medicine, 155–191. 10.1007/978-981-16-0522-2_7 [DOI] [Google Scholar]
  • 41.Hatem E. et al. (2017) Multifaceted Roles of Glutathione and Glutathione-Based Systems in Carcinogenesis and Anticancer Drug Resistance. Antioxid Redox Signal 27, 1217–1234. 10.1089/ars.2017.7134 [DOI] [PubMed] [Google Scholar]
  • 42.Malin D. et al. (2021) Methionine restriction exposes a targetable redox vulnerability of triple-negative breast cancer cells by inducing thioredoxin reductase. Breast Cancer Res Treat 190, 373–387. 10.1007/s10549-021-06398-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Jones CL et al. (2019) Cysteine depletion targets leukemia stem cells through inhibition of electron transport complex II. Blood 134, 389–394. 10.1182/blood.2019898114 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Cramer SL et al. (2017) Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat Med 23, 120–127. 10.1038/nm.4232 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Badgley MA 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]
  • 46.Novera W. et al. (2020) Cysteine Deprivation Targets Ovarian Clear Cell Carcinoma Via Oxidative Stress and Iron-Sulfur Cluster Biogenesis Deficit. Antioxid Redox Signal 33, 1191–1208. 10.1089/ars.2019.7850 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Peng H. et al. (2023) SLC43A2 and NFkappaB signaling pathway regulate methionine/cystine restriction-induced ferroptosis in esophageal squamous cell carcinoma via a feedback loop. Cell Death Dis 14, 347. 10.1038/s41419-023-05860-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Upadhyayula PS et al. (2023) Dietary restriction of cysteine and methionine sensitizes gliomas to ferroptosis and induces alterations in energetic metabolism. Nature Communications 14. ARTN 1187 10.1038/s41467-023-36630-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Swanda RV et al. (2023) Lysosomal cystine governs ferroptosis sensitivity in cancer via cysteine stress response. Mol Cell 83, 3347–3359 e3349. 10.1016/j.molcel.2023.08.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Elshorbagy AK et al. (2011) Cysteine supplementation reverses methionine restriction effects on rat adiposity: significance of stearoyl-coenzyme A desaturase. J Lipid Res 52, 104–112. 10.1194/jlr.M010215 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Gomez A. et al. (2015) Cysteine dietary supplementation reverses the decrease in mitochondrial ROS production at complex I induced by methionine restriction. J Bioenerg Biomembr 47, 199–208. 10.1007/s10863-015-9608-x [DOI] [PubMed] [Google Scholar]
  • 52.Yu X and Long YC (2016) Crosstalk between cystine and glutathione is critical for the regulation of amino acid signaling pathways and ferroptosis. Sci Rep 6, 30033. 10.1038/srep30033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Hayano M. et al. (2016) Loss of cysteinyl-tRNA synthetase (CARS) induces the transsulfuration pathway and inhibits ferroptosis induced by cystine deprivation. Cell Death Differ 23, 270–278. 10.1038/cdd.2015.93 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Szabo C. (2016) Gasotransmitters in cancer: from pathophysiology to experimental therapy. Nat Rev Drug Discov 15, 185–203. 10.1038/nrd.2015.1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Zhang D. et al. (2017) H(2)S-Induced Sulfhydration: Biological Function and Detection Methodology. Front Pharmacol 8, 608. 10.3389/fphar.2017.00608 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Wang RH et al. (2021) The Hidden Role of Hydrogen Sulfide Metabolism in Cancer. Int J Mol Sci 22. 10.3390/ijms22126562 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Kim J. et al. (2009) Expression of cystathionine beta-synthase is downregulated in hepatocellular carcinoma and associated with poor prognosis. Oncol Rep 21, 1449–1454. 10.3892/or_00000373 [DOI] [PubMed] [Google Scholar]
  • 58.Ascencao K and Szabo C. (2022) Emerging roles of cystathionine beta-synthase in various forms of cancer. Redox Biol 53, 102331. 10.1016/j.redox.2022.102331 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Brait M. et al. (2012) Cysteine dioxygenase 1 is a tumor suppressor gene silenced by promoter methylation in multiple human cancers. PLoS One 7, e44951. 10.1371/journal.pone.0044951 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Avila MA et al. (2000) Reduced mRNA abundance of the main enzymes involved in methionine metabolism in human liver cirrhosis and hepatocellular carcinoma. J Hepatol 33, 907–914. 10.1016/s0168-8278(00)80122-1 [DOI] [PubMed] [Google Scholar]
  • 61.Frau M. et al. (2012) Role of transcriptional and posttranscriptional regulation of methionine adenosyltransferases in liver cancer progression. Hepatology 56, 165–175. 10.1002/hep.25643 [DOI] [PubMed] [Google Scholar]
  • 62.Jin B. et al. (2016) Downregulation of betaine homocysteine methyltransferase (BHMT) in hepatocellular carcinoma associates with poor prognosis. Tumour Biol 37, 5911–5917. 10.1007/s13277-015-4443-6 [DOI] [PubMed] [Google Scholar]
  • 63.Pascale RM et al. (2018) Deregulation of methionine metabolism as determinant of progression and prognosis of hepatocellular carcinoma. Transl Gastroenterol Hepatol 3, 36. 10.21037/tgh.2018.06.04 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Strekalova E. et al. (2019) S-adenosylmethionine biosynthesis is a targetable metabolic vulnerability of cancer stem cells. Breast Cancer Res Treat 175, 39–50. 10.1007/s10549-019-05146-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Golbourn BJ et al. (2022) Loss of MAT2A compromises methionine metabolism and represents a vulnerability in H3K27M mutant glioma by modulating the epigenome. Nat Cancer 3, 629–648. 10.1038/s43018-022-00348-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Yang C. et al. (2023) Methionine orchestrates the metabolism vulnerability in cisplatin resistant bladder cancer microenvironment. Cell Death & Disease 14. ARTN 525 10.1038/S41419-023-06050-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Roy DG et al. (2020) Methionine Metabolism Shapes T Helper Cell Responses through Regulation of Epigenetic Reprogramming. Cell Metabolism 31, 250-+. 10.1016/j.cmet.2020.01.006 [DOI] [PubMed] [Google Scholar]
  • 68.Pandit M. et al. (2023) Methionine consumption by cancer cells drives a progressive upregulation of PD-1 expression in CD4 T cells. Nat Commun 14, 2593. 10.1038/s41467-023-38316-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Cheng H. et al. (2023) Extracellular acidosis restricts one-carbon metabolism and preserves T cell stemness. Nat Metab 5, 314–330. 10.1038/s42255-022-00730-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Dilek N. et al. (2020) Hydrogen sulfide: An endogenous regulator of the immune system. Pharmacol Res 161, 105119. 10.1016/j.phrs.2020.105119 [DOI] [PubMed] [Google Scholar]
  • 71.Castellano F and Molinier-Frenkel V. (2020) Control of T-Cell Activation and Signaling by Amino-Acid Catabolizing Enzymes. Front Cell Dev Biol 8, 613416. 10.3389/fcell.2020.613416 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Mak TW et al. (2017) Glutathione Primes T Cell Metabolism for Inflammation. Immunity 46, 675–689. 10.1016/j.immuni.2017.03.019 [DOI] [PubMed] [Google Scholar]
  • 73.Miller TW et al. (2012) Hydrogen sulfide is an endogenous potentiator of T cell activation. J Biol Chem 287, 4211–4221. 10.1074/jbc.M111.307819 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Carbonero F. et al. (2012) Microbial pathways in colonic sulfur metabolism and links with health and disease. Front Physiol 3, 448. 10.3389/fphys.2012.00448 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Huang J. et al. (2023) Modulation of gut microbiota: a novel approach to enhancing the effects of immune checkpoint inhibitors. Ther Adv Med Oncol 15, 17588359231204854. 10.1177/17588359231204854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Saini N. et al. (2022) Methionine uptake via the SLC43A2 transporter is essential for regulatory T-cell survival. Life Sci Alliance 5. 10.26508/lsa.202201663 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yang R. et al. (2015) Hydrogen Sulfide Promotes Tet1- and Tet2-Mediated Foxp3 Demethylation to Drive Regulatory T Cell Differentiation and Maintain Immune Homeostasis. Immunity 43, 251–263. 10.1016/j.immuni.2015.07.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Srivastava MK et al. (2010) Myeloid-derived suppressor cells inhibit T-cell activation by depleting cystine and cysteine. Cancer Res 70, 68–77. 10.1158/0008-5472.CAN-09-2587 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Hung MH et al. (2021) Tumor methionine metabolism drives T-cell exhaustion in hepatocellular carcinoma. NatCommun 12, 1455. 10.1038/s41467-021-21804-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Orillion A. et al. (2018) Dietary Protein Restriction Reprograms Tumor-Associated Macrophages and Enhances Immunotherapy. Clin Cancer Res 24, 6383–6395. 10.1158/1078-0432.CCR-18-0980 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Morehead LC et al. (2023) Increased Response to Immune Checkpoint Inhibitors with Dietary Methionine Restriction in a Colorectal Cancer Model. Cancers (Basel) 15. 10.3390/cancers15184467 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Joulia E and Metallo CM (2023) Methionine and H(2)S alter cancer-immune dialogue. Nat Metab 5, 1456–1458. 10.1038/s42255-023-00862-3 [DOI] [PubMed] [Google Scholar]
  • 83.Richie JP et al. (2023) Dietary Methionine and Total Sulfur Amino Acid Restriction in Healthy Adults. Journal of Nutrition Health & Aging 27, 111–123. 10.1007/s12603-023-1883-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Huang TH et al. (2014) A methionine-restricted diet and endurance exercise decrease bone mass and extrinsic strength but increase intrinsic strength in growing male rats. J Nutr 144, 621–630. 10.3945/jn.113.187922 [DOI] [PubMed] [Google Scholar]
  • 85.Epner DE et al. (2002) Nutrient intake and nutritional indexes in adults with metastatic cancer on a phase I clinical trial of dietary methionine restriction. Nutr Cancer 42,158–166. 10.1207/S15327914NC422_2 [DOI] [PubMed] [Google Scholar]
  • 86.Durando X. et al. (2008) Optimal methionine-free diet duration for nitrourea treatment: a Phase I clinical trial. Nutr Cancer 60, 23–30. 10.1080/01635580701525877 [DOI] [PubMed] [Google Scholar]
  • 87.Thivat E. et al. (2009) Phase II trial of the association of a methionine-free diet with cystemustine therapy in melanoma and glioma. Anticancer Res 29, 5235–5240 [PubMed] [Google Scholar]
  • 88.Durando X. et al. (2010) Dietary methionine restriction with FOLFOX regimen as first line therapy of metastatic colorectal cancer: a feasibility study. Oncology 78, 205–209. 10.1159/000313700 [DOI] [PubMed] [Google Scholar]

RESOURCES