Abstract
The potential for “anti-cancer” diets to markedly alter cancer risk and prognosis has captured the imagination of patients, physicians, and researchers alike, but many of these dietary recommendations come from correlative studies that attribute certain diets to altered cancer risk. While provocative, little is known about the molecular mechanisms behind how these dietary interventions might impact cancer progression. Within this context, however, changes in tumor lipid metabolism are emerging as a key contributor. In this review, we examine the current understanding of lipid metabolism in the tumor microenvironment, suggesting how diet-induced changes in lipid composition may regulate tumor progression and therapeutic efficacy. By dissecting various cellular pathways involved in lipid metabolism, we highlight how diet modulates the balance between saturated and unsaturated fatty acid species in tumors to impact cancer cell and stromal cell function. Finally, we describe how current cancer therapies may synergize with diet to improve therapeutic efficacy.
Graphical Abstract

Introduction
The possibility of using “anti-cancer” dietary interventions to prevent cancer onset or improve cancer prognosis is enthusiastically embraced by patients, clinicians, and researchers alike. Unfortunately, while dietary choices are known to contribute to cancer risk [1], the impacts of implementing dietary modifications after diagnosis to improve survival rates remains understudied. As many cancer patients are only motivated to change their diet after receiving a diagnosis, differentiating between prevention and prognosis is key. In addition, because any dietary advice must work in concert with standard-of-care cancer treatment, understanding how diet interacts with cancer therapies is also crucial [2,3]. Importantly, the heterogeneity of cancer (e.g., multiple cancer cell types, genetic alterations, and therapeutic options) makes individualized precision nutrition necessary to achieve optimal patient prognosis. Implementing this vision requires translational studies that test how diet-induced molecular changes synergize with different therapies to target specific tumor types. It is therefore critical to define the molecular mechanisms underlying the effects of diet on cancer progression and therapy responses.
Since environmental metabolite levels constrain cellular metabolic activity [4], recent studies have focused on how dietary modifications change metabolite levels within the tumor microenvironment (TME) to impact tumor metabolism, growth, and responses to therapy [2,5]. In particular, diet-induced changes to lipid metabolism have emerged as an important modulator of cancer progression [6,7]. Lipids contribute to key biological processes, including cellular membrane synthesis, energy generation, and signal transduction [8–10]. Fatty acids (FAs) – a major building block of complex lipids – are characterized by the number of double bonds they contain, including: saturated FAs (SFA, 0 double bonds), monounsaturated FAs (MUFA, 1 double bond), or polyunsaturated FAs (PUFA, 2+ double bonds). The degree to which cellular FAs are saturated is tightly regulated to maintain membrane fluidity and cellular function, with cancer cells altering FA desaturation pathways to improve their growth and survival [11–13]. In this review, we will discuss how various diets can impact tumor progression by controlling the amounts and types of FAs delivered to the TME, which directly alters lipid metabolism pathways within tumors. Specifically, we will focus on emerging evidence that diet can impact cancer progression and therapy by disrupting the balance between SFAs and MUFAs/PUFAs within tumors.
The degree of cellular fatty acid saturation is tightly regulated
Many cancer cells upregulate the synthesis of FAs during rapid cell division to support the production of complex lipids that make up cellular membranes (for a more in-depth review, see ref [8–10]). Mammalian cells use fatty acid synthase (FASN) for de novo synthesis of the SFA palmitate (16:0) from glucose and glutamine-derived acetyl-CoA and malonyl-CoA (Figure 1). 16:0 undergoes further elongation by FA elongases (ELOVL1–7) or desaturation by stearoyl-CoA desaturase (SCD) to produce palmitoleate (16:1(n-7)). Analogously, SCD converts the SFA stearate (18:0) into oleate (18:1(n-9)). These MUFAs can then be further elongated to generate longer-chain MUFAs. In contrast, omega-6 (n-6) and omega-3 (n-3) PUFAs cannot be de novo synthesized by mammalian cells and must be taken up from the environment. The most upstream PUFAs are linoleic acid (18:2(n-6)) and alpha-linolenic acid (18:3(n-3)), which, after being imported, are elongated by ELOVL2/5 and further desaturated by fatty acid desaturase 1/2 (FADS1/2) to produce long-chain, highly unsaturated PUFAs (Figure 1).
Figure 1. Schematic of cellular fatty acid metabolism pathways.

De novo fatty acid synthesis (top) uses carbons from acetyl-CoA, which is derived from nutrients such as glucose and glutamine, to produce saturated fatty acids (SFA) and monounsaturated fatty acids (MUFA). Essential fatty acid metabolism (bottom) elongates and desaturates diet-derived polyunsaturated fatty acids (PUFA). Fatty acids are designated X:Y(n-Z), where X indicates the number of carbons, Y indicates the number of double bonds, and n-Z (or omega-Z) indicates the position of the double bond closest to the methyl end of the fatty acid. ACC: acetyl-CoA carboxylase, FASN: fatty acid synthase, SCD: stearoyl-CoA desaturase, ELOVL1–7: elongation of very long chain fatty acids protein 1–7, FADS1/2: fatty acid desaturase 1/2, β-ox: β-oxidation.
These pathways are regulated, in part, by extracellular lipid availability. The presence of unsaturated FAs inhibits both the transcription and proteolytic activation of sterol regulatory element-binding proteins (SREBPs), which themselves control the expression of de novo FA synthesis and PUFA metabolism genes [14]. Conversely, environmental lipid limitation stimulates de novo FA synthesis [15,16]. Importantly, tight regulation of these pathways controls cellular FA saturation to maintain membrane fluidity and cellular function and to promote cell proliferation and survival [11]. For example, excess 16:0 is toxic to cells because increased cellular FA saturation disrupts the endoplasmic reticulum (ER) membrane, which causes ER stress and activates the unfolded protein response (UPR). How UPR signaling resolves lipotoxic stress remains poorly understood, but prolonged UPR activation induces apoptosis [11,17]. SCD is critical for disposing of 16:0 by desaturating it into 16:1(n-7), particularly when FASN is stimulated under lipid starvation conditions. Inhibiting SCD in these conditions leads to 16:0 accumulation, inducing lipotoxicity and cell death [17,18••]. Moreover, because SCD is an oxygen-dependent enzyme, hypoxic stress also inhibits SCD and promotes increased FA saturation. Some cancer cells can overcome SCD inhibition by employing FADS2 to convert 16:0 into the 16:1(n-10) MUFA sapienate [12], while others, particularly clear cell renal cell carcinoma cells, counter this stressor by releasing MUFAs stored in lipid droplets into phospholipid pools [19]. Similarly, exogenous unsaturated FAs can protect cells against 16:0 toxicity by channeling 16:0 into triglyceride pools [20] or hindering de novo 16:0 synthesis [18••]. However, excess levels of some PUFAs in the environment, such as arachidonic acid (20:4(n-6)), can also be toxic to cells [21,22], in part through oxidation to lipid peroxides that trigger a form of cell death known as ferroptosis [23]. These examples illustrate how cells, in conjunction with the specific FAs that are available, must carefully control the activity of intracellular FA metabolism pathways to maintain the required balance of saturated versus unsaturated FAs.
The dietary fat type affects lipid composition in the TME
The types of lipids that are available to tumors depends on the type of fats contained in the diet, in part because different fats have distinct amounts of SFAs, MUFAs, and PUFAs. The American Cancer Society (ACS) recommends higher consumption of MUFAs and PUFAs, relative to SFAs [24]. Similarly, we recently showed that among pancreatic ductal adenocarcinoma (PDAC) patients in the Nurses’ Health Study and Health Professionals Follow-up Study, a dietary pattern lower in carbohydrates and higher in plant-based, but not animal-based, fat was associated with longer survival [18••]. Because plant-based fats contain more unsaturated FAs [25], these observations suggest that the quantity and composition of fats in low carbohydrate diets can influence their effects on PDAC progression.
Despite these associations with dietary choices, the mechanisms underlying how dietary fat composition alters the metabolism of saturated versus unsaturated FAs within tumors remain poorly defined. By conducting studies in mouse cancer models, where both dietary fat intake and composition can be manipulated (e.g. ~10% in a low fat, high carbohydrate diet to ~90% in a ketogenic diet (KD)) (Figure 2), we can gain important insights into these effects. In Table 1, we highlight the FA compositions of common dietary fats, including SFA-enriched palm oil and cocoa butter, MUFA-enriched lard and olive oil, and PUFA-enriched fish and soybean oils. Interestingly, a recent study showed that a ~60% high fat diet (HFD), formulated with distinct types of fats, differentially impacted weight gain, blood lipid levels, and the gut microbiome composition in mice [26•].
Figure 2. Dietary fat compositions can be altered to formulate different types of diets.

Macronutrient compositions of diets with different percentages of dietary fat, including caloric restriction (CR), a low fat diet (10% fat), a western diet (45% fat), a high fat diet (60% fat), and a ketogenic diet (KD, 90% fat).
Table 1.
Approximate fatty acid compositions of common dietary fats.
| Fatty Acid | Milk Fat | Palm Oil | Cocoa Butter | Lard | Olive Oil | Fish Oil | Soybean Oil | Flaxseed Oil |
|---|---|---|---|---|---|---|---|---|
| 4:0 | 2 | - | - | - | - | - | - | - |
| 6:0 | 1.5 | - | - | - | - | - | - | - |
| 8:0 | 1 | - | - | - | - | - | - | - |
| 10:0 | 2.5 | - | - | - | - | - | - | - |
| 12:0 | 3 | - | - | - | - | - | - | - |
| 14:0 | 11 | 1 | <0.5 | 1 | - | 9 | - | - |
| 16:0 | 29 | 45 | 26 | 23 | 12 | 16 | 11 | 5 |
| 16:1(n-7) | 1.5 | <0.5 | <0.5 | 2 | 1 | 12 | - | - |
| 18:0 | 13 | 4.5 | 35 | 13 | 3 | 3 | 4 | 4 |
| 18:1(n-9) | 24 | 39 | 34 | 39 | 72 | 10.5 | 23 | 19.5 |
| 18:2(n-6) | 3 | 9.5 | 3 | 18 | 9 | 2 | 53 | 15.5 |
| 18:3(n-3) | 0.5 | <0.5 | <0.5 | 1 | 1 | 1.5 | 8 | 56 |
| 18:4(n-3) | - | - | - | - | - | 3 | - | - |
| 20:4(n-6) | <0.5 | - | - | <0.5 | - | 1.5 | - | - |
| 20:5(n-3) | - | - | - | - | - | 13.5 | - | - |
| 22:5(n-3) | - | - | - | - | - | 1 | - | - |
| 22:6(n-3) | - | - | - | - | - | 10.5 | - | - |
| SFA | 63 | 50.5 | 62 | 37 | 15 | 28 | 15 | 9 |
| MUFA | 29 | 39 | 34 | 41 | 73 | 22.5 | 23 | 19.5 |
| PUFA | 4 | 9.5 | 3 | 19 | 10 | 33 | 61 | 71.5 |
Values expressed as % of total fatty acids. Data adapted from Envigo Teklad.
Using mass spectrometry-based metabolomics and lipidomics methods, the tumor interstitial fluid (TIF), which closely mirrors the nutrient milieu experienced by tumor cells, can be analyzed to provide a direct measurement of how different diets alter metabolite and lipid levels in the TME [27]. For example, a 60% lard-based HFD increases diacylglyceride (DAG) and triacylglyceride (TAG) levels but decreases free FA levels in the TIF [28••]. Similarly, we demonstrated that a caloric restriction (CR) diet decreases, while a KD increases, FA availability in the TME [18••]. Critically, changing the fat type alters the plasma FA composition in a manner that reflects the SFA, MUFA, and PUFA composition of the diet. For example, palm oil is enriched with both 16:0 and 18:1(n-9) (Table 1), and a KD with palm oil raises 16:0 and overall SFA levels in the blood and in tumors. Likewise, the decreased levels of circulating 16:0 and 18:1(n-9) induced by CR can be reversed by feeding a high fat CR diet formulated with palm oil; a similar CR diet with soybean oil, which is enriched in 18:2(n-6) (Table 1), does not increase these levels [18••].
Despite the importance of the fat type in the diet, many dietary studies using mouse cancer models do not take this factor into account. Nevertheless, it has become clear that diet-induced changes to lipid availability in the TME interact with tumor cell lipid metabolism to influence cancer progression. In the following sections, we highlight several recent studies that illustrate how dietary modifications alter the balance of saturated versus unsaturated FAs in tumors to impact tumor biology, arguing for better control of dietary SFA, MUFA, and PUFA compositions in future studies that explore the links between diet and cancer.
Dietary changes influence tumor progression by altering cancer cell lipid metabolism
Because cancer cells exhibit high glucose consumption (i.e., the Warburg effect) [29], low glycemic diets such as CR or the KD are commonly thought to inhibit tumor growth by limiting glucose availability [30]. However, we recently found that in subcutaneous mouse allograft models of PDAC and lung cancer, both CR and the KD lower blood glucose, but only CR inhibits tumor growth. Therefore, CR may act by altering the availability of other nutrients beyond glucose [18••]. As discussed above, CR, but not the KD, lowers FA levels in the TME, creating a lipid-depleted environment under CR in which SCD is required for cancer cell proliferation. However, CR also suppresses tumor SCD activity. Thus, CR tumors lack access to environmental MUFAs as well as the ability to produce them via SCD, leading to reduced MUFA/SFA ratios that impair tumor growth. Either overexpressing SCD in these cancer cells or feeding a high fat CR diet that raises circulating 18:1(n-9) levels reverses these effects. Interestingly, the KD also impairs tumor SCD, but does not impair tumor growth because the high fat composition of the KD increases MUFA availability in the TME, thus maintaining the tumor MUFA/SFA ratios. This can be reversed by feeding a KD high in 16:0-enriched palm oil, thus increasing tumor SFA levels, which in conjunction with decreased tumor SCD activity, slows tumor growth. Therefore, it is the diet-induced changes to tumor MUFA/SFA ratios that determine whether a low glycemic diet will impair tumor growth (Figure 3A) [18••].
Figure 3. Overview of dietary effects on cancer cell and stromal cell lipid metabolism.

(A) Dietary modifications alter lipid availability in the tumor microenvironment and drive imbalances in ratios of SFAs, MUFAs, and PUFAs, which can affect tumor growth. Caloric restriction (CR) decreases TME lipid availability and impairs tumor SCD activity, resulting in a disrupted MUFA/SFA ratio that inhibits tumor growth. Similarly, a PUFA-rich diet increases PUFA levels in tumors to induce ferroptosis. (B) Fatty acid availability contributes to cancer metastases. Melanoma cells metastasize more efficiently when circulating in the lymph versus the blood, in part because the lymph contains more oleate, which protects against oxidative stress and ferroptosis. Increased palmitate availability in pre-metastatic niches can also promote metastasis formation by promoting nuclear factor-kappaB (NF-kB) signaling. (C) The distribution of saturated or unsaturated fatty acids affects the function of tumor-associated immune cells. For example, to activate naïve T cells, phosphoinositides (PIs) enriched in PUFAs are required, whereas saturated PIs allows differentiated T cells to sustain proliferation and function. Fatty acid transport protein 2 (FATP2) regulates the immune suppressive function of polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) via accumulation of arachidonic acid and synthesis of prostaglandin E2 (PGE2). (D) Dietary effects on lipid metabolism can mediate crosstalk between cancer cells and non-cancer stromal cells within a tumor. For example, in the context of high fat diet-induced obesity, cancer cells outcompete CD8+ T cells at uptaking fatty acids from the TME, leading to impaired T cell function and accelerated tumor growth.
Additional studies have highlighted how specific FAs can promote tumor progression. For example, breast cancer cells generate a pre-metastatic niche in the lung characterized by increased 16:0 levels, which is then oxidized to acetyl-CoA to promote p65 acetylation and pro-metastatic nuclear factor-kappaB (NF-kB) signaling. Therefore, a HFD that is high in 16:0 further increases 16:0 levels at metastatic sites, promoting the growth of breast cancer metastases (Figure 3B) [31••]. Recent studies also showed that circulating melanoma cells in the blood experience oxidative stress, leading to lipid peroxides that induce ferroptosis and limit their metastatic potential [32•,33]. To avoid this fate, more melanoma cells circulate in the lymph, which contains higher levels of 18:1(n-9) that protect against ferroptosis [34]. As such, melanoma cells in the lymph survive longer while in circulation, allowing them to form distant metastases (Figure 3B) [32•]. While diet was not explored in this study, determining whether dietary fats that raise 18:1(n-9) levels in the blood promote metastases in certain cancer types will be important going forward.
While 18:1(n-9) generally protects against ferroptosis, increased uptake of PUFAs promotes ferroptosis because oxidized PUFA-containing phospholipids are responsible for triggering this form of cell death. Indeed, compared to an 18:1(n-9)-rich olive oil diet, a 22:6(n-3) fish oil diet increases omega-3 FA levels in blood and tumors, thereby inducing ferroptosis and impairing tumor growth (Figure 3A). Moreover, the 22:6(n-3)-rich diet enhances the effects of ferroptosis inducers, such as sulfasalazine [35••]. This suggests that using dietary modifications that increase PUFA levels and decrease MUFA availability in the TME may be an effective strategy for promoting ferroptosis for cancer therapy.
Stromal cells and cancer cells compete for lipid species within the TME
Interactions between cancer cell and non-cancer stromal cell lipid metabolism also influence tumor progression. For example, FAs released from subcutaneous adipocytes can promote the proliferation and invasion of adjacent melanoma cells [36]. Dietary factors can also influence these cancer cell-stromal cell interactions. A recent study demonstrated that HFD-induced obesity accelerates the growth of some mouse tumor models by reducing the number and function of CD8+ T cells within tumors. The HFD increased FA uptake and oxidation in cancer cells, in part by lowering the expression of prolyl hydroxlase 3 (PHD3), a repressor of FA oxidation. As a result, decreased amounts of free FA were available in the TME for use by CD8+ T cells. By overexpressing PHD3 in these cancer cells, FA levels in the TME were restored, increasing T cell infiltration and blunting the growth promoting effects of the HFD. These data suggest that in the context of HFD-induced obesity, cancer cells outcompete T cells for FAs in the TME, which leads to impaired T cell function and accelerated tumor growth (Figure 3D) [[28••].
How dietary intake of saturated versus unsaturated FAs influences these interactions remains poorly explored; however, emerging evidence suggests that FA saturation regulates the function of tumor-associated immune cells. For example, naïve T cells are enriched in PUFA-containing phosphoinositides (PI) that are necessary for initiating T cell activation. Activated T cells then stimulate de novo synthesis of SFA-containing PI, which are required for signaling events that sustain T cell differentiation, proliferation, and cytokine production (Figure 3C) [37•]. In another case, polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), pathologically activated neutrophils that impair anti-tumor immunity, were found to elevate expression of fatty acid transport protein 2 (FATP2). FATP2 facilitates the uptake of 20:4(n-6) for conversion to the proinflammatory cytokine prostaglandin E2, thereby supporting the immunosuppressive activity of PMN-MDSCs. Pharmacological inhibition of FATP2 blocks 20:4(n-6) uptake and synergizes with immunotherapy to impair tumor growth (Figure 3C) [38•]. Together, these results suggest that it will be important to test whether diet-mediated changes to SFA versus PUFA availability in the TME influence anti-tumor immunity, revealing whether dietary interventions could enhance immunotherapy efficacy.
Clinical Implications and Conclusions
Even though some diets can slow tumor growth on their own, it is unlikely that any single diet will be sufficient to induce tumor regression. Moreover, dietary recommendations must be made within the context of each patient’s specific tumor and the therapies they are receiving. Therefore, it is important to identify how dietary interventions can be leveraged to enhance cancer therapies. For example, CR and fasting can improve tumor sensitivity to various chemotherapies and immunotherapies [30,39,40], and the KD can improve the efficacy of PI3K inhibitors in some contexts [41]. Recent studies have also shown that a KD enhances the efficacy of cytotoxic chemotherapies, such as nab-paclitaxel, gemcitabine, and cisplatin, on PDAC tumors in mice. These effects were associated with enhanced ketone body utilization, increased redox stress, inflammatory gene expression, and altered FA composition within tumor cells [42•,43•]. Conversely, other diets, such as a HFD, decrease the efficacies of chemotherapies in various mouse models [44,45].
How these diets influence responsiveness to drug therapies, especially through effects on lipid metabolism, remains undefined. However, some studies suggest that cancer cell FA composition can indeed influence chemotherapy sensitivity. For example, high FASN expression in PDAC cells is associated with poor responsiveness to gemcitabine, and pharmacological inhibition of FASN synergizes with gemcitabine to impair PDAC tumor growth [46]. Interestingly, tumors with high FASN activity exhibit increased FA saturation, while inhibiting FASN shifts cells towards increased polyunsaturation. In the latter case, the PUFA-rich cellular membranes exhibit altered membrane dynamics that promote the uptake of some chemotherapeutics, thereby increasing cellular sensitivity to these drugs [47]. A recent study demonstrated that in fasting mice and humans, erythrocyte membranes exhibited decreased SFA and MUFA, but increased PUFA, compositions, and the degree of these changes were correlated with increased sensitivity of normal tissues to chemotherapy toxicity [48••]. Finally, altering the fatty acid composition of a KD was recently shown to influence the response of a human head and neck cancer xenograft to radiation therapy in mice [49••]. These initial studies emphasize the important role that FAs play in tumor responses to chemotherapies, but much still remains unknown.
Finally, it is important to note that differences in lipid metabolism between sexes as well as among different species may have implications for future translational studies. For example, triglyceride and free fatty acid levels are reported to be higher in human males compared to females under basal conditions [50,51]. Further, while the enzymes that participate in fatty acid metabolism are largely similar between humans and mice, cholesterol is generally transported by LDL in humans, whereas rodents use HDL for cholesterol transport [52]. These observations suggest that further study is warranted to determine how these and other biological variables impact lipid metabolism before clinically relevant dietary interventions will be possible.
In summary, cellular FA saturation is an important regulator of tumor cell biology that influences disease progression and therapy responses. As we define how diets containing distinct fat types modulate TME lipid availability, tumor lipid metabolism, and the balance between saturated and unsaturated FAs in tumors, we will be able to identify strategies for using dietary interventions to enhance the efficacies of cancer therapies. These studies will also impact the development of lipid metabolism inhibitors, including those for FASN and SCD, as a treatment for various cancers [10]. Elucidating the molecular mechanisms underlying potential synergistic dietdrug combinations will provide scientific evidence that can benefit patients and physicians, providing guidance on how best to incorporate diet and nutrition into cancer therapy.
Highlights.
Dietary changes can perturb cancer progression by altering tumor lipid metabolism
Changes to dietary fat type can impact the lipid composition in the tumor microenvironment
Stromal cells and cancer cells compete for lipid species within the tumor microenvironment
Diet-induced dysregulation of saturated versus unsaturated fatty acid metabolism influences tumor growth and therapy responses
Dietary interventions may improve therapeutic efficacy in cancer patients
Acknowledgements
This work was supported by the National Institutes of Health (R00CA255928 to E.C.L.) and the Van Andel Institute Metabolism and Nutrition (MeNu) Program.
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 Competing Interest
The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.
References and recommended reading
Papers of particular interest, published within the period of review, have been highlighted as:
• of special interest
•• of outstanding interest
- 1.Mayne ST, Playdon MC, Rock CL: Diet, nutrition, and cancer: past, present and future. Nat Rev Clin Oncol 2016, 13:504–15. [DOI] [PubMed] [Google Scholar]
- 2.Muir A, Vander Heiden MG: The nutrient environment affects therapy. Science 2018, 360:962–963. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Kanarek N, Petrova B, Sabatini DM: Dietary modifications for enhanced cancer therapy. Nature 2020, 579:507–517. [DOI] [PubMed] [Google Scholar]
- 4.Muir A, Danai LV, Vander Heiden MG: Microenvironmental regulation of cancer cell metabolism: implications for experimental design and translational studies. Dis Model Mech 2018, 11:dmm035758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Lien EC, Vander Heiden MG: A framework for examining how diet impacts tumour metabolism. Nat Rev Cancer 2019, 19:651–661. [DOI] [PubMed] [Google Scholar]
- 6.Peck B, Schulze A: Lipid Metabolism at the Nexus of Diet and Tumor Microenvironment. Trends Cancer 2019, 5:693–703. [DOI] [PubMed] [Google Scholar]
- 7.Prendeville H, Lynch L: Diet, lipids, and antitumor immunity. Cell Mol Immunol 2022, 19:432–444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Snaebjornsson MT, Janaki-Raman S, Schulze A: Greasing the Wheels of the Cancer Machine: The Role of Lipid Metabolism in Cancer. Cell Metab 2020, 31:62–76. [DOI] [PubMed] [Google Scholar]
- 9.Hoy AJ, Nagarajan SR, Butler LM: Tumour fatty acid metabolism in the context of therapy resistance and obesity. Nat Rev Cancer 2021, 21:753–766. [DOI] [PubMed] [Google Scholar]
- 10.Yoon H, Shaw JL, Haigis MC, Greka A: Lipid metabolism in sickness and in health: Emerging regulators of lipotoxicity. Mol Cell 2021, 81:3708–3730. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Peck B, Schulze A: Lipid desaturation - the next step in targeting lipogenesis in cancer? FEBS J 2016, 283:2767–2778. [DOI] [PubMed] [Google Scholar]
- 12.Vriens K, Christen S, Parik S, Broekaert D, Yoshinaga K, Talebi A, Dehairs J, Escalona-Noguero C, Schmieder R, Cornfield T, et al. : Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity. Nature 2019, 566:403–406. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Triki M, Rinaldi G, Planque M, Broekaert D, Winkelkotte AM, Maier CR, Janaki Raman S, Vandekeere A, Van Elsen J, Orth MF, et al. : mTOR Signaling and SREBP Activity Increase FADS2 Expression and Can Activate Sapienate Biosynthesis. Cell Rep 2020, 31:107806. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Ye J, DeBose-Boyd RA: Regulation of cholesterol and fatty acid synthesis. Cold Spring Harb Perspect Biol 2011, 3.7:a004754. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Röhrig F, Schulze A: The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer 2016, 16:732–749. [DOI] [PubMed] [Google Scholar]
- 16.Shao W, Espenshade PJ: Expanding Roles for SREBP in Metabolism. Cell Metab 2012, 16:414–419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Piccolis M, Bond LM, Kampmann M, Pulimeno P, Chitraju C, Jayson CBK, Vaites LP, Boland S, Lai ZW, Gabriel KR, et al. : Probing the Global Cellular Responses to Lipotoxicity Caused by Saturated Fatty Acids. Mol Cell 2019, 74:32–44.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.••.Lien EC, Westermark AM, Zhang Y, Yuan C, Li Z, Lau AN, Sapp KM, Wolpin BM, Vander Heiden MG: Low glycaemic diets alter lipid metabolism to influence tumour growth. Nature 2021, 599:302–307. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates how caloric restriction (CR) and the ketogenic diet (KD) alter lipid availability in the TME and impair tumor SCD activity, resulting in imbalances in MUFA/SFA ratios that perturb tumor growth.
- 19.Ackerman D, Tumanov S, Qiu B, Michalopoulou E, Spata M, Azzam A, Xie H, Simon MC, Kamphorst JJ: Triglycerides Promote Lipid Homeostasis during Hypoxic Stress by Balancing Fatty Acid Saturation. Cell Rep 2018, 24:2596–2605.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Listenberger LL, Han X, Lewis SE, Cases S, Farese RV, Ory DS, Schaffer JE: Triglyceride accumulation protects against fatty acid-induced lipotoxicity. Proc Natl Acad Sci U S A 2003, 100:3077–3082. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Zhu XG, Nicholson Puthenveedu S, Shen Y, La K, Ozlu C, Wang T, Klompstra D, Gultekin Y, Chi J, Fidelin J, et al. : CHP1 Regulates Compartmentalized Glycerolipid Synthesis by Activating GPAT4. Mol Cell 2019, 74:45–58.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Pompeia C, Freitas JJS, Kim JS, Zyngier SB, Curi R: Arachidonic acid cytotoxicity in leukocytes: implications of oxidative stress and eicosanoid synthesis. Biol Cell 2002, 94:251–265. [DOI] [PubMed] [Google Scholar]
- 23.Dixon SJ, Stockwell BR: The Hallmarks of Ferroptosis. Annu Rev Cancer Biol 2019, 3:35–54. [Google Scholar]
- 24.Kushi LH, Doyle C, McCullough M, Rock CL, Demark-Wahnefried W, Bandera EV, Gapstur S, Patel AV, Andrews K, Gansler T, et al. : American Cancer Society guidelines on nutrition and physical activity for cancer prevention: Reducing the risk of cancer with healthy food choices and physical activity. CA Cancer J Clin 2012, 62:30–67. [DOI] [PubMed] [Google Scholar]
- 25.Coulston AM: The role of dietary fats in plant-based diets. Am J Clin Nutr 1999, 70:512S–515S. [DOI] [PubMed] [Google Scholar]
- 26.•.An J, Wang Q, Yi S, Liu X, Jin H, Xu J, Wen G, Zhu J, Tuo B: The source of the fat significantly affects the results of high-fat diet intervention. Sci Rep 2022, 12:4315. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study, twelve different types of dietary fat were tested as part of a high fat diet intervention in mice. The mice showed distinct effects with regards to weight gain, blood lipid levels, and the composition of the gut microbiome, depending on the fat source.
- 27.Sullivan MR, Danai LV, Lewis CA, Chan SH, Gui DY, Kunchok T, Dennstedt EA, Vander Heiden MG, Muir A: Quantification of microenvironmental metabolites in murine cancers reveals determinants of tumor nutrient availability. eLife 2019, 8:e44235. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.••.Ringel AE, Drijvers JM, Baker GJ, Catozzi A, García-Cañaveras JC, Gassaway BM, Miller BC, Juneja VR, Nguyen TH, Joshi S, et al. : Obesity shapes metabolism in the tumor microenvironment to suppress anti-tumor immunity. Cell 2020, 183:1848–1866.e26. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper suggests that in the context of HFD-induced obesity, cancer cells outcompete CD8+ T cells for fatty acids in the TME, leading to impaired T cell function and accelerated tumor growth.
- 29.Vander Heiden MG, Cantley LC, Thompson CB: Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324:1029–1033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Nencioni A, Caffa I, Cortellino S, Longo VD: Fasting and cancer: molecular mechanisms and clinical application. Nat Rev Cancer 2018, 18:707–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.••.Altea-Manzano P, Doglioni G, Liu Y, Cuadros AM, Nolan E, Fernández-García J, Wu Q, Planque M, Laue KJ, Cidre-Aranaz F, et al. : A palmitate-rich metastatic niche enables metastasis growth via p65 acetylation resulting in pro-metastatic NF-κB signaling. Nat Cancer 2023, 4:344–364. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study demonstrates that a HFD enhances palmitate availability at metastatic sites, which promotes pro-metastatic NF-kB signaling and the growth of breast cancer metastases.
- 32.•.Ubellacker JM, Tasdogan A, Ramesh V, Shen B, Mitchell EC, Martin-Sandoval MS, Gu Z, McCormick ML, Durham AB, Spitz DR, et al. : Lymph protects metastasizing melanoma cells from ferroptosis. Nature 2020, 585:113–118. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper shows that melanoma cells metastasize more efficiently when circulating in the lymph versus the blood, in part because the lymph contains higher levels of 18:1(n-9) that protect against oxidative stress and ferroptosis.
- 33.Piskounova E, Agathocleous M, Murphy MM, Hu Z, Huddlestun SE, Zhao Z, Leitch AM, Johnson TM, DeBerardinis RJ, Morrison SJ: Oxidative stress inhibits distant metastasis by human melanoma cells. Nature 2015, 527:186–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Magtanong L, Ko P-J, To M, Cao JY, Forcina GC, Tarangelo A, Ward CC, Cho K, Patti GJ, Nomura DK, et al. : Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State. Cell Chem Biol 2019, 26:420–432.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.••.Dierge E, Debock E, Guilbaud C, Corbet C, Mignolet E, Mignard L, Bastien E, Dessy C, Larondelle Y, Feron O: Peroxidation of n-3 and n-6 polyunsaturated fatty acids in the acidic tumor environment leads to ferroptosis-mediated anticancer effects. Cell Metab 2021, 33:1701–1715.e5. [DOI] [PubMed] [Google Scholar]; This study shows how compared to an 18:1(n-9)-rich olive oil diet, a 22:6(n-3)-rich fish oil diet induces ferroptosis in tumors and enhances the effects of ferroptosis inducers.
- 36.Zhang M, Di Martino JS, Bowman RL, Campbell NR, Baksh SC, Simon-Vermot T, Kim IS, Haldeman P, Mondal C, Yong-Gonzales V, et al. : Adipocyte-Derived Lipids Mediate Melanoma Progression via FATP Proteins. Cancer Discov 2018, 8:1006–1025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.•.Edwards-Hicks J, Apostolova P, Buescher JM, Maib H, Stanczak MA, Corrado M, Klein Geltink RI, Maccari ME, Villa M, Carrizo GE, et al. : Phosphoinositide acyl chain saturation drives CD8+ effector T cell signaling and function. Nat Immunol 2023, 24:516–530. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows that the acyl chains on PI shift from PUFA-rich to SFA-rich during T cell activation, a necessary step for T cell differentiation, proliferation, and cytokine production.
- 38.•.Veglia F, Tyurin VA, Blasi M, De Leo A, Kossenkov AV, Donthireddy L, To TKJ, Schug Z, Basu S, Wang F, et al. : Fatty acid transport protein 2 reprograms neutrophils in cancer. Nature 2019, 569:73–78. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study, PMN-MDSCs are shown to upregulate FATP2 to import 20:4(n-6), which is converted to prostaglandin E2 to support their immunosuppressive activity. Pharmacological FATP2 inhibition was also shown to synergize with immunotherapy.
- 39.Vernieri C, Fucà G, Ligorio F, Huber V, Vingiani A, Iannelli F, Raimondi A, Rinchai D, Frigè G, Belfiore A, et al. : Fasting-Mimicking Diet Is Safe and Reshapes Metabolism and Antitumor Immunity in Patients with Cancer. Cancer Discov 2022, 12:90–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.de Groot S, Lugtenberg RT, Cohen D, Welters MJP, Ehsan I, Vreeswijk MPG, Smit VTHBM, de Graaf H, Heijns JB, Portielje JEA, et al. : Fasting mimicking diet as an adjunct to neoadjuvant chemotherapy for breast cancer in the multicentre randomized phase 2 DIRECT trial. Nat Commun 2020, 11:3083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hopkins BD, Pauli C, Du X, Wang DG, Li X, Wu D, Amadiume SC, Goncalves MD, Hodakoski C, Lundquist MR, et al. : Suppression of insulin feedback enhances the efficacy of PI3K inhibitors. Nature 2018, 560:499–503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.•.Yang L, TeSlaa T, Ng S, Nofal M, Wang L, Lan T, Zeng X, Cowan A, McBride M, Lu W, et al. : Ketogenic diet and chemotherapy combine to disrupt pancreatic cancer metabolism and growth. Med 2022, 3:119–136.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study, a ketogenic diet (KD) is shown to synergize with chemotherapies in a mouse model of pancreatic cancer. These effects are associated with changes to ketone body and redox metabolism.
- 43.•.Cortez NE, Rodriguez Lanzi C, Hong BV, Xu J, Wang F, Chen S, Ramsey JJ, Pontifex MG, Müller M, Vauzour D, et al. : A Ketogenic Diet in Combination with Gemcitabine Increases Survival in Pancreatic Cancer KPC Mice. Cancer Res Commun 2022, 2:951–965. [DOI] [PMC free article] [PubMed] [Google Scholar]; This paper demonstrates that a ketogenic diet (KD) enhances the effects of gemcitabine in a mouse model of pancreatic cancer. These effects are associated with changes to oncogenic cell signaling, fatty acid metabolism, and gut microbiota composition.
- 44.Mentoor I, Nell T, Emjedi Z, Van Jaarsveld PJ, De Jager L, Engelbrecht A-M: Decreased Efficacy of Doxorubicin Corresponds With Modifications in Lipid Metabolism Markers and Fatty Acid Profiles in Breast Tumors From Obese vs. Lean Mice. Front Oncol 2020, 10:306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kesh K, Mendez R, Mateo-Victoriano B, Garrido VT, Durden B, Gupta VK, Oliveras Reyes A, Merchant N, Datta J, Banerjee S, et al. : Obesity enriches for tumor protective microbial metabolites and treatment refractory cells to confer therapy resistance in PDAC. Gut Microbes 2022, 14:2096328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Tadros S, Shukla SK, King RJ, Gunda V, Vernucci E, Abrego J, Chaika NV, Yu F, Lazenby AJ, Berim L, et al. : De Novo Lipid Synthesis Facilitates Gemcitabine Resistance through Endoplasmic Reticulum Stress in Pancreatic Cancer. Cancer Res 2017, 77:5503–5517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rysman E, Brusselmans K, Scheys K, Timmermans L, Derua R, Munck S, Van Veldhoven PP, Waltregny D, Daniëls VW, Machiels J, et al. : De novo Lipogenesis Protects Cancer Cells from Free Radicals and Chemotherapeutics by Promoting Membrane Lipid Saturation. Cancer Res 2010, 70:8117–8126. [DOI] [PubMed] [Google Scholar]
- 48.••.Barradas M, Plaza A, Colmenarejo G, Lázaro I, Costa-Machado LF, Martín-Hernández R, Micó V, López-Aceituno JL, Herranz J, Pantoja C, et al. : Fatty acids homeostasis during fasting predicts protection from chemotherapy toxicity. Nat Commun 2022, 13:5677. [DOI] [PMC free article] [PubMed] [Google Scholar]; This study shows an association between the fasting-induced shifts in SFA/MUFA versus PUFA levels in erythrocyte membranes with the sensitivity of normal tissues to chemotherapy toxicity.
- 49.••.Ma DC, Anderson CM, Rodman SN, Buranasudja V, McCormick ML, Davis A, Loth E, Bodeker KL, Ahmann L, Parkhurst JR, et al. : Ketogenic Diet with Concurrent Chemoradiation in Head and Neck Squamous Cell Carcinoma: Preclinical and Phase 1 Trial Results. Radiat Res 2021, 196:213–224. [DOI] [PMC free article] [PubMed] [Google Scholar]; In this study, a commercially available KD (KetoCal) was shown to enhance the efficacy of radiation therapy on impairing the growth of a human head and neck cancer xenograft in mice. The fat composition of KetoCal was altered mid-study, and the new formulation no longer synergized with radiation.
- 50.Link JC, Reue K: Genetic Basis for Sex Differences in Obesity and Lipid Metabolism. Annu Rev Nutr 2017, 37:225–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Zore T, Palafox M, Reue K: Sex differences in obesity, lipid metabolism, and inflammation—A role for the sex chromosomes? Mol Metab 2018, 15:35–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bergen WG, Mersmann HJ: Comparative Aspects of Lipid Metabolism: Impact on Contemporary Research and Use of Animal Models. J Nutr 2005, 135:2499–2502. [DOI] [PubMed] [Google Scholar]
