Next Article in Journal
Omega-3 Fatty Acids and Their Interaction with the Gut Microbiome in the Prevention and Amelioration of Type-2 Diabetes
Previous Article in Journal
Perilla Fruit Oil-Fortified Soybean Milk Intake Alters Levels of Serum Triglycerides and Antioxidant Status, and Influences Phagocytotic Activity among Healthy Subjects: A Randomized Placebo-Controlled Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Exploring the Role of Metabolites in Cancer and the Associated Nerve Crosstalk

1
Department of Food Science, Division of Agriculture, University of Arkansas, Fayetteville, AR 72704, USA
2
Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, USA
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2022, 14(9), 1722; https://doi.org/10.3390/nu14091722
Submission received: 15 March 2022 / Revised: 12 April 2022 / Accepted: 15 April 2022 / Published: 21 April 2022
(This article belongs to the Section Micronutrients and Human Health)

Abstract

:
Since Otto Warburg’s first report on the increased uptake of glucose and lactate release by cancer cells, dysregulated metabolism has been acknowledged as a hallmark of cancer that promotes proliferation and metastasis. Over the last century, studies have shown that cancer metabolism is complex, and by-products of glucose and glutamine catabolism induce a cascade of both pro- and antitumorigenic processes. Some vitamins, which have traditionally been praised for preventing and inhibiting the proliferation of cancer cells, have also been proven to cause cancer progression in a dose-dependent manner. Importantly, recent findings have shown that the nervous system is a key player in tumor growth and metastasis via perineural invasion and tumor innervation. However, the link between cancer–nerve crosstalk and tumor metabolism remains unclear. Here, we discuss the roles of relatively underappreciated metabolites in cancer–nerve crosstalk, including lactate, vitamins, and amino acids, and propose the investigation of nutrients in cancer–nerve crosstalk based on their tumorigenicity and neuroregulatory capabilities. Continued research into the metabolic regulation of cancer–nerve crosstalk will provide a more comprehensive understanding of tumor mechanisms and may lead to the identification of potential targets for future cancer therapies.

1. Introduction

Cancer places a heavy burden on the global population as the second leading cause of death, with severe detriment in countries without access to sufficient medical care [1]. In 2020 alone, the World Health Organization’s GLOBOCAN reported that over 10 million men and 9 million women were diagnosed with some form of cancer [2]. The most prominent cancers among women include breast, colorectal, lung, cervical, and thyroid; while lung, prostate, colorectal, stomach, and liver cancers are the most common among men [2]. Though genetic factors may play a role in cancer onset, a heavy emphasis has been placed on the preventability of cancer, as highlighted by the increased probability of disease onset by smoking, sun exposure, and virus and bacterial incubation [3,4]. Over the last 20 years, increasing evidence has implicated diet as a major contributor in up to 20% of cancer cases [5]. This is made even more apparent by the variance in cancer dominance between countries and cultures [6] and the incidence of cancer among populations without access to proper nutrition [7].
The role of an altered metabolic state in which cells exhibit an increased conversion of glucose into lactate even in highly oxygenated areas, denoted the Warburg effect, has been an accepted hallmark of cancer since the 1920s [8,9]. This reliance on aerobic glycolysis leaves cells with a large deficiency in ATP and increased by-products such as reactive oxygen species (ROS) [9,10,11]. For cancer cells, maintaining ROS homeostasis via glutathione is essential, as moderate levels are proven to contribute to tumorigenesis and invasion while an overabundance leads to cell and DNA damage [10,12,13]. Moreover, increased ROS leads to upregulation of glutathione to maintain homeostasis; this increased defense can allow cancer cells to become resistant to therapies that rely on the induction of oxidative stress [14]. As the catabolism of glucose leaves them starved of energy, cancer cells can become dependent on glutamine for ATP [15,16,17,18]. Glutamine plays a vital role in macromolecule synthesis through the tricarboxylic acid (TCA) cycle (Figure 1) [16,19,20]. Therefore, glucose, glutamine, and their by-products are prevalent in studies searching for metabolic targets in cancer therapies [21,22,23,24].
Alternatively, researchers have come to acknowledge the role of lactate not only as a waste product but also as the driving force of mitochondrial respiration in oxidating cancer cells [25]. Thus, there has been continued interest in the investigation of metabolites in cancer progression. Amino-acid products of the TCA, urea, folate, and methionine cycles as well as those exogenously supplemented have proven important in facilitating cancer-cell protein and DNA synthesis as well as waste (e.g., ROS and nitric oxide synthase (NOS)), which progresses tumorigenesis [11,26]. Recent studies have targeted many amino acids to inhibit cancer progression, as many induce adaptive resistance to chemotherapy and radiation-therapy treatments [27,28,29,30,31,32]. In order to slow cancer progression, some clinicians have even suggested patients adhere to an “anti-cancer diet” to starve tumor cells of key nutrients [33,34]. However, many cancers alter their reliance on specific metabolites, rendering dietary restrictions ineffective, which has caused researchers to investigate pathways via targeted therapies [35,36]. Alternatively, some vitamins (e.g., vitamins A, C, and D) have been identified as antitumorigenic agents, presenting them as promising treatments in inhibiting tumor growth, while the upregulation of others (e.g., vitamin B12) is associated with decreased patient outcomes [37,38,39,40,41,42]. Continued examination of the contributions of lactate, amino acids, and vitamins in cancer mechanisms may provide more significant therapeutic options.
Within the past two decades, researchers have also acknowledged the interconnection of the nervous system with cancer progression and metastasis [43,44,45,46,47,48]. This cancer–nerve crosstalk occurs in cancers of highly innervated areas, including the pancreas [49,50,51,52], head and neck [53,54,55], colon and rectum [56,57,58], breasts [59,60,61,62], prostate [63,64,65], cervix [66,67,68], lungs [69], etc. There are two established subtypes of cancer–nerve crosstalk: perineural invasion (PNI) occurs when cancer cells migrate into the perineural layer of adjacent nerves [70,71,72] while its relative opposite, tumor innervation (TI), describes the phenomenon in which neurites extend into solid tumors [73,74]. PNI has been deemed a prognostic factor of select cancers, including cervical [68], liver [75], and colorectal [48], as well as a signifier of tumor recurrence in breast [61] and prostate cancers [76]. However, there still lack effective treatments of either form of the crosstalk, as the lack of sufficient pre-surgical screenings for PNI and TI complicate resection [77] and radiation and chemotherapeutic options are limited [78,79]. It is vital that there be continued exploration into the mechanisms of cancer–nerve crosstalk, as both PNI and TI contribute to progressive cancer-related pain, metastases, and worsened patient prognosis [74].
Previous studies have presented neurotransmitters and neurotrophins (e.g., substance P [80], nerve-growth factor (NGF) [81,82,83] and brain-derived neurotrophic factor (BDNF) [84]), chemokines (e.g., CX3CL1 [85] and CCL2 [86,87]), extracellular vesicles [66,88], and Schwann cells [52,89] as key players in cancer–nerve crosstalk. Moreover, glutamate, the product of glutaminolysis, acts as a potent central nervous system neurotransmitter that facilitates PNI in endometrial cancer [90] and TI in gliomas [57]. Glucose also works alongside neurotransmitters to modulate cancer metabolism and immune cell activity in the tumor microenvironment [91], while hyperglycemia in patients with type 2 diabetes mellitus correlates with an increased incidence of PNI in pancreatic cancer [92,93]. However, because there is presently a strong focus on targeting glutamine and glucose metabolism in cancer, this review will focus on the less-appreciated metabolites in cancer–nerve crosstalk that may be targeted to advance cancer therapeutics. As known contributors to cancer, nutrients should now be investigated in the context of cancer–nerve crosstalk.
Using literature obtained via PubMed and Google Scholar databases (published in English through March 2022), this review will highlight select nutrients and their anti- and/or pro-cancer effects on metabolism, invasion, and metastasis. As discussed above, there exist many reviews that highlight the value of glutamine, glucose, and their by-products in cancer; therefore, this review will focus on the roles other key nutrients play in cancer metabolism and aggressiveness. Additionally, we identify potential target metabolites as key players in cancer–nerve crosstalk based on their roles in cancer, as well as their known mechanistic contributions to neurite outgrowth and nerve regeneration. Further investigation into these nutrients will provide vital insights to develop targeted therapies and promote improved global patient outcomes.

2. Metabolites and Cancer

2.1. Background

In 1924, Otto Warburg published his findings, introducing dysregulated metabolism as a hallmark of cancer [94]. This altered metabolic state, aptly titled the Warburg effect, is described by the phenomenon in which cancer cells increase their uptake of glucose and production of lactate with a heavy reliance on aerobic glycolysis [94]. In this metabolic state, cancer cells produce very limited ATP via glycolysis and may become dependent on glutamine as an energy source [21]. Therefore, suppression of glucose and glutamine metabolism is heavily investigated in cancer therapeutics [21,22,23,24].
Lactate has gained a new appreciation for its contribution to oxidative respiration under normoxic conditions [95]. The discovery that lactate is both a waste product and vital tumor energy source highlights that metabolites other than glucose and glutamine may promote an environment fit for cancer-cell growth and proliferation. Downstream amino-acid by-products, e.g., asparagine [96], arginine [97], cysteine [98], serine [99], and glycine [100], have also been investigated for their contribution to the survival of cancer cells. While the deprivation of these nutrients appears to be an effective treatment in some cases [101,102], more investigation into therapies targeting each metabolic pathway is needed. Alternatively, some amino acids and key vitamins, e.g., vitamins A [37], B [103], D [104], E [105], K [106], and C [107], act as antitumorigenic factors and suppress cancer progression. To bring attention to these relatively underappreciated metabolites, this review highlights lactate, vitamins, and amino acids for their roles in progressing, suppressing, and preventing cancer (Table 1).

2.2. Lactate

Lactate is the major metabolite of glycolysis, which is catabolized from pyruvate by lactate dehydrogenase (LDH) (Figure 1) [94,137]. The primary isomer of lactate produced in humans is L-lactate, while D-lactate is synthesized at 1 to 5% the concentration of L-lactate [131,138]. Therefore, this review will focus on L-lactate unless stated otherwise. In healthy tissues, endogenous lactate levels are around 1.5 to 3 mM; however, under Warburg effect-like conditions, cancer cells contain 30 to 40 mM lactate [131]. In the past, lactate was recorded only as the waste product of glycolysis under hypoxic conditions; however, within the last 2 decades, researchers have gained an appreciation for its facilitation of oxidative metabolism in oxygenating cancer cells [25]. As such, when fed glucose-rich media with 10 mM sodium lactate supplementation, cervical cancer cells favored lactate and switched from glycolysis to lactate oxidation [25]. These cells continued to thrive following glucose starvation [25]. Conversely, glycolytic colorectal cancer cells had an adverse reaction when glucose was removed from their media [25]. In the oxidative cervical cancer cells, mRNA for monocarboxylate transporter-1 (MCT1), a protein that imports lactate, is transcribed at a higher rate than MCT4 mRNA, the protein exporter of lactate. MCT4 exhibited higher expression versus MCT1 in glycolytic colorectal cancer cells [25]. The study concluded that MCT1 and hypoxia are mutually exclusive in glycolytic tumors, and MCT1 inhibition induced lung carcinoma cell necrosis and sensitizes cells to irradiation in vivo [25]. One breast cancer study found that endogenous lactate receptor G protein-coupled receptor 81 (GPR81) mRNA expression is elevated in clinical estrogen receptor alpha-positive (ERα+) and human epidermal receptor-growth factor 2-positive (Her2+) breast cancer versus triple-negative breast cancer (TNBC) and benign tissue samples [139]. In vitro, GPR81 regulates MCT1 expression and lactate uptake in MCF-7 breast epithelial breast cancer cells and is responsible for cancer-cell proliferation and survival when lactate is the main nutrient source [139].
Bonuccelli et al. found that 10 mM L-lactate acted as a chemoattractant to almost double breast cancer-cell migration versus untreated cells [129]. In vivo, mice treated with L-lactate at 500 mg/kg of body weight (BW) increased lung metastases by 10.6-fold versus control mice [129]. Apicella et al. cultured non-small-cell lung cancer (NSCLC) tumors ex vivo before being injected into mice resistant to mesenchymal-to-epithelial transition (MET) tyrosine kinase inhibitors (TKI) [132]. This study found that resistant tumors upregulated lactate production and nuclear factor kappa-B (NF-κB), which induced hepatocyte growth factor (HGF) transcription by adjacent cancer-associated fibroblasts [133]. This pathway was shown to contribute to tumor resistance to MET TKI; stromal HGF and tumor MCT4 were also upregulated in epidermal growth-factor receptor TKI-resistant tumors of NSCLC patients [132].
In a genetic evaluation of in vitro breast cancer, MCF-7 cells were incubated with 0 to 20 mM exogenous lactate and analyzed for the expression of proto-oncogenes (i.e., NRAS, MYC, and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha PIK3CA), proliferative genes (i.e., ATM, CCND1, CDK4, CDK1A, CDK2b, AKT1, and MIF), tumor suppressors (i.e., BRCA1 and BRCA2), and transcription factors (i.e., HIF1A and E2F1) [130]. Following 48 h, cells treated with 4500 mg/L glucose (endogenous lactate) upregulated oncogenes, proliferative gene expression (except for MIF), tumor-suppressor genes, and transcription factors by over 2-fold [130]. Oncogenes, transcription factors, tumor suppressors, and proliferation genes were upregulated after 6 h of 10 mM treatment [130]. Expression was lower, yet still significant, after 48 h for all genes except for BRCA1 [130]. Following 6 h of 20 mM exogenous lactate therapy, MYC, transcription factors, tumor suppressors, and proliferation genes except ATM and CDK2b increased in expression [130]. Similarly, all but NRAS, PIK3CA, ATM, and CDK2b slightly decreased between 6 and 48 h [130]. The authors concluded that lactate changes cancer-cell transcription and acts as both a pro- and antitumorigenic factor in in vitro breast cancer [130].
A variety of therapies have been investigated in targeting glycolysis and lactate. Docetaxel-resistant prostate cancer cells were sensitized to chemotherapy following treatment with sodium oxamate, a structural analog to pyruvate which inhibits LDH and glycolysis [30]. The study found success in targeting LDH-A expression in vitro [30]. In papillary thyroid carcinoma patients, LDH-A mRNA and protein levels correlate with an aggressive phenotype and poor prognosis [140], while also associated with high glycolytic activity, radioresistance, and poor survival of NSCLC patients [133]. LDH-A promotes migration, proliferation, and epithelial-to-mesenchymal transition (EMT), regulates tumorigenicity and autophagy via the AMP-activated protein kinase (AMPK) pathway, and inhibits apoptosis of thyroid cancer in vitro and in vivo [140]. LDH-A inhibition sensitizes NSCLC cells to radiation therapy by blocking cellular energy metabolism and increasing X-ray-induced DNA injury via ROS production [133]. Inhibition, alone [140] and in conjunction with irradiation [134], induces apoptosis and autophagy of cancer cells.
Alternatively, D-lactate dimers (DLAD) have been found to be cytotoxic in some neuroblastoma and cervical cancer studies; therefore, Dikshit et al. utilized DLAD 3-(4,5- dimethylthiazol-2-yl)-2,5 diphenyl tetrazolium bromide to evaluate melanoma cell growth in dose-dependent manner (1.25 to 10 mg/mL) [131]. Following only 1 day of 10 mg/mL treatment, melanoma cells experienced complete cytotoxicity [131]. DLAD was found to contribute to dose-dependent inhibition of mitochondrial function, with inhibitory effects on cell growth even with pH neutralization [131]. In vivo, melanoma xenografts regressed in size and showed significantly decreased proliferation over a 21-day period. Furthermore, immunohistochemistry for innate immune cells indicated a significant increase in immune response and tumor growth inhibition [131]. With minimal metabolic effect on fibroblasts versus squamous-cell carcinoma cells in vitro, there exists promise for DLAD treatment in targeting tumor lactate metabolism [131].

2.3. Vitamin A

Vitamin A is a group of fat-soluble retinols and their derivatives, including retinyl ester, retinal, retinoic acid, and pro-vitamin A carotenoids, including α- and β-carotene [108]. Retinol is mostly acquired from the diet in precursor forms, e.g., retinyl esters from animal sources and pro-vitamin A carotenoids from plants [141].
Since retinoids have an important role in regulating cell growth, proliferation, and differentiation, studies have investigated the correlation between vitamin A and cancer patient prognosis [142,143]. A meta-analysis with 31 studies highlighted an inverse association between vitamin A intake and gastric cancer risk [108]. Another meta-analysis of 17 studies reported that higher blood concentrations of α- and β-carotene, total carotenoids, and retinol are significantly associated with decreased lung cancer risk [109]. Fulan et al. found an inverse association between the total intake of vitamin A/retinol and breast cancer risk [105]. Both natural and synthetic retinoids have been shown to exert chemotherapeutic effects in cancer with antiproliferative, proapoptotic and antioxidant activities [37]. Synthetic retinoid derivative fenretinide (4-HPR) has shown succuss in tumor-cell cytotoxicity in many cancers such as breast, prostate, colon, head and neck, and lung [37].

2.4. Vitamin B

Cobalamin (vitamin B12) is a water-soluble vitamin that is abundant in foods of animal origin [144]. Vitamin B12 can also be supplemented in foods in which it is not naturally synthesized. For humans to absorb vitamin B12 from food, hydrochloric acid in the stomach must separate B12 from the protein to which it is attached and then should combine with intrinsic factors to be absorbed by the body [145]. Analogs of cobalamin include: methylcobalamin (MeCbl), adenosylcobalamin (AdCbl), hydroxycobalamin (OHCbl), and cyanocobalamin (CNCbl). MeCbl and 5-deoxyadenosylcobalamin are active in human metabolism, while OHCbl is found in foods and is used as the first line of treatment for cyanide poisoning. CNCbl is a synthetic B12 analog used in food fortification and supplementation [146]. Vitamin B12 plays an important role in two biochemical reactions in humans: the formation of methionine by methylation of homocysteine (Figure 1), by which tetrahydrofolate, a precursor of purine and pyrimidine synthesis, is produced [147] as well as the conversion of L-methylmalonyl coenzyme A into succinyl coenzyme A, which is essential for the formation of the neuronal myelin sheath [148].
There has been some evidence of a relationship between intake of vitamin B and cancer [149]. In a review of the United Kingdom’s Health Improvement Network primary-care database, elevated plasma B12 levels were linked with a higher 1-year cancer risk, particularly for myeloid, liver, and pancreatic cancers. This suggests that some cancers may affect B12 metabolism [113]. Cancer caused by chromosomal breaks from the incorporation of uracil instead of the appropriate base is linked to folate deficiency and potentially vitamin B12 and B6 deficiencies [150]. In a nested study, higher blood B12 levels were associated with increased overall lung cancer risk [114]. One meta-analysis, however, showed that vitamins B1, B3, B6, and B9 are associated with reduced risk of esophageal cancer, while vitamin B12 was associated with induced esophageal cancer risk [103].
Furthermore, the decrease in thiamine (vitamin B1) transporter gene SLC9A3 has been evident in in vitro breast cancer cells as well as breast and gastric tumors [111,112,151]. Ng et al. highlighted a significant increase in plasma SLC9A3 methylation levels in cancer patients versus healthy participants, resulting in thiamine deficiency in patients [112]. In an in vivo study of breast cancer, mice were supplemented with 0 to 2500 times the recommended daily amount to combat this deficiency; mice given 12.5 to 75 times the recommended dose exhibited a significant increase in cancer-cell proliferation versus untreated mice [110]. However, it was also noted that oversupplementation, i.e., mice given 2500 times the recommended dosage, saw a decrease in tumor growth, suggesting the potential of thiamine overdose as a cancer therapy [110].
Thiamine is a coenzyme of pyruvate dehydrogenase (PDH), which converts pyruvate into acetyl-co-A to connect glycolysis and the TCA cycle [152,153]. However, similar to thiamine, the PDH subunit PDH-E1β is downregulated in breast cancer and HeLa cervical cancer cells as well as in vitro mouse fibroblasts under prolonged hypoxia [154]. This reduction in PDH was found to induce aerobic glycolysis and lactate production [154]. Therefore, the authors concluded that the activation status of PDH-E1β informs cancers cells whether to perform in a Warburg effect-like metabolic state (when silenced) or to enhance tumor growth (when upregulated) [154]. Interestingly, the group also found that bladder, melanoma, ovarian, and prostate cancer patients presented with high ratios of PDH-E1β, with prostate cancer patients experiencing up to a 50% amplification in total PDH expression.

2.5. Vitamin C

Ascorbic acid (vitamin C) is a water-soluble compound rich in fruits and vegetables. Unlike other species, humans need to consume vitamin C from external sources, as humans do not have gulonolactone oxidase to synthesize endogenous vitamin C [155]. Vitamin C is an important antioxidant and reducing cofactor in many enzymatic reactions related to collagen synthesis, tyrosine metabolism, hypoxia-inducible transcription factor (HIF) regulation, and epigenetic regulation of histone and DNA demethylation [156].
Due to its antioxidant activities, vitamin C has been widely researched as an anticancer agent for decades [38]. However, the use of vitamin C is cancer therapeutics is limited, as its anticancer effects are only observed in vitro [38,39]. However, it was recently shown that a high dose of intravenous vitamin C selectively reduces the proliferation and growth of cancer cells, with limited interference in healthy cell metabolism [108,157,158]. Yang et al. showed that high concentrations (1 to 5 mM) of vitamin C significantly inhibit cell proliferation and increase apoptosis in melanoma cells, while low concentrations (100 µM) support cell growth and migration [116]. Daily oral administration of low concentrations of vitamin C (0.0075 g/g of BW) promotes significant melanoma tumor growth in mice (p < 0.05) compared to control mice [116]. Zeng et al. treated breast cancer cells with low, intermediate, and high doses of vitamin C (0.01 to 2 mM) and demonstrated that low and intermediate doses induce cell migration and invasion while high doses suppress EMT [115]. High doses of vitamin C (4 g/kg of BW) also significantly suppressed the metastasis of breast cancer in a xenograft mouse model [115]. These studies suggest that pharmacological doses of vitamin C over 0.1 mM may increase the production of ROS and DNA damage in cancer cells, while physiological doses promote the tumor growth and metabolism [158]. Although potential antitumorigenic mechanisms of high-dose vitamin C (e.g., hydrogen peroxide generation, enzymatic cofactor reactions, antioxidant, and anti-inflammatory activities) have been proposed, more clinical investigations are needed to support the potential of vitamin C as an effective cancer-therapy option [159,160,161,162].

2.6. Vitamin D

Vitamin D is a precursor to the fat-soluble steroid hormone calcitriol (1, 25(OH)2D3) [163]. Vitamin D3 (cholecalciferol)and vitamin D2 (i.e., ergocalciferol) can be synthesized by exposing skin to ultraviolet light or consumed from fortified food and supplements [104]. Vitamin D is hydroxylated into 25-hydroxyvitamin D (25(OH)D) in liver and is activated to produce calcitriol in the kidneys [164]. Calcitriol notably regulates calcium-phosphate homeostasis for bone health [165]. Upon binding to the vitamin D receptor (VDR), calcitriol regulates gene transcription related to cellular growth, differentiation, apoptosis, and the function of the immune, nerve, and cardiovascular systems [165,166].
Decades of research have demonstrated that low serum 25(OH)D is related to the initiation of many cancers, such as colon, breast, prostate, and gastric [164,167]. Accumulating evidence indicates that calcitriol has antitumorigenic effects through its antiproliferative, proapoptotic, antiangiogenic, and antimetastatic activities in various cancers [104,164,168]. Calcitriol and vitamin D3 analogs suppress the secretion of proteolytic enzymes in breast and prostate cancer cells while also inhibiting the expression of receptors of cell-adhesion molecules, e.g., vascular cell-adhesion protein 1 (VCAM-1) [117]. This is a noteworthy observation, as proteolytic enzyme activation and cell-adhesion-molecule expression in cancer cells can increase invasiveness and metastasis [117]. Some studies have shown that 1,25(OH)2D3 and vitamin D analogs have potential anticancer effects [40]. Chiang et al. compared the effects of 1,25(OH)2D3 and 19-nor-2-(3-hydroxypropyl)-1α,25-dihydroxyvitamin D3 (MART-10) on hypopharyngeal and tongue cancer cells [120]. MART-10 showed the greater inhibiting effect on both cancer-cell growth via G0/G1 cell-cycle arrest and the upregulation of p21 and p27 expression. Milczarek et al. reported that vitamin D analogs sensitize colon cancer to 5-fluorouracil in vivo [118]. Gorham et al. suggested a daily intake of 1000 to 2000 IU/day of vitamin D3 to decrease the risk of colorectal cancer [119].

2.7. Vitamin E

Vitamin E is a group of hydrophobic compounds in eight isoforms consisting of α, β, γ, and δ subtypes of tocopherols and tocotrienols [169]. Tocopherols are saturated forms of vitamin E, while tocotrienols are unsaturated with short tails and three double bonds [121]. They are rich in oils such as vegetable, palm, and rice bran, along with cereal grains [170]. After being absorbed in the small intestine, vitamin E, specifically α-tocopherol, binds to an α-tocopherol transfer protein in the liver [171].
Due to their lower bioavailability versus α-tocopherol, tocotrienols have been neglected for years [172]. However, it has been reported that some tocopheroland tocotrienol isoforms (i.e., γ and δ) have greater anticancer activities than the most abundant isoform, α-tocopherols [173]. In addition, there is increasing evidence that supports the anticancer effects of tocotrienols in breast, colorectal, lung, prostate, pancreas, and liver cancers [41]. γ-tocotrienol, for instance, increases cell-cycle arrest in G1/S phase in MCF-7 and MDA-MB-231 breast cancer cells [121,122] and induces apoptosis in HT-29 colon cancer cells by increasing apoptotic caspase-3 and downregulating NF-κB signaling [123]. Moreover, tocotrienols significantly sensitize cancer cells to chemotherapeutic drugs [174].

2.8. Vitamin K

Vitamin K is a group of lipid-soluble vitamins that has two natural forms: vitamin K1 (phylloquinone) and vitamin K2 (menaquinone) [175]. Vitamin K1 can be found in green leafy vegetables and olive and soybean oils, while vitamin K2 is mainly present in fermented food such as cheese, natto, and curds, endogenously generated by intestinal bacteria [176].
In the prospective cohort study of the European Prospective Investigation into Cancer and Nutrition-Heidelberg, dietary intake of vitamin K2 had an inverse association with cancer mortality [177]. An alternate prospective cohort analysis in a Mediterranean population also indicated that dietary intake of vitamin K is inversely associated with the risk of cancer mortality [175]. Vitamin K2 also increases nonapoptotic cell death with autophagy in TNBC cells [42]. However, the relationship between vitamin K and cancer is still unclear, and further studies are required to properly understand the mechanistic contributions of vitamin K to cancer.

2.9. Asparagine

Asparagine is a nonessential amino acid that is either synthesized from glutamate and aspartate by asparagine synthetase (Figure 1) or acquired from exogenous sources, e.g., cereal grains [178]. In recent years, asparagine, a precursor to the Class 2A carcinogen acrylamide, has been found to be important in maintaining the health of glutamine-independent liposarcoma and breast cancer cells [19,96,178]. As an exchange factor, asparagine works to import exogenous amino acids, e.g., arginine and serine, and 2 mM asparagine activates the mammalian target of rapamycin-1 (mTOR1) protein complex, a major complex that regulates cell growth and proliferation, in the absence of glutamine in HeLa cervical cancer cells [96,179]. Krall et al. noted that these functions promote asparagine-induced protein and nucleotide synthesis, contributing to the longevity of cancer cells [96]. Furthermore, Knott et al. demonstrated that while dietary asparagine restriction does not affect tumor growth, high-asparagine diets (4%) promote EMT and tumor metastasis in vivo versus low-asparagine diets (0.6%) [180].
Previously, studies focused more on the importance of L-asparaginase, a bacterial enzyme utilized as the first treatment of acute lymphoblastic leukemia and lymphoma [181,182,183,184]. One in vitro study found that silencing asparagine synthase increased ovarian cancer cells’ sensitivity to L-asparaginase up to 500-fold, concluding that asparagine synthetase may be used as a predictor of L-asparaginase therapy efficacy [181]. Unfortunately, the effectiveness of the treatment does not translate to in vivo models [185]. A more recent preclinical study showed that the administration of 20,000 U/kg asparaginase alone for 14 days insufficiently produced leukemia cytotoxicity and required additional glutaminase activity for both asparagine synthetase-positive and -negative mice [186]. Moreover, a metabolic analysis of 19 leukemia cells and 26 leukemia patients proved that cancer cells of lower mitochondrial respiration and glycolytic function are more sensitive to asparaginase therapy than higher glycolytic cells [187]. Continued investigation into targeting asparagine may contribute to more effective and clinically translatable cancer therapies.

2.10. Arginine

Arginine (2-amino-5-guanidinovaleric acid) is the semi-essential amino-acid precursor of nitric oxides, polyamines, and glutamate, which is upregulated in times of stress and directly activates mTOR [97,188]. In the urea cycle, citrulline and aspartate synthesize arginosuccinate by arginosuccinate synthase, arginosuccinate synthase synthesizes arginine by arginosuccinate lysate, and arginine synthesizes nitric oxide and citrulline by nitric oxide synthase (Figure 1) [97,189]. Recent studies have suggested that utilizing the presence of diminished plasma arginine levels acts as a biomarker of clinical prostate (<67.18 µmol/L) and breast cancers [126,190]. While not proposing a cutoff for diagnosis, Hu et al. reported that luminal A, luminal B, HER2+, and TNBC patients showed significantly lower arginine levels (7.34 ± 5.64, 9.98 ± 6.84, 8.27 ± 6.78, and 4.18 ± 3.34 µmol/L, respectively) compared to healthy patients [97,189,190]. Additionally, a diet rich in soy protein, fish, walnuts, and peanuts can supplement intracellular arginine synthesis [191], on which melanoma and ovarian cancer cells have been found to be dependent on in cases of arginosuccinate synthase deficiency [25,125]. Therefore, arginine starvation, which functions by inducing asparagine synthetase and depleting aspartate, has been used to treat arginosuccinate synthase-1-deficient breast cancer in vitro [124] and in vivo [101]. Ji et al. utilized PEGylated arginine deiminase 20,000 molecular weight (ADI-PEG20) to degrade arginine in ovarian cancer cells and xenograft models [125], while Izzo et al. found the therapy unsuccessful in a limited clinical study of liver cancer [192]. One in vitro study of ADI-PEG20-resistant melanoma reported that the oncogene c-MYC binds to the arginosuccinate synthase-1 promoter to upregulate arginosuccinate lysate and PI3K/AKT sensitivity [26]. mTOR signaling, glutamine, glutamine dehydrogenase, and sensitivity to glutamine inhibitors are also enhanced [26].

2.11. Serine and Glycine

Serine is a nonessential amino acid synthesized when 3-phospho-glycerate is oxidized to 3-phospho-hydroxypyruvate by phosphoglycerate dehydrogenase (PHGDH), transaminated to phosphoserine by phosphoserine aminotransferase (PSAT), and finally dephosphorylated via phosphoserine phosphatase (PSPH) [128,193,194]. Glycine, which is transformed from serine de novo in mitochondria via serine hydroxymethyltransferase (SHMT) and vitamin B6 (Figure 1) [128,136], is a nonessential amino acid and has been shown to work alongside serine in protein synthesis in 60 tumor-derived cell lines (NCI60) [195]. Dolfi et al. also highlighted that glycine exchange rates significantly correlate with cell proliferation and DNA synthesis in these 60 cancer-cell lines [195]. Both serine and glycine are highly regarded for their roles in protein, phospholipid, and glutathione synthesis via the serine synthesis pathway (SSP) [99,196,197]. As key players in one-carbon metabolism (i.e., folate and methionine metabolism), both serine and glycine collaborate in nucleotide synthesis, e.g., purine, in healthy patients [198,199]. However, while serine is accepted to contribute to cancer-cell purine synthesis, glycine has been demonstrated to work in a cancer-type-dependent manner [128]. For example, Labuschagne et al. demonstrated that in vitro breast and colon cancer cells prefer serine over glycine, and when starved of serine, cells show reduced nucleotide synthesis; furthermore, cells fed 0.4 to 2 mM serine exhibit increased proliferation [99]. The study found that cancer cells react to glycine in a dose-dependent manner: while low concentrations (0.4 mM) moderately increase cell proliferation, higher concentrations (1 to 2 mM) inhibit cancer cell proliferation [99]. Another study found that brain metastatic breast cancer cells upregulate de novo serine when deprived of the exogenous alternative, and PHGDH suppression reduces brain metastases of NCSLC and TNBC in vivo [134]. A recent in vitro genetic analysis found that when fed serine-free media, serine-starvation-resistant colon cancer cells increased serine, glycine, and threonine metabolic pathways [135]. In the short term, this serine inhibition decreases Yes-associated protein activation, which controls tumorigenesis, but does the opposite over a prolonged period, as SSP promotes the catabolism of PHGDH, PSAT1, and PSPH [135]. Furthermore, Meiser et al. found that serine–glycine catabolism is induced by stress and contributes to formate levels in in vivo colorectal cancer [136]. Interestingly, a study of NSCLC found that most de novo serine and glycine are allocated for glutathione synthesis [128], an important antioxidant in ROS homeostasis. Maddocks et al. proposed that serine and glycine inhibition must be used in parallel in vivo to induce a significant decrease in tumor size in colon cancer [102]. The study concluded that serine starvation in vitro induces cell metabolic stress and causes cells to rely on p53-mediated glycolysis [102].
As previously noted, SSP and serine–glycine metabolism enzymes are also implicated in cancer [193,197]. One breast cancer study found that PHGDH and PSPH are highly expressed in in vitro TNBC, and these, along with SHMT-1, are also elevated in stromal TNBC tumors [193]. Tumor PSPH positivity, stromal PSPH positivity, and stromal SHMT-1 negativity are linked to decreased survival in TNBC and HER-2 breast cancers [193]. Additionally, glycine decarboxylase, which transforms glycine into the one-carbon metabolism intermediate methylenetetrahydrofolate, was elevated in MDA-MD-453 and MDA-MB-435 breast cancer cell lines [193].

2.12. Cysteine

Cysteine is the rate-limiting substrate in glutathione production in cells [200]. Cancer cells can become dependent on cysteine in order to uphold the functions of glutathione, e.g., ROS depletion, protein modification, and cell signaling [14,98]. Cysteine is regulated via the cystine–cysteine cycle, in which extracellular cystine is captured, imported via the cystine/glutamate antiporter system, and transformed into cysteine by thioredoxin reductase-1 (Figure 1) [14,201]. Because cancer cells produce an abundance of ROS as a product of aerobic glycolysis, increased levels of glutathione, and thus increased cysteine levels, are vital in maintaining ROS homeostasis to promote tumorigenesis [10,14]. While a recent study of in vitro and in vivo ovarian cancer found cysteine depletion to be a successful therapy [202], many patients do not respond to this method of induced oxidative stress [14,98]. Consequently, studies have investigated targeting the cystine/glutamate antiporter system [29,98]. Tarragó-Celada et al. evaluated liver-metastatic colon cancer in vitro and discovered that metastatic cell proliferation significantly decreases following cystine starvation, and cells are particularly sensitive to cystine/glutamate antiporter-targeted therapies [29]. Alothaim et al. demonstrated that inhibitors of histone deacetylase-6, a moderator of tumor-cell proliferation and metastasis, sensitize cystine/glutamate antiporter-targeted therapy-resistant TNBC cells to cysteine deprivations by signaling necroptosis and ferroptosis cell death [98]. Alternatively, one in vitro study utilized autophagy inhibition to diminish cysteine homeostasis via the deletion of the SLC7A11 cystine transporter gene in pancreatic cancer cells [127]. However, more clinically translatable studies are needed to develop the complete mechanistic understanding of cysteine in cancer and cysteine-targeted therapies.

3. Metabolites and Cancer–Nerve Crosstalk

3.1. Background

In the last two decades, an emergence of evidence has supported the role of the nervous system as a key player in cancer progression, increasing patient pain and poor outcomes [70,203]. PNI occurs when cancer cells invade adjacent nerves to aid in metastasis to secondary sites, with a high prevalence in cancer such as colorectal, head and neck, liver, pancreatic, and prostate [75,204]. TI, however, is the event in which neurites from adjacent nerves infiltrate nearby solid tumors, e.g., breast, cervical, head and neck, lung, and pancreatic cancers [62,66,69,205]. This crosstalk complicates cancer treatment, as there is limited success in preoperative diagnoses of nerve involvement in cancer, making surgical interventions more difficult to successfully complete [77], and few studies have explored radiation and chemotherapy as a method of blocking cancer and/or neurite invasion [78,79]. Though more studies have investigated the mechanism of PNI, it is evident that both forms of cancer–nerve crosstalk are progressed via chemokines, neurotrophins, and neurotransmitters [45]. However, the role of tumor metabolic dysregulation in cancer–nerve crosstalk remains underappreciated. Only within the last 5 years has literature immerged presenting metabolic players, e.g., vitamin C uptake gene SLC2A3 [206], asparagine synthetase [28], neuron-secreted serine [207], and lactate importer MCT1 [205], as contributors to PNI and/or TI. Alternatively, there is abundant literature examining the neuroregulatory properties of these metabolites. Combining this knowledge with the present understanding of tumor metabolism may aid in developing a more comprehensive understanding of cancer–nerve crosstalk mechanisms and in educating future potential therapies.

3.2. Known Contributors of Cancer–Nerve Crosstalk

3.2.1. Vitamin C and SLC2A3

Vitamin C has an important role in synthesizing neurotransmitters. Recently, some studies showed the effect of vitamin C on peripheral nerve regeneration after traumatic injury [208,209]. In addition, Gao et al. reported that the solute carrier family 2 member 3 (SLC2A3) expression was remarkably associated with PNI in colorectal cancer (Figure 2A) [205]. SLC2A3 gene upregulation, which encodes glucose transporter 3, showed decreased disease-free survival in colorectal cancer patients [210]. Liu et al. showed that low SLC2A3 expression in acute myeloid leukemia significantly suppressed the effect of vitamin C, resulting in diminished overall survival [211]. Further investigation on tumor SLC2A3 expression should be conducted to develop a more comprehensive understanding of SLC2A3 and vitamin C in cancer–nerve crosstalk.

3.2.2. Asparagine and Asparagine Synthetase

While the present literature does not highlight the importance of asparagine in the nervous system or neural disorders, the mutation of asparagine synthetase-promoting genes is linked to severe impairments. Asparagine synthetase deficiency is a congenital disorder that causes cognitive impairment, microcephaly, seizures, and progressive cerebral atrophy [212]. In a clinical and in vitro study of oral squamous-cell carcinoma, Fu et al. presented a correlation between high asparagine synthetase levels and histological and mRNA evidence of PNI (Figure 2B,C) [28]. Moreover, the study found that L-asparagine was the only amino acid with high sensitivity and specificity in diagnosing PNI, and patients with high asparagine synthetase exhibited PNI-positive tumors [28]. More studies are necessary to examine the roles of asparagine and asparagine synthetase in PNI and TI for the potential of developing future therapeutics targeting these pathways.

3.2.3. Serine and Glycine

Serine not only promotes cancer-cell proliferation, but also aids in maintaining the health of neurites. Nusser et al. observed that NGF treatment activates protein kinase A, leading to the phosphorylation of RhoA, a G protein that regulates cell-cycle progression, gene expression, and cell motility [213], on serine [214]. This cascade inhibits the RhoA-Rho-associated kinase binding necessary to restrict neurite outgrowth [214]. Moreover, a more recent in vitro analysis found that NGF-induced serine phosphorylation promotes signal transducer and activator of transcription 3 (STAT3), which is then responsible for induced neurite outgrowth [215]. While limited studies exist, Tapia et al. discovered glycine (50 µM) activates chloride-ion membrane currents of neurons in vitro, and glycine receptor activation correlates to depolarizing excitatory potentials [216]. Interestingly, glycine was found to affect neurite outgrowth in a dose-dependent manner, but neurons become desensitized after long exposure, e.g., higher concentrations are needed to induce outgrowth [216].
Banh et al. published the first documented study implicating the role of serine in tumor innervation [207]. In the study, in vitro pancreatic ductal adenocarcinoma cells upregulated serine synthesis following serine/glycine starvation, which promoted cell growth in a dose-dependent manner [207]. Additionally, in this nutrient-poor environment, axons release amino acids, e.g., serine, to support the health of exogenous serine-dependent cancer cells, and in vivo, pancreatic tumors in mice starved of serine/glycine showed increased innervation by sympathetic and sensory nerves [207]. The group found that blocking innervation via NGF receptor TRK inhibitor, LOXO-101, slowed tumor growth in serine/glycine-deprived mice, suggesting the promise of utilizing serine/glycine starvation with innervation inhibitors as cancer therapies [207]. However, more studies should be conducted to prove its therapeutic efficacy in other tumor types and the clinical setting.

3.3. Proposed Targets for Cancer–Nerve Crosstalk Research

3.3.1. Lactate

In the exercise sciences, it is understood that muscles release lactate (and its protonated form lactic acid) into the bloodstream following physical activity [217]. Studies have shown that while some lactate is processed by the liver and utilized for oxidative respiration, lactate can also cross the blood–brain barrier and mediate cognitive function [218]. Once imported into neurons by MCT1 in vivo, lactate signals silent information regulator-1 (SIRT1) to induce upregulation of transcriptional factor PGCa and secreted factor FNDC5 to ultimately facilitate BDNF expression [218]. A recent meta-analysis of interval training, characterized by increased lactate output, concluded that physical activity induces BDNF release in humans [219]. While no distinct link between lactate and cancer–nerve crosstalk has been confirmed, BDNF is a known contributor of both PNI [220] and TI [88]; therefore, continued examinations of the role of lactate in this relationship are warranted.
Alternatively, Sandforth et al. found a distinct correlation between MCT1, which imports lactate, and PNI in pancreatic cancer (Figure 2D) [209]. In humans, MCT1 has been found in oligodendrocytes, astrocytes, microglia, endothelial cells, and neurons, and the wellbeing of glia–neuron metabolic crosstalk relies on MCT functionality [221]. Additionally, Lin et al. showed that LDH-A release from damaged neurons facilitates angiogenesis in the central nervous system via interaction with adjacent vimentin-expressing endothelial cells [222]. Further studies should be conducted to fully comprehend the value of lactate, its transporters, and LDH in cancer–nerve crosstalk and investigate the efficacy of lactate silencing/inhibition as a treatment option.

3.3.2. Vitamin A

Retinoic acid and NGF have been reported to show synergistic effects on neuroprotection related to neuronal survival and growth [223]. Combination treatment of NGF and all-trans retinoic acid (ATRA) on 8705-C thyroid papillary tumor cells inhibited their proliferation and invasion [224]. Arrieta et al. investigated the effect of ATRA on chemotherapy-induced peripheral neuropathy in the male Wistar rat model [225] by administering 20 mg/kg per os (PO) of ATRA for 15 days. ATRA was found to suppress chemotherapy-induced neuropathy by increasing NGF and retinoic-acid-receptor beta (RAR-β) expression. Additionally, the group conducted a randomized, double-blinded, controlled study in which 95 NSCLC patients were administered 20 mg/m2 of ATRA per day for 1 week before undergoing chemotherapy over two courses (21 days per course) [225]. ATRA-treated patients presented with a reduction in axonal degeneration [225]. Higashi et al. investigated the effect of retinoic acid on neuroblastoma cell lines and found that treatment with 1 to 10 μM of 13-cis-retinoic acid for 3 to 12 days upregulated cell expression of chromodomain-helicase DNA-binding protein 5 (CHD5), a tumor-suppressing gene, and induced neuronal differentiation in SH-SY5Y, NGP, and SK-N-DZ cells [226]. NGF also increased CHD5 expression and neuronal differentiation in SY5Y cells. The study concluded that 13-cis-retinoic acid administration for a year after surgery has a preventive effect on recurrence in neuroblastoma patients [227]. As NGF is one of the key players in cancer–nerve crosstalk, the link between retinoic acid and NFG in cancer of the nervous system calls for future studies to determine the full extent of this relationship [81,82,83].

3.3.3. Vitamin B

Neurodegenerative disease is caused by alterations in the central nervous system and has been credited to protein misfolding induced by disordered metabolite control of proteins [228]. Protein misfolding is the cause of many types of neurodegenerative diseases, including Alzheimer’s disease (AD) [229]. Studies have presented NGF as a beneficial treatment of AD due its role in promoting neurite outgrowth [230]. Therefore, Ina and Kamei probed the mechanism of vitamin B12-mediated neurite outgrowth and found that low concentrations of NGF (10 ng/mL), vitamin B12 (6 to 100 µM) promoted neurite outgrowth and the differentiation of PC12 pheochromocytoma cells (a type of neuroendocrine tumor) into neuron-like cells [231]. Upon further investigation, the study found that by using protein kinase inhibitors, vitamin B12 stimulates PC12 differentiation in a manner that involves the same signal-transduction pathways activated by NGF. The results suggest that vitamin B12 can stimulate neural differentiation, and that like NGF, it stimulates the mitogen-activated protein kinase/extracellular receptor kinase (MAPK/ERK)-signaling pathway [231]. In addition, Okada et al. showed that MeCbl (≥100 nM) promotes neurite outgrowth and neuronal survival [232]. These outcomes were mediated by the methylation cycle and demonstrated that MeCbl increases ERK1/2 and AKT activities through this process [232]. Neurotrophins, such as NGF and BDNF, also activate ERK1/2 and AKT [233]. This activation of ERK1/2 promotes neurite outgrowth and AKT initiates branching of dorsal root ganglia neurites [234]. In a follow-up study, Okada et al. revisited this mechanism and determined that MeCbl increases mTOR activity, a protein kinase that regulates neurite outgrowth and nerve regeneration, through the activation of AKT, which in turn promotes neurite outgrowth in cerebellar granule neurons [235,236].
In addition to vitamin B12, other isoforms of vitamin B are linked to promoting peripheral nerve regeneration, including B1 and B6 [208]. These neurotrophic B vitamins support the development of new cell structures and are key players in nerve regeneration while also maintaining neuronal viability. Vitamin B1 acts as a site-directed antioxidant and facilitates the use of carbohydrates for energy production, vitamin B6 balances nerve metabolism, and vitamin B12 promotes neural cell survival and myelin sheath maintenance [236]. It has been determined that a combination of vitamin B complex is necessary to optimize regeneration in cases of peripheral neuropathy [237]. Deficiencies in these vitamins have been associated with nerve dysfunction and damage and can lead to peripheral neuropathy [238]. Altun and Kurutaş suggested that tissue vitamin B complex levels vary during crush-induced peripheral nerve injury, and supplementation during these acute time periods may accelerate nerve regeneration [239]. These neuroregulatory properties of vitamin B, especially vitamin B12 (in an NGF-like manner) [231] show the potential value of targeting vitamin B in PNI and TI therapies. However, further research may provide a clearer mechanism by which vitamin B contributes to this crosstalk.

3.3.4. Vitamin D

The active form of vitamin D, 1,25(OH)2D3, is a hormone that has a similar influence as that of neurosteroids [240]. Vitamin D has an important role in neuronal differentiation and maturation via neurotrophin regulation [241]. NGF, glial-cell-derived neurotrophic factor (GDNF), and neurotrophin 3 levels are increased by vitamin D, to facilitate neuronal growth [242]. Male Sprague Dawley rats injected with vitamin D3 for 8 days showed significant increased GDNF in the cortex with significantly decreased infarction amount after middle cerebral-artery ligation [120]. Rats from mothers with vitamin D3 deficiency showed significant changes in brain structure and low NGF and GDNF levels at birth [243]. Even though there has been no apparent link between vitamin D and cancer–nerve crosstalk, the value of vitamin D in controlling neurotrophin expression [241] highlights its potential in mediating PNI and/or TI and requires further investigation.

3.3.5. Vitamin E and K

Vitamin E has an important role in maintaining normal neurological function and structure [244]. In in vivo sciatic-nerve-crush injury models, vitamin E acetate showed a neuroprotective effect with steady improvement in motor-nerve-conduction velocity and thermal hyperalgesia [245]. In addition, Pace et al. showed that vitamin E significantly reduced the incidence of chemotherapy-induced peripheral neuropathy and its severity in patients following cisplatin chemotherapy, without affecting the antitumorigenic activity of cisplatin [246]. These neuroprotective properties of vitamin E suggest its potential involvement in cancer–nerve crosstalk, and therefore warrant additional studies. Vitamin K1 and K2 treatment of PC12D cells (100 µg/mL) in the presence of 2.5 to 50 ng/mL NGF significantly increased neurite outgrowth [247]. One study found that vitamin K-induced neurite outgrowth is potentially mediated via protein kinase A and MAPK cascades [248]. In the nervous system, vitamin K-dependent growth-arrest-specific protein 6 activates receptors of TAM, which includes tyrosine-protein kinase receptor 3, receptor tyrosine kinase, and MER proto-oncogene tyrosine kinase [249]. Similar defects in TAM are associated with cancer, indicating the potential for vitamin K to facilitate cancer initiation [249]. More studies are needed to fully understand the mechanisms and contributions of vitamin K in the nervous system, cancer progression, and cancer–nerve crosstalk.

4. Conclusions

In this review, we have summarized the current knowledge of significant yet relatively underappreciated metabolites in cancer development and metastasis (i.e., lactate, vitamins A, B, C, D, E and K, asparagine, arginine, serine, glycine, and cysteine). In addition, we discussed metabolites and their regulators currently established as contributors to cancer–nerve crosstalk (i.e., SLC2A3, asparagine synthetase, and serine) and suggested metabolites that may be implicated in cancer–nerve crosstalk based on their tumorigenic and neuroregulatory properties (i.e., lactate and vitamins A, B, D, E, and K). However, there remains limited information to make a clear connection between these nutrients and cancer–nerve crosstalk. It is our hope that this review serves researchers as a guide to developing future studies to determine the roles of metabolites in cancer and nerve crosstalk in order to expand the collective understanding of cancer mechanisms that may be beneficial in developing potential therapies for cancer, PNI, and TI.

Author Contributions

Conceptualization, Y.H.S. and S.-O.L.; writing—original draft preparation, I.G., E.G., and C.A.; writing—review and editing, Y.H.S., S.-O.L., I.G. and E.G.; supervision, Y.H.S. and S.-O.L.; funding acquisition, Y.H.S. and S.-O.L. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health through the award number P20GM139768 awarded to Y.H.S., as well as Chancellor’s Fund for Innovation & Collaboration at the University of Arkansas awarded to Y.H.S. and S.-O.L.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. American Cancer Society. Cancer Facts & Figures; American Cancer Society: Atlanta, GA, USA, 2022. [Google Scholar]
  2. International Agency for Research on Cancer; World Health Organization: Geneva, Switzerland, 2020.
  3. Wu, S.; Zhu, W.; Thompson, P.; Hannun, Y.A. Evaluating intrinsic and non-intrinsic cancer risk factors. Nat. Commun. 2018, 9, 3490. [Google Scholar] [CrossRef] [PubMed]
  4. Emmons, K.M.; Colditz, G.A. Realizing the Potential of Cancer Prevention—The Role of Implementation Science. N. Engl. J. Med. 2017, 376, 986–990. [Google Scholar] [CrossRef] [Green Version]
  5. Blot, W.J.; Tarone, R.E. Doll and Peto’s quantitative estimates of cancer risks: Holding generally true for 35 years. J. Natl. Cancer Inst. 2015, 107, djv044. [Google Scholar] [CrossRef] [PubMed]
  6. Key, T.J.; Bradbury, K.E.; Perez-Cornago, A.; Sinha, R.; Tsilidis, K.K.; Tsugane, S. Diet, nutrition, and cancer risk: What do we know and what is the way forward? BMJ 2020, 368, m511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Haskins, C.P.; Champ, C.E.; Miller, R.; Vyfhuis, M.A.L. Nutrition in Cancer: Evidence and Equality. Adv. Radiat. Oncol. 2020, 5, 817–823. [Google Scholar] [CrossRef] [PubMed]
  8. Potter, M.; Newport, E.; Morten, K.J. The Warburg effect: 80 years on. Biochem. Soc. Trans. 2016, 44, 1499–1505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. VanderHeiden, M.G.; Cantley, L.C.; Thompson, C.B. Understanding the Warburg Effect: The Metabolic Requirements of Cell Proliferation. Science 2009, 324, 1029–1033. [Google Scholar] [CrossRef] [Green Version]
  10. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211–218. [Google Scholar] [CrossRef] [Green Version]
  11. Jang, M.; Kim, S.S.; Lee, J. Cancer cell metabolism: Implications for therapeutic targets. Exp. Mol. Med. 2013, 45, e45. [Google Scholar] [CrossRef] [Green Version]
  12. Rodic, S.; Vincent, M.D. Reactive oxygen species (ROS) are a key determinant of cancer’s metabolic phenotype. Int. J. Cancer 2018, 142, 440–448. [Google Scholar] [CrossRef]
  13. Panieri, E.; Santoro, M.M. Ros homeostasis and metabolism: A dangerous liason in cancer cells. Cell Death Dis. 2016, 7, e2253. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Liu, J.; Liu, M.; Zhang, H.; Wei, X.; Wang, J.; Xian, M.; Guo, W. Exploring cysteine regulation in cancer cell survival with a highly specific “lock and Key” fluorescent probe for cysteine. Chem. Sci. 2019, 10, 10065–10071. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Cluntun, A.A.; Lukey, M.J.; Cerione, R.A.; Locasale, J.W. Glutamine Metabolism in Cancer: Understanding the Heterogeneity. Trends Cancer 2017, 3, 169–180. [Google Scholar] [CrossRef] [Green Version]
  16. Jiang, J.; Srivastava, S.; Zhang, J. Starve cancer cells of glutamine: Break the spell or make a hungry monster? Cancers 2019, 11, 804. [Google Scholar] [CrossRef] [Green Version]
  17. Wise, D.R.; Thompson, C.B. Glutamine addiction: A new therapeutic target in cancer. Trends Biochem. Sci. 2010, 35, 427–433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Yu, L.; Teoh, S.T.; Ensink, E.; Ogrodzinski, M.P.; Yang, C.; Vazquez, A.I.; Lunt, S.Y. Cysteine catabolism and the serine biosynthesis pathway support pyruvate production during pyruvate kinase knockdown in pancreatic cancer cells. Cancer Metab. 2019, 7, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Zhang, J.; Fan, J.; Venneti, S.; Cross, J.R.; Takagi, T.; Bhinder, B.; Djaballah, H.; Kanai, M.; Cheng, E.H.; Judkins, A.R.; et al. Asparagine Plays a Critical Role in Regulating Cellular Adaptation to Glutamine Depletion. Mol. Cell 2014, 23, 205–218. [Google Scholar] [CrossRef] [Green Version]
  20. Zhu, Y.; Li, T.; Da Silva, S.R.; Lee, J.J.; Lu, C.; Eoh, H.; Jung, J.U.; Gao, S.J. A critical role of glutamine and asparagine γ-Nitrogen in nucleotide biosynthesis in cancer cells hijacked by an oncogenic virus. MBio 2017, 8, e01179-17. [Google Scholar] [CrossRef] [Green Version]
  21. Choi, Y.; Park, K. Targeting Glutamine Metabolism for Cancer Treatment. Biomol. Ther. 2018, 26, 19–28. [Google Scholar] [CrossRef] [Green Version]
  22. Butler, M.; van der Meer, L.T.; van Leeuwen, F.N. Amino Acid Depletion Therapies: Starving Cancer Cells to Death. Trends Endocrinol. Metab. 2021, 32, 367–381. [Google Scholar] [CrossRef]
  23. Tsai, P.Y.; Lee, M.S.; Jadhav, U.; Naqvi, I.; Madha, S.; Adler, A.; Mistry, M.; Naumenko, S.; Lewis, C.A.; Hitchcock, D.S.; et al. Adaptation of pancreatic cancer cells to nutrient deprivation is reversible and requires glutamine synthetase stabilization by mTORC1. Proc. Natl. Acad. Sci. USA 2021, 118, e2003014118. [Google Scholar] [CrossRef] [PubMed]
  24. Hamanaka, R.B.; Chandel, N.S. Targeting glucose metabolism for cancer therapy. J. Exp. Med. 2012, 209, 211–215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Sonveaux, P.; Végran, F.; Schroeder, T.; Wergin, M.C.; Verrax, J.; Rabbani, Z.N.; De Saedeleer, C.J.; Kennedy, K.M.; Diepart, C.; Jordan, B.F.; et al. Targeting lactate-fueled respiration selectively kills hypoxic tumor cells in mice. J. Clin. Investig. 2008, 118, 3930–3942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Long, Y.; Tsai, W.B.; Wangpaichitr, M.; Tsukamoto, T.; Savaraj, N.; Feun, L.G.; Kuo, M.T. Arginine deiminase resistance in melanoma cells is associated with metabolic reprogramming, glucose dependence, and glutamine addiction. Mol. Cancer Ther. 2013, 12, 2581–2590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Truman, J.P.; Ruiz, C.F.; Montal, E.; Garcia-Barros, M.; Mileva, I.; Snider, A.J.; Hannun, Y.A.; Obeid, L.M.; Mao, C. 1-Deoxysphinganine initiates adaptive responses to serine and glycine starvation in cancer cells via proteolysis of sphingosine kinase. J. Lipid Res. 2022, 63, 100154. [Google Scholar] [CrossRef]
  28. Fu, Y.; Ding, L.; Yang, X.; Ding, Z.; Huang, X.; Zhang, L.; Chen, S.; Hu, Q.; Ni, Y. Asparagine Synthetase-Mediated l-Asparagine Metabolism Disorder Promotes the Perineural Invasion of Oral Squamous Cell Carcinoma. Front. Oncol. 2021, 11, 637226. [Google Scholar] [CrossRef]
  29. Tarragó-Celada, J.; Foguet, C.; Tarrado-Castellarnau, M.; Marin, S.; Hernández-Alias, X.; Perarnau, J.; Morrish, F.; Hockenbery, D.; Gomis, R.R.; Ruppin, E.; et al. Cysteine and folate metabolism are targetable vulnerabilities of metastatic colorectal cancer. Cancers 2021, 13, 425. [Google Scholar] [CrossRef]
  30. Muramatsu, H.; Sumitomo, M.; Morinaga, S.; Kajikawa, K.; Kobayashi, I.; Nishikawa, G.; Kato, Y.; Watanabe, M.; Zennami, K.; Kanao, K.; et al. Targeting lactate dehydrogenase-A promotes docetaxel-induced cytotoxicity predominantly in castration-resistant prostate cancer cells. Oncol. Rep. 2019, 42, 224–230. [Google Scholar] [CrossRef]
  31. Jeschke, J.; O’Hagan, H.M.; Zhang, W.; Vatapalli, R.; Calmon, M.F.; Danilova, L.; Nelkenbrecher, C.; Van Neste, L.; Bijsmans, I.T.G.W.; Van Engeland, M.; et al. Frequent inactivation of Cysteine dioxygenase type 1 contributes to survival of breast cancer cells and resistance to anthracyclines. Clin. Cancer Res. 2013, 19, 3201–3211. [Google Scholar] [CrossRef] [Green Version]
  32. Nicholson, L.J.; Smith, P.R.; Hiller, L.; Szlosarek, P.W.; Kimberley, C.; Sehouli, J.; Koensgen, D.; Mustea, A.; Schmid, P.; Crook, T. Epigenetic silencing of argininosuccinate synthetase confers resistance to platinum-induced cell death but collateral sensitivity to arginine auxotrophy in ovarian cancer. Int. J. Cancer 2009, 125, 1454–1463. [Google Scholar] [CrossRef]
  33. Papadimitriou, N.; Markozannes, G.; Kanellopoulou, A.; Critselis, E.; Alhardan, S.; Karafousia, V.; Kasimis, J.C.; Katsaraki, C.; Papadopoulou, A.; Zografou, M.; et al. An umbrella review of the evidence associating diet and cancer risk at 11 anatomical sites. Nat. Commun. 2021, 12, 4579. [Google Scholar] [CrossRef] [PubMed]
  34. Soldati, L.; Di Renzo, L.; Jirillo, E.; Ascierto, P.A.; Marincola, F.M.; De Lorenzo, A. The influence of diet on anti-cancer immune responsiveness. J. Transl. Med. 2018, 16, 75. [Google Scholar] [CrossRef] [PubMed]
  35. Tsai, W.B.; Aiba, I.; Long, Y.; Lin, H.K.; Feun, L.; Savaraj, N.; Kuo, M.T. Activation of Ras/PI3K/ERK pathway induces c-Myc stabilization to upregulate argininosuccinate synthetase, leading to arginine deiminase resistance in melanoma cells. Cancer Res. 2012, 72, 2622–2633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Kremer, J.C.; Prudner, B.C.; Lange, S.E.S.; Bean, G.R.; Schultze, M.B.; Brashears, C.B.; Radyk, M.D.A.; Redlich, N.; Tzeng, S.C.; Kami, K.; et al. Arginine Deprivation Inhibits the Warburg Effect and Upregulates Glutamine Anaplerosis and Serine Biosynthesis in ASS1-Deficient Cancers. Cell Rep. 2017, 18, 991–1004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Doldo, E.; Costanza, G.; Agostinelli, S.; Tarquini, C.; Ferlosio, A.; Arcuri, G.; Passeri, D.; Scioli, M.G.; Orlandi, A. Vitamin A, cancer treatment and prevention: The new role of cellular retinol binding proteins. Biomed Res. Int. 2015, 2015, 624627. [Google Scholar] [CrossRef] [Green Version]
  38. Frei, B.; Lawson, S. Vitamin C and cancer revisited. Proc. Natl. Acad. Sci. USA 2008, 105, 11037–11038. [Google Scholar] [CrossRef] [Green Version]
  39. Unlu, A.; Kirca, O.; Ozdogan, M.; Nayır, E. High-dose vitamin C and cancer. J. Oncol. Sci. 2016, 1, 10–12. [Google Scholar] [CrossRef]
  40. Norlin, M. Effects of vitamin D in the nervous system: Special focus on interaction with steroid hormone signalling and a possible role in the treatment of brain cancer. J. Neuroendocrinol. 2020, 32, e12799. [Google Scholar] [CrossRef]
  41. Sailo, B.L.; Banik, K.; Padmavathi, G.; Javadi, M.; Bordoloi, D.; Kunnumakkara, A.B. Tocotrienols: The promising analogues of vitamin E for cancer therapeutics. Pharmacol. Res. 2018, 130, 259–272. [Google Scholar] [CrossRef]
  42. Miyazawa, S.; Moriya, S.; Kokuba, H.; Hino, H.; Takano, N.; Miyazawa, K. Vitamin K2 induces non-apoptotic cell death along with autophagosome formation in breast cancer cell lines. Breast Cancer 2020, 27, 225–235. [Google Scholar] [CrossRef]
  43. Mancino, M.; Ametller, E.; Gascón, P.; Almendro, V. The neuronal influence on tumor progression. Biochim. Biophys. Acta-Rev. Cancer 2011, 1816, 105–118. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Silverman, D.A.; Martinez, V.K.; Dougherty, P.M.; Myers, J.N.; Calin, G.A.; Amit, M. Cancer-associated neurogenesis and nerve-cancer crosstalk. Cancer Res. 2021, 6, 1431–1440. [Google Scholar] [CrossRef] [PubMed]
  45. Gregory, E.; Dugan, R.; David, G.; Song, Y.H. The biology and engineered modeling strategies of cancer-nerve crosstalk. BBA-Rev. Cancer 2020, 1874, 188406. [Google Scholar] [CrossRef] [PubMed]
  46. Wang, H.; Zheng, Q.; Lu, Z.; Wang, L.; Ding, L.; Xia, L.; Zhang, H.; Wang, M.; Chen, Y.; Li, G. Role of the nervous system in cancers: A review. Cell Death Discov. 2021, 7, 76. [Google Scholar] [CrossRef]
  47. Wang, W.; Li, L.; Chen, N.; Niu, C.; Li, Z.; Hu, J.; Cui, J. Nerves in the Tumor Microenvironment: Origin and Effects. Front. Cell Dev. Biol. 2020, 8, 601738. [Google Scholar] [CrossRef]
  48. Ueno, H.; Hase, K.; Mochizuki, H. Criteria for extramural perineural invasion as a prognostic factor in rectal cancer. J. Br. Surg. 2001, 88, 994–1000. [Google Scholar] [CrossRef]
  49. Secq, V.; Leca, J.; Bressy, C.; Guillaumond, F.; Skrobuk, P.; Nigri, J.; Lac, S.; Lavaut, M.N.; Bui, T.T.; Thakur, A.K.; et al. Stromal SLIT2 impacts on pancreatic cancer-associated neural remodeling. Cell Death Dis. 2015, 6, e1592. [Google Scholar] [CrossRef]
  50. Su, D.; Guo, X.; Huang, L.; Ye, H.; Li, Z.; Lin, L.; Chen, R.; Zhou, Q. Tumor-neuroglia interaction promotes pancreatic cancer metastasis. Theranostics 2020, 10, 5029–5047. [Google Scholar] [CrossRef]
  51. Ferdoushi, A.; Li, X.; Griffin, N.; Faulkner, S.; Jamaluddin, M.F.B.; Gao, F.; Jiang, C.C.; van Helden, D.F.; Tanwar, P.S.; Jobling, P.; et al. Schwann Cell Stimulation of Pancreatic Cancer Cells: A Proteomic Analysis. Front. Oncol. 2020, 10, 1601. [Google Scholar] [CrossRef]
  52. Deborde, S.; Omelchenko, T.; Lyubchik, A.; Zhou, Y.; He, S.; McNamara, W.F.; Chernichenko, N.; Lee, S.Y.; Barajas, F.; Chen, C.H.; et al. Schwann cells induce cancer cell dispersion and invasion. J. Clin. Investig. 2016, 126, 1538–1554. [Google Scholar] [CrossRef] [Green Version]
  53. Jia, S.; Wang, W.; Hu, Z.; Shan, C.; Wang, L.; Wu, B.; Yang, Z.; Yang, X.; Lei, D. BDNF mediated TrkB activation contributes to the EMT progression and the poor prognosis in human salivary adenoid cystic carcinoma. Oral Oncol. 2015, 51, 64–70. [Google Scholar] [CrossRef] [PubMed]
  54. Mahjour, F.; Dambal, V.; Shrestha, N.; Singh, V.; Noonan, V.; Kantarci, A.; Trackman, P.C. Mechanism for oral tumor cell lysyl oxidase like-2 in cancer development: Synergy with PDGF-AB. Oncogenesis 2019, 8, 34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Ma, C.; Gao, T.; Ju, J.; Zhang, Y.; Ni, Q.; Li, Y.; Zhao, Z.; Chai, J.; Yang, X.; Sun, M. Sympathetic innervation contributes to perineural invasion of salivary adenoid cystic carcinoma via the β2-adrenergic receptor. Onco Targets Ther. 2019, 12, 1475–1495. [Google Scholar] [CrossRef] [Green Version]
  56. Huang, S.M.; Lin, C.; Lin, H.Y.; Chiu, C.M.; Fang, C.W.; Liao, K.F.; Chen, D.R.; Yeh, W.L. Brain-derived neurotrophic factor regulates cell motility in human colon cancer. Endocr. Relat. Cancer 2015, 22, 455–464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Schonkeren, S.L.; Thijssen, M.S.; Vaes, N.; Boesmans, W.; Melotte, V. The emerging role of nerves and glia in colorectal cancer. Cancers 2021, 13, 152. [Google Scholar] [CrossRef]
  58. Duchalais, E.; Guilluy, C.; Nedellec, S.; Touvron, M.; Bessard, A.; Touchefeu, Y.; Bossard, C.; Boudin, H.; Louarn, G.; Neunlist, M.; et al. Colorectal Cancer Cells Adhere to and Migrate Along the Neurons of the Enteric Nervous System. Cell. Mol. Gastroenterol. Hepatol. 2018, 5, 31–49. [Google Scholar] [CrossRef] [Green Version]
  59. Zhao, Q.; Yang, Y.; Liang, X.; Du, G.; Liu, L.; Lu, L.; Dong, J.; Han, H.; Zhang, G. The clinicopathological significance of neurogenesis in breast cancer. BMC Cancer 2014, 14, 724209. [Google Scholar] [CrossRef] [Green Version]
  60. Sloan, E.K.; Priceman, S.J.; Cox, B.F.; Yu, S.; Pimentel, M.A.; Tangkanangnukul, V.; Arevalo, J.M.G.; Morizono, K.; Karanikolas, B.D.W.; Wu, L.; et al. The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res. 2010, 70, 7042–7052. [Google Scholar] [CrossRef] [Green Version]
  61. Narayan, P.; Flynn, J.; Zhang, Z.; Gillespie, E.F.; Mueller, B.; Xu, A.J.; Cuaron, J.; McCormick, B.; Khan, A.J.; Cahlon, O.; et al. Perineural invasion as a risk factor for locoregional recurrence of invasive breast cancer. Sci. Rep. 2021, 11, 12781. [Google Scholar] [CrossRef]
  62. Kamiya, A.; Hayama, Y.; Kato, S.; Shimomura, A.; Shimomura, T.; Irie, K.; Kaneko, R.; Yanagawa, Y.; Kobayashi, K.; Ochiya, T. Genetic manipulation of autonomic nerve fiber innervation and activity and its effect on breast cancer progression. Nat. Neurosci. 2019, 22, 1289–1305. [Google Scholar] [CrossRef]
  63. Magnon, C.; Hall, S.J.; Lin, J.; Xue, X.; Gerber, L.; Freedland, S.J.; Frenette, P.S. Autonomic Nerve Development Contributes to Prostate Cancer Progression. Science 2013, 15, 713–714. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Sigorski, D.; Gulczyński, J.; Sejda, A.; Rogowski, W.; Iżycka-Świeszewska, E. Investigation of Neural Microenvironment in Prostate Cancer in Context of Neural Density, Perineural Invasion, and Neuroendocrine Profile of Tumors. Front. Oncol. 2021, 11, 710899. [Google Scholar] [CrossRef] [PubMed]
  65. You, H.; Shang, W.; Min, X.; Weinreb, J.; Li, Q.; Leapman, M.; Wang, L.; Tian, J. Sight and switch off: Nerve density visualization for interventions targeting nerves in prostate cancer. Sci. Adv. 2020, 6, eaax6040. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Lucido, C.T.; Wynja, E.; Madeo, M.; Williamson, C.S.; Schwartz, L.E.; Imblum, B.A.; Drapkin, R.; Vermeer, P.D. Innervation of cervical carcinoma is mediated by cancer-derived exosomes. Gynecol. Oncol. 2019, 154, 228–235. [Google Scholar] [CrossRef] [Green Version]
  67. Zhu, Y.; Zhang, G.-N.; Shi, Y.; Cui, L.; Leng, X.-F.; Huang, J.-M. Perineural invasion in cervical cancer: Pay attention to the indications of nerve-sparing radical hysterectomy. Ann. Transl. Med. 2019, 7, 203. [Google Scholar] [CrossRef]
  68. Tang, M.; Liu, Q.; Yang, X.; Chen, L.; Yu, J.; Qi, X.; Wang, Y. Perineural invasion as a prognostic risk factor in patients with early cervical cancer. Oncol. Lett. 2019, 17, 1101–1107. [Google Scholar] [CrossRef] [Green Version]
  69. Shao, J.X.; Wang, B.; Yao, Y.N.; Pan, Z.J.; Shen, Q.; Zhou, J.Y. Autonomic nervous infiltration positively correlates with pathological risk grading and poor prognosis in patients with lung adenocarcinoma. Thorac. Cancer 2016, 7, 588–598. [Google Scholar] [CrossRef]
  70. Azam, S.H.; Pecot, C.V. Cancer’s got nerve: Schwann cells drive perineural invasion. J. Clin. Investig. 2016, 126, 1242–1244. [Google Scholar] [CrossRef] [Green Version]
  71. Ju, J.; Li, Y.; Chai, J.; Ma, C.; Ni, Q.; Shen, Z.; Wei, J.; Sun, M. The role of perineural invasion on head and neck adenoid cystic carcinoma prognosis: A systematic review and meta-analysis. Oral Surg. Oral Med. Oral Pathol. Oral Radiol. 2016, 122, 691–701. [Google Scholar] [CrossRef]
  72. Chen, S.-H.; Zhang, B.-Y.; Zhou, B.; Zhu, C.-Z.; Sun, L.-Q.; Feng, Y.-J. Perineural invasion of cancer: A complex crosstalk between cells and molecules in the perineural niche. Am. J. Cancer Res. 2019, 9, 1–21. [Google Scholar]
  73. Vermeer, P.D. Exosomal induction of tumor innervation. Cancer Res. 2019, 79, 3529–3535. [Google Scholar] [CrossRef]
  74. Gysler, S.M.; Drapkin, R. Tumor innervation: Peripheral nerves take control of the tumor microenvironment. J. Clin. Investig. 2021, 131, e147276. [Google Scholar] [CrossRef] [PubMed]
  75. Shirai, K.; Ebata, T.; Oda, K.; Nishio, H.; Nagasaka, T.; Nimura, Y.; Nagino, M. Perineural invasion is a prognostic factor in intrahepatic cholangiocarcinoma. World J. Surg. 2008, 32, 2395–2402. [Google Scholar] [CrossRef] [PubMed]
  76. Sun, G.; Huang, R.; Zhang, X.; Shen, P.; Gong, J.; Zhao, J.; Liu, J.; Tang, Q.; Shu, K.; Yin, X.; et al. The impact of multifocal perineural invasion on biochemical recurrence and timing of adjuvant androgen-deprivation therapy in high-risk prostate cancer following radical prostatectomy. Prostate 2017, 77, 1279–1287. [Google Scholar] [CrossRef] [PubMed]
  77. Chang, S.T.; Jeffrey, R.B.; Patel, B.N.; Dimaio, M.A.; Rosenberg, J.; Willmann, J.K.; Olcott, E.W. Preoperative Multidetector cT Diagnosis of extrapancreatic Perineural or Duodenal invasion is associated with reduced Postoperative survival after Pancreaticoduodenectomy for Pancreatic adenocarcinoma: Preliminary Experience and Implications for Patient C. Radiology 2016, 281, 816–825. [Google Scholar] [CrossRef]
  78. Tao, Q.; Zhu, W.; Zhao, X.; Li, M.; Shu, Y.; Wang, D.; Li, X. Perineural Invasion and Postoperative Adjuvant Chemotherapy Efficacy in Patients With Gastric Cancer. Front. Oncol. 2020, 10, 530. [Google Scholar] [CrossRef] [Green Version]
  79. Bakst, R.L.; Lee, N.; He, S.; Chernichenko, N.; Chen, C.H.; Linkov, G.; Le, H.C.; Koutcher, J.; Vakiani, E.; Wong, R.J. Radiation impairs perineural invasion by modulating the nerve microenvironment. PLoS ONE 2012, 7, e39925. [Google Scholar] [CrossRef] [Green Version]
  80. Li, X.; Ma, G.; Ma, Q.; Li, W.; Liu, J.; Han, L.; Duan, W.; Xu, Q.; Liu, H.; Wang, Z.; et al. Neurotransmitter substance P mediates pancreatic cancer perineural invasion via NK-1R in cancer cells. Mol. Cancer Res. 2013, 11, 294–302. [Google Scholar] [CrossRef] [Green Version]
  81. Kolokythas, A.; Cox, D.P.; Dekker, N.; Schmidt, B.L. Nerve Growth Factor and Tyrosine Kinase A Receptor in Oral Squamous Cell Carcinoma: Is There an Association With Perineural Invasion? J. Oral Maxillofac. Surg. 2010, 68, 1290–1295. [Google Scholar] [CrossRef]
  82. Pundavela, J.; Roselli, S.; Faulkner, S.; Attia, J.; Scott, R.J.; Thorne, R.F.; Forbes, J.F.; Bradshaw, R.A.; Walker, M.M.; Jobling, P.; et al. Nerve fibers infiltrate the tumor microenvironment and are associated with nerve growth factor production and lymph node invasion in breast cancer. Mol. Oncol. 2015, 9, 1626–1635. [Google Scholar] [CrossRef] [Green Version]
  83. Hayakawa, Y.; Sakitani, K.; Konishi, M.; Asfaha, S.; Jiang, Z.; Tanaka, T.; Dubeykovskaya, Z.A.; Chen, X.; Urbanska, A.M.; Nagar, K.; et al. Nerve growth factor promotes gastric tumorigenesis through aberrant cholinergic signaling. Cancer Cell 2017, 31, 21–34. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Kowalski, P.J.; Paulino, A.F.G. Perineural invasion in adenoid cystic carcinoma: Its causation/promotion by brain-derived neurotrophic factor. Hum. Pathol. 2002, 33, 933–936. [Google Scholar] [CrossRef] [PubMed]
  85. Lv, C.Y.; Zhou, T.; Chen, W.; Yin, X.D.; Yao, J.H.; Zhang, Y.F. Preliminary study correlating CX3CL1/CX3CR1 expression with gastric carcinoma and gastric carcinoma perineural invasion. World J. Gastroenterol. 2014, 20, 4428–4432. [Google Scholar] [CrossRef] [PubMed]
  86. He, S.; He, S.; Chen, C.H.; Deborde, S.; Bakst, R.L.; Chernichenko, N.; McNamara, W.F.; Lee, S.Y.; Barajas, F.; Yu, Z.; et al. The chemokine (CCL2-CCR2) signaling axis mediates perineural invasion. Mol. Cancer Res. 2015, 13, 380–390. [Google Scholar] [CrossRef] [Green Version]
  87. Huang, T.; Fan, Q.; Wang, Y.; Cui, Y.; Wang, Z.; Yang, L.; Sun, X.; Wang, Y. Schwann Cell-Derived CCL2 Promotes the Perineural Invasion of Cervical Cancer. Front. Oncol. 2020, 10, 19. [Google Scholar] [CrossRef]
  88. Madeo, M.; Colbert, P.L.; Vermeer, D.W.; Lucido, C.T.; Cain, J.T.; Vichaya, E.G.; Grossberg, A.J.; Muirhead, D.R.; Rickel, A.P.; Hong, Z.; et al. Cancer exosomes induce tumor innervation. Nat. Commun. 2018, 9, 4284. [Google Scholar] [CrossRef] [Green Version]
  89. Demir, I.E.; Ceyhan, G.O.; Liebl, F.; D’Haese, J.G.; Maak, M.; Friess, H. Neural invasion in pancreatic cancer: The past, present and future. Cancers 2010, 2, 1513–1527. [Google Scholar] [CrossRef] [Green Version]
  90. Ni, T.; Huang, T.; Gu, S.L.; Wang, J.; Liu, Y.; Sun, X.; Wang, Y.D. DRG neurons promote perineural invasion of endometrial cancer via GluR2. J. Cancer 2020, 11, 2518–2528. [Google Scholar] [CrossRef]
  91. Sejda, A.; Sigorski, D.; Gulczyński, J.; Wesołowski, W.; Kitlińska, J.; Iżycka-Świeszewska, E. Complexity of Neural Component of Tumor Microenvironment in Prostate Cancer. Pathobiology 2020, 87, 87–99. [Google Scholar] [CrossRef]
  92. Li, J.; Ma, J.; Han, L.; Xu, Q.; Lei, J.; Duan, W.; Li, W.; Wang, F.; Wu, E.; Ma, Q.; et al. Hyperglycemic tumor microenvironment induces perineural invasion in pancreatic cancer. Cancer Biol. Ther. 2015, 16, 912–921. [Google Scholar] [CrossRef] [Green Version]
  93. Zhang, L.; Zhang, W.; Zhang, X.; Yihe, M.I.N.; Zhao, Y.; Wang, B.; Wei, L.I.; Shuai, M.A.O.; Weili, M.I.N. High-glucose microenvironment promotes perineural invasion of pancreatic cancer via activation of hypoxia inducible factor 1α. Oncol. Rep. 2022, 47, 1–11. [Google Scholar] [CrossRef] [PubMed]
  94. Fantin, V.R.; St-Pierre, J.; Leder, P. Attenuation of LDH-A expression uncovers a link between glycolysis, mitochondrial physiology, and tumor maintenance. Cancer Cell 2006, 9, 425–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Wang, Z.H.; Peng, W.B.; Zhang, P.; Yang, X.P.; Zhou, Q. Lactate in the tumour microenvironment: From immune modulation to therapy. EBioMedicine 2021, 73, 103627. [Google Scholar] [CrossRef] [PubMed]
  96. Krall, A.S.; Xu, S.; Graeber, T.G.; Braas, D.; Christofk, H.R. Asparagine promotes cancer cell proliferation through use as an amino acid exchange factor. Nat. Commun. 2016, 7, 11457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Chen, C.L.; Hsu, S.C.; Ann, D.K.; Yen, Y.; Kung, H.J. Arginine signaling and cancer metabolism. Cancers 2021, 13, 3541. [Google Scholar] [CrossRef]
  98. Alothaim, T.; Charbonneau, M.; Tang, X. HDAC6 inhibitors sensitize non-mesenchymal triple-negative breast cancer cells to cysteine deprivation. Sci. Rep. 2021, 11, 10956. [Google Scholar] [CrossRef]
  99. Labuschagne, C.F.; van den Broek, N.J.F.; Mackay, G.M.; Vousden, K.H.; Maddocks, O.D.K. Serine, but not glycine, supports one-carbon metabolism and proliferation of cancer cells. Cell Rep. 2014, 7, 1248–1258. [Google Scholar] [CrossRef] [Green Version]
  100. Maneikyte, J.; Bausys, A.; Leber, B.; Horvath, A.; Feldbacher, N.; Hoefler, G.; Strupas, K.; Stiegler, P.; Schemmer, P. Dietary glycine decreases both tumor volume and vascularization in a combined colorectal liver metastasis and chemotherapy model. Int. J. Biol. Sci. 2019, 15, 1582–1590. [Google Scholar] [CrossRef] [Green Version]
  101. Cheng, C.T.; Qi, Y.; Wang, Y.C.; Chi, K.K.; Chung, Y.; Ouyang, C.; Chen, Y.R.; Oh, M.E.; Sheng, X.; Tang, Y.; et al. Arginine starvation kills tumor cells through aspartate exhaustion and mitochondrial dysfunction. Commun. Biol. 2018, 1, 178. [Google Scholar] [CrossRef] [Green Version]
  102. Maddocks, O.D.K.; Berkers, C.R.; Mason, S.M.; Zheng, L.; Blyth, K. Serine starvation induces stress and p53 dependent metabolic remodeling in cancer cells. Nature 2019, 493, 542–546. [Google Scholar] [CrossRef]
  103. Ma, J.L.; Zhao, Y.; Guo, C.Y.; Hu, H.T.; Zheng, L.; Zhao, E.J.; Li, H.L. Dietary vitamin B intake and the risk of esophageal cancer: A meta-analysis. Cancer Manag. Res. 2018, 10, 5395–5410. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Ness, R.A.; Miller, D.D.; Li, W. The role of vitamin D in cancer prevention. Chin. J. Nat. Med. 2015, 13, 481–497. [Google Scholar] [CrossRef] [Green Version]
  105. Fulan, H.; Changxing, J.; Yi Baina, W.; Wencui, Z.; Chunqing, L.; Fan, W.; Dandan, L.; Dianjun, S.; Tong, W.; Da, P.; et al. Retinol, vitamins A, C, and e and breast cancer risk: A meta-analysis and meta-regression. Cancer Causes Control 2011, 22, 1383–1396. [Google Scholar] [CrossRef] [PubMed]
  106. Fusaro, M.; Gallieni, M.; Porta, C.; Nickolas, T.L.; Khairallah, P. Vitamin K effects in human health: New insights beyond bone and cardiovascular health. J. Nephrol. 2020, 33, 239–249. [Google Scholar] [CrossRef]
  107. Ngo, B.; Van Riper, J.M.; Cantley, L.C.; Yun, J. Targeting cancer vulnerabilities with high-dose vitamin C. Nat. Rev. Cancer 2019, 19, 271–282. [Google Scholar] [CrossRef]
  108. Wu, Y.; Ye, Y.; Shi, Y.; Li, P.; Xu, J.; Chen, K.; Xu, E.; Yang, J. Association between vitamin A, retinol intake and blood retinol level and gastric cancer risk: A meta-analysis. Clin. Nutr. 2015, 34, 620–626. [Google Scholar] [CrossRef]
  109. Abar, L.; Vieira, A.R.; Aune, D.; Stevens, C.; Vingeliene, S.; Navarro Rosenblatt, D.A.; Chan, D.; Greenwood, D.C.; Norat, T. Blood concentrations of carotenoids and retinol and lung cancer risk: An update of the WCRF–AICR systematic review of published prospective studies. Cancer Med. 2016, 5, 2069–2083. [Google Scholar] [CrossRef] [Green Version]
  110. Comín-Anduix, B.; Boren, J.; Martinez, S.; Moro, C.; Centelles, J.J.; Trebukhina, R.; Petushok, N.; Lee, W.N.P.; Boros, L.G.; Cascante, M. The effect of thiamine supplementation on tumour proliferation: A metabolic control analysis study. Eur. J. Biochem. 2001, 268, 4177–4182. [Google Scholar] [CrossRef]
  111. Liu, S.; Huang, H.; Lu, X.; Golinski, M.; Comesse, S.; Watt, D.; Grossman, R.B.; Moscow, J.A. Down-regulation of thiamine transporter THTR2 gene expression in breast cancer and its association with resistance to apoptosis. Mol. Cancer Res. 2003, 1, 665–673. [Google Scholar]
  112. Ng, E.K.O.; Leung, C.P.H.; Shin, V.Y.; Wong, C.L.P.; Ma, E.S.K.; Jin, H.C.; Chu, K.M.; Kwong, A. Quantitative analysis and diagnostic significance of methylated SLC19A3 DNA in the plasma of breast and gastric cancer patients. PLoS ONE 2011, 6, e22233. [Google Scholar] [CrossRef] [Green Version]
  113. Arendt, J.F.H.; Sørensen, H.T.; Horsfall, L.J.; Petersen, I. Elevated vitamin B12 levels and cancer risk in UK primary care: A thin database cohort study. Cancer Epidemiol. Biomark. Prev. 2019, 28, 814–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Fanidi, A.; Carreras-Torres, R.; Larose, T.L.; Yuan, J.M.; Stevens, V.L.; Weinstein, S.J.; Albanes, D.; Prentice, R.; Pettinger, M.; Cai, Q.; et al. Is high vitamin B12 status a cause of lung cancer? Int. J. Cancer 2019, 145, 1499–1503. [Google Scholar] [CrossRef] [Green Version]
  115. Zeng, L.H.; Wang, Q.M.; Feng, L.Y.; Ke, Y.D.; Xu, Q.Z.; Wei, A.Y.; Zhang, C.; Ying, R.B. High-dose vitamin C suppresses the invasion and metastasis of breast cancer cells via inhibiting epithelial-mesenchymal transition. Onco Targets Ther. 2019, 12, 7405–7413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Yang, G.; Yan, Y.; Ma, Y.; Yang, Y. Vitamin C at high concentrations induces cytotoxicity in malignant melanoma but promotes tumor growth at low concentrations. Mol. Carcinog. 2017, 56, 1965–1976. [Google Scholar] [CrossRef] [PubMed]
  117. van den Bemd, G.J.C.M.; Chang, G.T.G. Vitamin D and Vitamin D Analogs in Cancer Treatment. Curr. Drug Targets 2005, 3, 85–94. [Google Scholar] [CrossRef]
  118. Milczarek, M.; Psurski, M.; Kutner, A.; Wietrzyk, J. Vitamin D analogs enhance the anticancer activity of 5-fluorouracil in an in vivo mouse colon cancer model. BMC Cancer 2013, 13, 294. [Google Scholar] [CrossRef] [Green Version]
  119. Gorham, E.D.; Garland, C.F.; Garland, F.C.; Grant, W.B.; Mohr, S.B.; Lipkin, M.; Newmark, H.L.; Giovannucci, E.; Wei, M.; Holick, M.F. Optimal Vitamin D Status for Colorectal Cancer Prevention. A Quantitative Meta Analysis. Am. J. Prev. Med. 2007, 32, 210–216. [Google Scholar] [CrossRef]
  120. Wang, Y.; Chiang, Y.H.; Su, T.P.; Hayashi, T.; Morales, M.; Hoffer, B.J.; Lin, S.Z. Vitamin D3 attenuates cortical infarction induced by middle cerebral arterial ligation in rats. Neuropharmacology 2000, 39, 873–880. [Google Scholar] [CrossRef]
  121. Kannappan, R.; Gupta, S.C.; Kim, J.H.; Aggarwal, B.B. Tocotrienols fight cancer by targeting multiple cell signaling pathways. Genes Nutr. 2012, 7, 43–52. [Google Scholar] [CrossRef] [Green Version]
  122. Yap, W.N.; Zaiden, N.; Tan, Y.L.; Ngoh, C.P.; Zhang, X.W.; Wong, Y.C.; Ling, M.T.; Yap, Y.L. Id1, inhibitor of differentiation, is a key protein mediating anti-tumor responses of gamma-tocotrienol in breast cancer cells. Cancer Lett. 2010, 291, 187–199. [Google Scholar] [CrossRef]
  123. Xu, W.L.; Liu, J.R.; Liu, H.K.; Qi, G.Y.; Sun, X.R.; Sun, W.G.; Chen, B.Q. Inhibition of proliferation and induction of apoptosis by γ-tocotrienol in human colon carcinoma HT-29 cells. Nutrition 2009, 25, 555–566. [Google Scholar] [CrossRef] [PubMed]
  124. Qiu, F.; Chen, Y.-R.; Liu, X.; Chu, C.-Y.; Shen, L.-J.; Xu, J.; Gaur, S.; Forman, H.J.; Zhang, H.; Zheng, S.; et al. Arginine Starvation Impairs Mitochondrial Respiratory Function in ASS1-Deficient Breast Cancer Cells. Sci. Signal. 2015, 7, ra31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Ji, J.X.; Cochrane, D.R.; Tessier-Cloutier, B.; Chen, S.Y.; Ho, G.; Pathak, K.V.; Alcazar, I.N.; Farnell, D.; Leung, S.; Cheng, A.; et al. Arginine Depletion Therapy with ADI-PEG20 Limits Tumor Growth in Argininosuccinate Synthase–Deficient Ovarian Cancer, including Small-Cell Carcinoma of the Ovary, Hypercalcemic Type. Clin. Cancer Res. 2020, 26, 4402–4413. [Google Scholar] [CrossRef] [PubMed]
  126. Selvi, I.; Basar, H.; Baydilli, N.; Murat, K.; Kaymaz, O. The importance of plasma arginine level and its downstream metabolites in diagnosing prostate cancer. Int. Urol. Nephrol. 2019, 51, 1975–1983. [Google Scholar] [CrossRef]
  127. Mukhopadhyay, S.; Biancur, D.E.; Parker, S.J.; Yamamoto, K.; Banh, R.S.; Paulo, J.A.; Mancias, J.D.; Kimmelman, A.C. Autophagy is required for proper cysteine homeostasis in pancreatic cancer through regulation of SLC7A11. Proc. Natl. Acad. Sci. USA 2021, 118, e2021475118. [Google Scholar] [CrossRef]
  128. Fan, T.W.M.; Bruntz, R.C.; Yang, Y.; Song, H.; Chernyavskaya, Y.; Deng, P.; Zhang, Y.; Shah, P.P.; Beverly, L.J.; Qi, Z.; et al. De novo synthesis of serine and glycine fuels purine nucleotide biosynthesis in human lung cancer tissues. J. Biol. Chem. 2019, 294, 13464–13477. [Google Scholar] [CrossRef]
  129. Bonuccelli, G.; Tsirigos, A.; Whitaker-Menezes, D.; Pavlides, S.; Pestell, R.G.; Chiavarina, B.; Frank, P.G.; Flomenberg, N.; Howell, A.; Martinez-Outschoorn, U.E.; et al. Ketones and lactate “fuel” tumor growth and metastasis: Evidence that epithelial cancer cells use oxidative mitochondrial metabolism. Cell Cycle 2010, 9, 3506–3514. [Google Scholar] [CrossRef]
  130. San-Millán, I.; Julian, C.G.; Matarazzo, C.; Martinez, J.; Brooks, G.A. Is Lactate an Oncometabolite? Evidence Supporting a Role for Lactate in the Regulation of Transcriptional Activity of Cancer-Related Genes in MCF7 Breast Cancer Cells. Front. Oncol. 2020, 9, 1536. [Google Scholar] [CrossRef]
  131. Dikshit, A.; Lu, J.; Ford, A.E.; Degan, S.; Jin, Y.J.; Sun, H.; Nichols, A.; Salama, A.K.S.; Beasley, G.; Gooden, D.; et al. Potential Utility of Synthetic D-Lactate Polymers in Skin Cancer. JID Innov. 2021, 1, 100043. [Google Scholar] [CrossRef]
  132. Apicella, M.; Giannoni, E.; Fiore, S.; Ferrari, K.J.; Fernández-Pérez, D.; Isella, C.; Granchi, C.; Minutolo, F.; Sottile, A.; Comoglio, P.M.; et al. Increased Lactate Secretion by Cancer Cells Sustains Non-cell-autonomous Adaptive Resistance to MET and EGFR Targeted Therapies. Cell Metab. 2018, 28, 848–865.e6. [Google Scholar] [CrossRef] [Green Version]
  133. Yang, Y.; Chong, Y.; Chen, M.; Dai, W.; Zhou, X.; Ji, Y.; Qiu, G.; Du, X. Targeting lactate dehydrogenase a improves radiotherapy efficacy in non-small cell lung cancer: From bedside to bench. J. Transl. Med. 2021, 19, 170. [Google Scholar] [CrossRef] [PubMed]
  134. Ngo, B.; Kim, E.; Osorio-Vasquez, V.; Doll, S.; Bustraan, S.; Liang, R.J.; Luengo, A.; Davidson, S.M.; Ali, A.; Ferraro, G.B.; et al. Limited environmental serine and glycine confer brain metastasis sensitivity to PHGDH inhibition. Cancer Discov. 2020, 10, 1352–1373. [Google Scholar] [CrossRef] [PubMed]
  135. Zhao, X.; Fu, J.; Hu, B.; Chen, L.; Wang, J.; Fang, J.; Ge, C.; Lin, H.; Pan, K.; Fu, L.; et al. Serine Metabolism Regulates YAP Activity Through USP7 in Colon Cancer. Front. Cell Dev. Biol. 2021, 9, 639111. [Google Scholar] [CrossRef]
  136. Meiser, J.; Tumanov, S.; Maddocks, O.; Labuschagne, C.F.; Athineos, D.; Van Den Broek, N.; Mackay, G.M.; Gottlieb, E.; Blyth, K.; Vousden, K.; et al. Serine one-carbon catabolism with formate overflow. Sci. Adv. 2016, 2, e1601273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Goodwin, M.L.; Gladden, L.B.; Nijsten, M.W.N.; Jones, K.B. Lactate and Cancer: Revisiting the Warburg Effect in an Era of Lactate Shuttling. Front. Nutr. 2015, 1, 2014–2016. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  138. de la Cruz-López, K.G.; Castro-Muñoz, L.J.; Reyes-Hernández, D.O.; García-Carrancá, A.; Manzo-Merino, J. Lactate in the Regulation of Tumor Microenvironment and Therapeutic Approaches. Front. Oncol. 2019, 9, 1143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  139. Tafur, D.; Svrcek, P.; White, B. Differential expression and function of the endogenous lactate receptor, GPR81, in ER alpha-positive/HER2-positive epithelial vs. post-EMT triple-negative mesenchymal breast cancer cells. J. Cancer Metastasis Treat. 2019, 5, 46. [Google Scholar] [CrossRef] [Green Version]
  140. Hou, X.; Shi, X.; Zhang, W.; Li, D.; Hu, L.; Yang, J.; Zhao, J.; Wei, S.; Wei, X.; Ruan, X.; et al. LDHA induces EMT gene transcription and regulates autophagy to promote the metastasis and tumorigenesis of papillary thyroid carcinoma. Cell Death Dis. 2021, 12, 347. [Google Scholar] [CrossRef]
  141. Siddikuzzaman, C.G.; Berlin Grace, V.M. All trans retinoic acid and cancer. Immunopharmacol. Immunotoxicol. 2011, 33, 241–249. [Google Scholar] [CrossRef]
  142. Niles, R.M. Signaling pathways in retinoid chemoprevention and treatment of cancer. Mutat. Res. Fundam. Mol. Mech. Mutagen. 2004, 555, 97–105. [Google Scholar] [CrossRef]
  143. Huang, Z.; Liu, Y.; Qi, G.; Brand, D.; Zheng, S.G. Role of vitamin A in the immune system. J. Clin. Med. 2018, 7, 258. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Ryan-Harshman, M.; Aldoori, W. Vitamin B12 and health. Can. Fam. Physician 2008, 54, 536–541. [Google Scholar] [PubMed]
  145. Vitamin B12: Fact Sheet for Health Professionals. Available online: https://ods.od.nih.gov/factsheets/VitaminB12-HealthProfessional/ (accessed on 14 March 2022).
  146. Paul, C.; Brady, D.M. Comparative Bioavailability and Utilization of Particular Forms of B12 Supplements with Potential to Mitigate B12-related Genetic Polymorphisms. Integr. Med. 2017, 16, 42–49. [Google Scholar]
  147. Weissbach, H.; Dickerman, H. Biochemical Role of Vitamin B12. Physiol. Rev. 1965, 45, 80–97. [Google Scholar] [CrossRef] [PubMed]
  148. Hammond, N.; Wang, Y.; Dimachkie, M.M.; Barohn, R.J. Nutritional neuropathies. Neurol. Clin. 2013, 31, 477–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Alberg, A.J.; Selhub, J.; Shah, K.V.; Viscidi, R.P.; Comstock, G.W.; Helzlsouer, K.J. The risk of cervical cancer in relation to serum concentrations of folate, vitamin B12, and homocysteine. Cancer Epidemiol. Biomark. Prev. 2000, 9, 761–764. [Google Scholar]
  150. Ames, B.N. DNA damage from micronutrient deficiencies is likely. Univ. Calif. Berkeley 2001, 475, 7–20. [Google Scholar]
  151. Ikehata, M.; Ueda, K.; Iwakawa, S. Different involvement of DNA methylation and histone deacetylation in the expression of solute-carrier transporters in 4 colon cancer cell lines. Biol. Pharm. Bull. 2012, 35, 301–307. [Google Scholar] [CrossRef] [Green Version]
  152. Kikuchi, D.; Minamishima, Y.A.; Nakayama, K. Prolyl-hydroxylase PHD3 interacts with pyruvate dehydrogenase (PDH)-E1β and regulates the cellular PDH activity. Biochem. Biophys. Res. Commun. 2014, 451, 288–294. [Google Scholar] [CrossRef]
  153. Lương, K.V.Q.; Nguyễn, L.T.H. Genetic and Cellular Signaling Mechanisms The Role of Thiamine in Cancer: Possible. Cancer Genom. Proteom. 2013, 10, 169–186. [Google Scholar] [CrossRef]
  154. Yonashiro, R.; Eguchi, K.; Wake, M.; Takeda, N.; Nakayama, K. Pyruvate dehydrogenase PDH-E1b controls tumor progression by altering the metabolic status of cancer cells. Cancer Res. 2018, 78, 1592–1603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Verrax, J.; Buc Calderon, P. The controversial place of vitamin C in cancer treatment. Biochem. Pharmacol. 2008, 76, 1644–1652. [Google Scholar] [CrossRef] [PubMed]
  156. Roa, F.J.; Peña, E.; Gatica, M.; Escobar-Acuña, K.; Saavedra, P.; Maldonado, M.; Cuevas, M.E.; Moraga-Cid, G.; Rivas, C.I.; Muñoz-Montesino, C. Therapeutic Use of Vitamin C in Cancer: Physiological Considerations. Front. Pharmacol. 2020, 11, 211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Fritz, H.; Flower, G.; Weeks, L.; Cooley, K.; Callachan, M.; McGowan, J.; Skidmore, B.; Kirchner, L.; Seely, D. Intravenous vitamin C and cancer: A systematic review. Integr. Cancer Ther. 2014, 13, 280–300. [Google Scholar] [CrossRef] [PubMed]
  158. Gillberg, L.; Ørskov, A.D.; Liu, M.; Harsløf, L.B.S.; Jones, P.A.; Grønbæk, K. Vitamin C—A new player in regulation of the cancer epigenome. Semin. Cancer Biol. 2018, 51, 59–67. [Google Scholar] [CrossRef] [PubMed]
  159. Carr, A.C.; Cook, J. Intravenous vitamin C for cancer therapy—Identifying the current gaps in our knowledge. Front. Physiol. 2018, 9, 1182. [Google Scholar] [CrossRef]
  160. Park, S.; Ahn, S.; Shin, Y.; Yang, Y.; Yeom, C.H. Vitamin C in cancer: A metabolomics perspective. Front. Physiol. 2018, 9, 762. [Google Scholar] [CrossRef] [Green Version]
  161. Vissers, M.C.M.; Das, A.B. Potential mechanisms of action for vitamin C in cancer: Reviewing the evidence. Front. Physiol. 2018, 9, 809. [Google Scholar] [CrossRef] [Green Version]
  162. Pawlowska, E.; Szczepanska, J.; Blasiak, J. Pro- and antioxidant effects of Vitamin C in cancer in correspondence to its dietary and pharmacological concentrations. Oxid. Med. Cell. Longev. 2019, 2019, 7286737. [Google Scholar] [CrossRef] [Green Version]
  163. Bao, Y.; Li, Y.; Gong, Y.; Huang, Q.; Cai, S.; Peng, J. Vitamin D Status and Survival in Stage II-III Colorectal Cancer. Front. Oncol. 2020, 10, 581597. [Google Scholar] [CrossRef]
  164. Jeon, S.M.; Shin, E.A. Exploring vitamin D metabolism and function in cancer. Exp. Mol. Med. 2018, 50, 1–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Zmijewski, M.A. Vitamin D and human health. Int. J. Mol. Sci. 2019, 20, 145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Mena, J.M.O.; Brenner, H. Vitamin D and cancer: An overview on epidemiological studies. Adv. Exp. Med. Biol. 2014, 810, 17–32. [Google Scholar]
  167. Szkandera, J.; Absenger, G.; Pichler, M.; Stotz, M.; Langsenlehner, T.; Samonigg, H.; Renner, W.; Gerger, A. Association of common gene variants in vitamin D modulating genes and colon cancer recurrence. J. Cancer Res. Clin. Oncol. 2013, 139, 1457–1464. [Google Scholar] [CrossRef] [PubMed]
  168. Sun, H.; Wang, C.; Hao, M.; Sun, R.; Wang, Y.; Liu, T.; Cong, X.; Liu, Y. CYP24A1 is a potential biomarker for the progression and prognosis of human colorectal cancer. Hum. Pathol. 2016, 50, 101–108. [Google Scholar] [CrossRef]
  169. Yang, C.S.; Suh, N.; Kong, A.N.T. Does vitamin E prevent or promote cancer? Cancer Prev. Res. 2012, 5, 701–705. [Google Scholar] [CrossRef] [Green Version]
  170. Kanchi, M.M.; Shanmugam, M.K.; Rane, G.; Sethi, G.; Kumar, A.P. Tocotrienols: The unsaturated sidekick shifting new paradigms in vitamin E therapeutics. Drug Discov. Today 2017, 22, 1765–1781. [Google Scholar] [CrossRef]
  171. Constantinou, C.; Charalambous, C.; Kanakis, D. Vitamin E and cancer: An update on the emerging role of γ and δ tocotrienols. Eur. J. Nutr. 2020, 59, 845–857. [Google Scholar] [CrossRef]
  172. Peh, H.Y.; Tan, W.S.D.; Liao, W.; Wong, W.S.F. Vitamin E therapy beyond cancer: Tocopherol versus tocotrienol. Pharmacol. Ther. 2016, 162, 152–169. [Google Scholar] [CrossRef]
  173. Jiang, Q. Natural forms of vitamin E as effective agents for cancer prevention and therapy. Adv. Nutr. 2017, 8, 850–867. [Google Scholar] [CrossRef]
  174. Chang, P.N.; Yap, W.N.; Wing Lee, D.T.; Ling, M.T.; Wong, Y.C.; Yap, Y.L. Evidence of -tocotrienol as an apoptosis-inducing, invasion-suppressing, and chemotherapy drug-sensitizing agent in human melanoma cells. Nutr. Cancer 2009, 61, 357–366. [Google Scholar] [CrossRef]
  175. Juanola-Falgarona, M.; Salas-Salvadó, J.; Martínez-Gonzaĺez, M.A.; Corella, D.; Estruch, R.; Ros, E.; Fitó, M.; Arós, F.; Gómez-Gracia, E.; Fiol, M.; et al. Dietary intake of vitamin K is inversely associated with mortality risk. J. Nutr. 2014, 144, 743–750. [Google Scholar] [CrossRef] [PubMed]
  176. Dahlberg, S.; Ede, J.; Schött, U. Vitamin K and cancer. Scand. J. Clin. Lab. Investig. 2017, 77, 555–567. [Google Scholar] [CrossRef]
  177. Nimptsch, K.; Rohrmann, S.; Kaaks, R.; Linseisen, J. Dietary vitamin K intake in relation to cancer incidence and mortality: Results from the Heidelberg cohort of the European Prospective Investigation into Cancer and Nutrition (EPIC-Heidelberg). Am. J. Clin. Nutr. 2010, 91, 1348–1358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. Postles, J.; Curtis, T.Y.; Powers, S.J.; Elmore, J.S.; Mottram, D.S.; Halford, N.G. Changes in free amino acid concentration in rye grain in response to nitrogen and sulfur availability, and expression analysis of genes involved in asparagine metabolism. Front. Plant Sci. 2016, 7, 917. [Google Scholar] [CrossRef] [PubMed]
  179. Sabatini, D.M. Twenty-five years of mTOR: Uncovering the link from nutrients to growth. Proc. Natl. Acad. Sci. USA 2017, 114, 11818–11825. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Knott, S.R.V.; Wagenblast, E.; Khan, S.; Kim, S.Y.; Soto, M.; Wagner, M.; Turgeon, M.O.; Fish, L.; Erard, N.; Gable, A.L.; et al. Asparagine bioavailability governs metastasis in a model of breast cancer. Nature 2018, 554, 378–381. [Google Scholar] [CrossRef]
  181. Lorenzi, P.L.; Reinhold, W.C.; Rudelius, M.; Gunsior, M.; Shankavaram, U.; Bussey, K.J.; Scherf, U.; Eichler, G.S.; Martin, S.E.; Chin, K.; et al. Asparagine synthetase as a causal, predictive biomarker for L-asparaginase activity in ovarian cancer cells. Mol. Cancer Ther. 2006, 5, 2613–2623. [Google Scholar] [CrossRef] [Green Version]
  182. Brumano, L.P.; da Silva, F.V.; Costa-Silva, T.A.; Apolinário, A.C.; Santos, J.H.; Kleingesinds, E.K.; Monteiro, G.; Rangel-Yagui, C.D.; Benyahia, B.; Junior, A.P. Development of L-asparaginase biobetters: Current research status and review of the desirable quality profiles. Front. Bioeng. Biotechnol. 2019, 6, 212. [Google Scholar] [CrossRef] [Green Version]
  183. Garcia-Bermudez, J.; Williams, R.T.; Guarecuco, R.; Birsoy, K. Targeting extracellular nutrient dependencies of cancer cells. Mol. Metab. 2020, 33, 67–82. [Google Scholar] [CrossRef]
  184. Clarkson, B.; Krakoff, I.; Burchenal, J.; Karnofsky, D.; Golby, R.; Dowling, M.; Oettegen, H.; Lipton, A. Clinical Results of Treatment with E. Coli L-Asparaginase in Adults with Leukemia, Lymphoma, and Solid Tumors. Cancer 1969, 25, 279–305. [Google Scholar] [CrossRef]
  185. Su, N.; Pan, Y.-X.; Zhou, M.; Harvey, R.; Hunger, S.; Kilberg, M. Correlation between Asparaginase Sensitivity and Asparagine Synthetase Protein Content, but not mRNA, in Acute Lymphoblastic Leukemia Cell Lines. Pediatr. Blood Cancer 2008, 50, 1018–1025. [Google Scholar] [CrossRef] [PubMed]
  186. Chan, W.K.; Horvath, T.D.; Tan, L.; Link, T.; Harutyunyan, K.G.; Pontikos, M.A.; Anishkin, A.; Du, D.; Martin, L.A.; Yin, E.; et al. Glutaminase activity of L-asparaginase contributes to durable preclinical activity against acute lymphoblastic leukemia. Mol. Cancer Ther. 2019, 18, 1587–1592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Hlozkova, K.; Pecinova, A.; Alquezar-Artieda, N.; Pajuelo-Reguera, D.; Simcikova, M.; Hovorkova, L.; Rejlova, K.; Zaliova, M.; Mracek, T.; Kolenova, A.; et al. Metabolic profile of leukemia cells influences treatment efficacy of L-asparaginase. BMC Cancer 2020, 20, 526. [Google Scholar] [CrossRef] [PubMed]
  188. Albaugh, V.L.; Pinzon-Guzman, C.; Barbul, A. Arginine Metabolism and Cancer. J. Surg. Oncol. 2017, 115, 273–280. [Google Scholar] [CrossRef] [PubMed]
  189. Shi, L.Y.; Wang, Y.Y.; Jing, Y.; Xu, M.H.; Zhu, Z.T.; Wang, Q.J. Abnormal arginine metabolism is associated with prognosis in patients of gastric cancer. Transl. Cancer Res. 2021, 10, 2451–2469. [Google Scholar] [CrossRef] [PubMed]
  190. Hu, L.; Gao, Y.; Cao, Y.; Zhang, Y.; Xu, M.; Wang, Y.; Jing, Y.; Guo, S.; Jing, F.; Hu, X.; et al. Association of plasma arginine with breast cancer molecular subtypes in women of Liaoning province. IUBMB Life 2016, 68, 980–984. [Google Scholar] [CrossRef]
  191. Mirmiran, P.; Moghadam, S.K.; Bahadoran, Z.; Ghasemi, A.; Azizi, F. Dietary L-Arginine Intakes and the Risk of Metabolic Syndrome: A 6-Year Follow-Up in Tehran Lipid and Glucose Study. Prev. Nutr. Food Sci. 2017, 22, 263–270. [Google Scholar] [CrossRef] [Green Version]
  192. Izzo, F.; Marra, P.; Beneduce, G.; Castello, G.; Vallone, P.; De Rosa, V.; Cremona, F.; Ensor, C.M.; Holtsberg, F.W.; Bomalaski, J.S.; et al. Pegylated arginine deiminase treatment of patients with unresectable hepatocellular carcinoma: Results from phase I/II studies. J. Clin. Oncol. 2004, 22, 1815–1822. [Google Scholar] [CrossRef]
  193. Kim, S.K.; Jung, W.H.; Koo, J.S. Differential expression of enzymes associated with serine/glycine metabolism in different breast cancer subtypes. PLoS ONE 2014, 9, e101004. [Google Scholar] [CrossRef]
  194. Perry, C.; Yu, S.; Chen, J.; Matharu, K.S.; Stover, P.J. Effect of vitamin B6 availability on serine hydroxymethyltransferase in MCF-7 cells. Arch. Biochem. Biophys. 2007, 462, 21–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Dolfi, S.C.; Chan, L.L.-Y.; Qiu, J.; Tedeschi, P.M.; Bertino, J.R.; Hirshfield, K.M.; Oltvai, Z.N.; Vazquez, A. The metabolic demands of cancer cells are coupled to their size and protein synthesis rates. Cancer Metab. 2013, 1, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  196. Locasale, J.W. Serine, Glycine and the one-carbon cycle. Nat. Rev Cancer 2013, 13, 572–583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Tajan, M.; Hennequart, M.; Cheung, E.C.; Zani, F.; Hock, A.K.; Legrave, N.; Maddocks, O.D.K.; Ridgway, R.A.; Athineos, D.; Suárez-Bonnet, A.; et al. Serine synthesis pathway inhibition cooperates with dietary serine and glycine limitation for cancer therapy. Nat. Commun. 2021, 12, 366. [Google Scholar] [CrossRef]
  198. Ducker, G.S.; Rabinowitz, J.D. One-Carbon Metabolism in Health and Disease. Cell Metab. 2017, 25, 27–42. [Google Scholar] [CrossRef] [Green Version]
  199. Asai, A.; Konno, M.; Koseki, J.; Taniguchi, M.; Vecchione, A.; Ishii, H. One-carbon metabolism for cancer diagnostic and therapeutic approaches. Cancer Lett. 2020, 470, 141–148. [Google Scholar] [CrossRef]
  200. Banjac, A.; Perisic, T.; Sato, H.; Seiler, A.; Bannai, S.; Weiss, N.; Kölle, P.; Tschoep, K.; Issels, R.D.; Daniel, P.T.; et al. The cystine/cysteine cycle: A redox cycle regulating susceptibility versus resistance to cell death. Oncogene 2008, 27, 1618–1628. [Google Scholar] [CrossRef] [Green Version]
  201. Lewerenz, J.; Hewett, S.J.; Huang, Y.; Lambros, M.; Gout, P.W.; Kalivas, P.W.; Massie, A.; Smolders, I.; Methner, A.; Pergande, M.; et al. The cystine/glutamate antiporter system xc- in health and disease: From molecular mechanisms to novel therapeutic opportunities. Antioxid. Redox Signal. 2013, 18, 522–555. [Google Scholar] [CrossRef] [Green Version]
  202. Novera, W.; Lee, Z.W.; Nin, D.S.; Dai, M.Z.Y.; Binte Idres, S.; Wu, H.; Damen, J.M.A.; Tan, T.Z.; Sim, A.Y.L.; Long, Y.C.; et al. Cysteine Deprivation Targets Ovarian Clear Cell Carcinoma Via Oxidative Stress and Iron-Sulfur Cluster Biogenesis Deficit. Antioxid. Redox Signal. 2020, 33, 1191–1208. [Google Scholar] [CrossRef]
  203. Cervantes-Villagrana, R.D.; Albores-García, D.; Cervantes-Villagrana, A.R.; García-Acevez, S.J. Tumor-induced neurogenesis and immune evasion as targets of innovative anti-cancer therapies. Signal Transduct. Target. Ther. 2020, 5, 99. [Google Scholar] [CrossRef]
  204. Brown, I.S. Pathology of Perineural Spread. J. Neurol. Surg. Part B Skull Base 2016, 77, 124–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  205. Sandforth, L.; Ammar, N.; Dinges, L.A.; Röcken, C.; Arlt, A.; Sebens, S.; Schäfer, H. Impact of the Monocarboxylate transporter-1 (MCT1)-mediated cellular import of lactate on stemness properties of human pancreatic adenocarcinoma cells. Cancers 2020, 12, 581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Gao, H.; Liang, J.; Duan, J.; Chen, L.; Li, H.; Zhen, T.; Zhang, F.; Dong, Y.; Shi, H.; Han, A. A Prognosis Marker SLC2A3 Correlates With EMT and Immune Signature in Colorectal Cancer. Front. Oncol. 2021, 11, 638099. [Google Scholar] [CrossRef] [PubMed]
  207. Banh, R.S.; Biancur, D.E.; Yamamoto, K.; Sohn, A.S.W.; Walters, B.; Kuljanin, M.; Gikandi, A.; Wang, H.; Mancias, J.D.; Schneider, R.J.; et al. Neurons Release Serine to Support mRNA Translation in Pancreatic Cancer. Cell 2020, 183, 1202–1218.e25. [Google Scholar] [CrossRef] [PubMed]
  208. El Soury, M.; Fornasari, B.E.; Carta, G.; Zen, F.; Haastert-Talini, K.; Ronchi, G. The role of dietary nutrients in peripheral nerve regeneration. Int. J. Mol. Sci. 2021, 22, 7417. [Google Scholar] [CrossRef] [PubMed]
  209. Lu, H.; Klaassen, C. Tissue distribution and thyroid hormone regulation of Pept1 and Pept2 mRNA in rodents. Peptides 2006, 27, 850–857. [Google Scholar] [CrossRef] [PubMed]
  210. Kim, E.; Jung, S.; Park, W.S.; Lee, J.H.; Shin, R.; Heo, S.C.; Choe, E.K.; Lee, J.H.; Kim, K.; Chai, Y.J. Upregulation of SLC2A3 gene and prognosis in colorectal carcinoma: Analysis of TCGA data. BMC Cancer 2019, 19, 302. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  211. Liu, J.; Hong, J.; Han, H.; Park, J.; Kim, D.; Park, H.; Ko, M.; Koh, Y.; Shin, D.Y.; Yoon, S.S. Decreased vitamin C uptake mediated by SLC2A3 promotes leukaemia progression and impedes TET2 restoration. Br. J. Cancer 2020, 122, 1445–1452. [Google Scholar] [CrossRef]
  212. Sprute, R.; Ardicli, D.; Oguz, K.K.; Malenica-Mandel, A.; Daimagüler, H.S.; Koy, A.; Coskun, T.; Wang, H.; Topcu, M.; Cirak, S. Clinical outcomes of two patients with a novel pathogenic variant in ASNS: Response to asparagine supplementation and review of the literature. Hum. Genome Var. 2019, 6, 24. [Google Scholar] [CrossRef] [Green Version]
  213. Ellerbroek, S.M.; Wennerberg, K.; Burridge, K. Serine phosphorylation negatively regulates RhoA in vivo. J. Biol. Chem. 2003, 278, 19023–19031. [Google Scholar] [CrossRef] [Green Version]
  214. Nusser, N.; Gosmanova, E.; Makarova, N.; Fujiwara, Y.; Yang, L.; Guo, F.; Luo, Y.; Zheng, Y.; Tigyi, G. Serine phosphorylation differentially affects RhoA binding to effectors: Implications to NGF-induced neurite outgrowth. Cell. Signal. 2006, 18, 704–714. [Google Scholar] [CrossRef] [PubMed]
  215. Zhou, L.; Too, H.P. Mitochondrial localized STAT3 is involved in NGF induced neurite outgrowth. PLoS ONE 2011, 6, e21680. [Google Scholar] [CrossRef] [PubMed]
  216. Tapia, J.C.; Mentis, G.Z.; Navarrete, R.; Nualart, F.; Figueroa, E.; Sánchez, A.; Aguayo, L.G. Early expression of glycine and GABAA receptors in developing spinal cord neurons. Effects on neurite outgrowth. Neuroscience 2001, 108, 493–506. [Google Scholar] [CrossRef]
  217. El Hayek, L.; Khalifeh, M.; Zibara, V.; Abi Assaad, R.; Emmanuel, N.; Karnib, N.; El-Ghandour, R.; Nasrallah, P.; Bilen, M.; Ibrahim, P.; et al. Lactate mediates the effects of exercise on learning and memory through sirt1-dependent activation of hippocampal brain-derived neurotrophic factor (BDNF). J. Neurosci. 2019, 39, 2369–2382. [Google Scholar] [CrossRef] [Green Version]
  218. Lezi, E.; Lu, J.; Selfridge, J.E.; Burns, J.M.; Swerdlow, R.H. Lactate administration reproduces specific brain and liver exercise-related changes. J. Neurochem. 2013, 127, 91–100. [Google Scholar] [CrossRef] [Green Version]
  219. García-Suárez, P.C.; Rentería, I.; Plaisance, E.P.; Moncada-Jiménez, J.; Jiménez-Maldonado, A. The effects of interval training on peripheral brain derived neurotrophic factor (BDNF) in young adults: A systematic review and meta-analysis. Sci. Rep. 2021, 11, 8937. [Google Scholar] [CrossRef]
  220. Bakst, R.L.; Glastonbury, C.M.; Parvathaneni, U.; Katabi, N.; Hu, K.S.; Yom, S.S. Perineural Invasion and Perineural Tumor Spread in Head and Neck Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2019, 103, 1109–1124. [Google Scholar] [CrossRef] [Green Version]
  221. Jha, M.K.; Morrison, B.M. Lactate Transporters Mediate Glia-Neuron Metabolic Crosstalk in Homeostasis and Disease. Front. Cell. Neurosci. 2020, 14, 589582. [Google Scholar] [CrossRef]
  222. Lin, H.; Muramatsu, R.; Maedera, N.; Tsunematsu, H.; Hamaguchi, M.; Koyama, Y.; Kuroda, M.; Ono, K.; Sawada, M.; Yamashita, T. Extracellular Lactate Dehydrogenase A Release From Damaged Neurons Drives Central Nervous System Angiogenesis. EBioMedicine 2018, 27, 71–85. [Google Scholar] [CrossRef] [Green Version]
  223. De Oliveira, M.R.; Da Rocha, R.F.; Stertz, L.; Fries, G.R.; De Oliveira, D.L.; Kapczinski, F.; Moreira, J.C.F. Total and mitochondrial nitrosative stress, decreased brain- Derived neurotrophic factor (BDNF) levels and glutamate uptake, and evidence of endoplasmic reticulum stress in the hippocampus of vitamin A-treated rats. Neurochem. Res. 2011, 36, 506–517. [Google Scholar] [CrossRef]
  224. Páez Pereda, M.; Missale, C.; Grübler, Y.; Arzt, E.; Schaaf, L.; Stalla, G.K. Nerve growth factor and retinoic acid inhibit proliferation and invasion in thyroid tumor cells. Mol. Cell. Endocrinol. 2000, 167, 99–106. [Google Scholar] [CrossRef]
  225. Arrieta, O.; Hernández-Pedro, N.; Fernández-González-Aragón, M.C.; Saavedra-Pérez, D.; Campos-Parra, A.D.; Ríos-Trejo, M.A.; Cerón-Lizárraga, T.; Martínez-Barrera, L.; Pineda, B.; Ordóñez, G.; et al. Retinoic acid reduces chemotherapyinduced neuropathy in an animal model and patients with lung cancer. Neurology 2011, 77, 987–995. [Google Scholar] [CrossRef] [PubMed]
  226. Higashi, M.; Kolla, V.; Iyer, R.; Naraparaju, K.; Zhuang, T.; Kolla, S.; Brodeur, G.M. Retinoic acid-induced CHD5 upregulation and neuronal differentiation of neuroblastoma. Mol. Cancer 2015, 14, 150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Wagner, L.M.; Danks, M.K. New therapeutic targets for the treatment of high-risk neuroblastoma. J. Cell. Biochem. 2009, 107, 46–57. [Google Scholar] [CrossRef] [PubMed]
  228. Sweeney, P.; Park, H.; Baumann, M.; Dunlop, J.; Frydman, J.; Kopito, R.; McCampbell, A.; Leblanc, G.; Venkateswaran, A.; Nurmi, A.; et al. Protein misfolding in neurodegenerative diseases: Implications and strategies. Transl. Neurodegener. 2017, 6, 6. [Google Scholar] [CrossRef] [Green Version]
  229. Chaudhuri, O.; Koshy, S.T.; Branco Da Cunha, C.; Shin, J.W.; Verbeke, C.S.; Allison, K.H.; Mooney, D.J. Extracellular matrix stiffness and composition jointly regulate the induction of malignant phenotypes in mammary epithelium. Nat. Mater. 2014, 13, 970–978. [Google Scholar] [CrossRef]
  230. Xu, C.J.; Wang, J.L.; Jin, W.L. The Emerging Therapeutic Role of NGF in Alzheimer’s Disease. Neurochem. Res. 2016, 41, 1211–1218. [Google Scholar] [CrossRef]
  231. Ina, A.; Kamei, Y. Vitamin B12, a chlorophyll-related analog to pheophytin a from marine brown algae, promotes neurite outgrowth and stimulates differentiation in PC12 cells. Cytotechnology 2006, 52, 181–187. [Google Scholar] [CrossRef] [Green Version]
  232. Okada, K.; Tanaka, H.; Temporin, K.; Okamoto, M.; Kuroda, Y.; Moritomo, H.; Murase, T.; Yoshikawa, H. Methylcobalamin increases Erk1/2 and Akt activities through the methylation cycle and promotes nerve regeneration in a rat sciatic nerve injury model. Exp. Neurol. 2010, 222, 191–203. [Google Scholar] [CrossRef]
  233. Segal, R.A. Selectivity in neurotrophin signaling: Theme and variations. Annu. Rev. Neurosci. 2003, 26, 299–330. [Google Scholar] [CrossRef]
  234. Markus, A.; Zhong, J.; Snider, W.D. Raf and Akt mediate distinct aspects of sensory axon growth. Neuron 2002, 35, 65–76. [Google Scholar] [CrossRef] [Green Version]
  235. Okada, K.; Tanaka, H.; Temporin, K.; Okamoto, M.; Kuroda, Y.; Moritomo, H.; Murase, T.; Yoshikawa, H. Akt/mammalian target of rapamycin signaling pathway regulates neurite outgrowth in cerebellar granule neurons stimulated by methylcobalamin. Neurosci. Lett. 2011, 495, 201–204. [Google Scholar] [CrossRef] [PubMed]
  236. Baltrusch, S. The Role of Neurotropic B Vitamins in Nerve Regeneration. Biomed Res. Int. 2021, 2021, 9968228. [Google Scholar] [CrossRef] [PubMed]
  237. Al-saaeed, S.M.; Al-khalisy, M.H. The Regenerative Role of Vitamins B1, B6, B12 in Treatment of Peripheral Neuropathy. Int. J. Sci. Res. 2015, 6, 2319–7064. [Google Scholar] [CrossRef]
  238. Schloss, J.; Colosimo, M. B Vitamin Complex and Chemotherapy-Induced Peripheral Neuropathy. Curr. Oncol. Rep. 2017, 19, 10–14. [Google Scholar] [CrossRef] [PubMed]
  239. Altun, I.; Kurutaş, E.B. Vitamin B complex and vitamin B12 levels after peripheral nerve injury. Neural Regen. Res. 2016, 11, 842–845. [Google Scholar] [CrossRef]
  240. Wrzosek, M.; Lukaszkiewicz, J.; Wrzosek, M.; Jakubczyk, A.; Matsumoto, H.; Piatkiewicz, P.; Radziwon-Zaleska, M.; Wojnar, M.; Nowicka, G. Vitamin D and the central nervous system. Pharmacol. Rep. 2013, 65, 271–278. [Google Scholar] [CrossRef]
  241. Deluca, G.C.; Kimball, S.M.; Kolasinski, J.; Ramagopalan, S.V.; Ebers, G.C. Review: The role of vitamin D in nervous system health and disease. Neuropathol. Appl. Neurobiol. 2013, 39, 458–484. [Google Scholar] [CrossRef]
  242. Annweiler, C.; Schott, A.M.; Berrut, G.; Chauviré, V.; Le Gall, D.; Inzitari, M.; Beauchet, O. Vitamin D and ageing: Neurological issues. Neuropsychobiology 2010, 62, 139–150. [Google Scholar] [CrossRef]
  243. Eyles, D.; Brown, J.; Mackay-Sim, A.; McGrath, J.; Feron, F. Vitamin D3 and brain development. Neuroscience 2003, 118, 641–653. [Google Scholar] [CrossRef]
  244. Sen, C.K.; Khanna, S.; Roy, S. Tocotrienol: The natural vitamin E to defend the nervous system? Ann. N. Y. Acad. Sci. 2004, 1031, 127–142. [Google Scholar] [CrossRef] [PubMed]
  245. Morani, A.S.; Bodhankar, S.L. Neuroprotective effect of vitamin E acetate in models of mononeuropathy in rats. Neuroanatomy 2008, 7, 33–37. [Google Scholar]
  246. Pace, A.; Savarese, A.; Picardo, M.; Maresca, V.; Pacetti, U.; Del Monte, G.; Biroccio, A.; Leonetti, C.; Jandolo, B.; Cognetti, F.; et al. Neuroprotective effect of vitamin E supplementation in patients treated with cisplatin chemotherapy. J. Clin. Oncol. 2003, 21, 927–931. [Google Scholar] [CrossRef] [PubMed]
  247. Tsang, C.K.; Kamei, Y. Novel effect of vitamin K1 (phylloquinone) and vitamin K2 (menaquinone) on promoting nerve growth factor-mediated neurite outgrowth from PC12D cells. Neurosci. Lett. 2002, 323, 9–12. [Google Scholar] [CrossRef]
  248. Graham, D.K.; Deryckere, D.; Davies, K.D.; Earp, H.S. The TAM family: Phosphatidylserine-sensing receptor tyrosine kinases gone awry in cancer. Nat. Rev. Cancer 2014, 14, 769–785. [Google Scholar] [CrossRef]
  249. Shafit-Zagardo, B.; Gruber, R.C.; DuBois, J.C. The role of TAM family receptors and ligands in the nervous system: From development to pathobiology. Pharmacol. Ther. 2018, 188, 97–117. [Google Scholar] [CrossRef]
Figure 1. Complex mechanisms induced by glucose, glutamine, and lactate uptake encompass the metabolism of tumor cells. 3-PG: 3-phosphoglycerate, 3-PH: 3-phosphohydroxypyruvate, ASL: arginosuccinate lyase, ASNS: asparagine synthetase, ASS: arginosuccinate synthase, αKG: alpha-ketoglutarate, GDH: glutamine dehydrogenase, GS: glutamine synthetase, LDH: lactate dehydrogenase, MCT1/4: monocarboxylate transporter 1/4, NOS: nitric oxide synthase, PDH: pyruvate dehydrogenase, PHGDH: phosphoglycerate dehydrogenase, PSAT: phosphoserine aminotransferase, PSPH: phosphoserine phosphatase, ROS: reactive oxygen species, SHMT: serine hydroxymethyltransferase, TXNRD1: thioredoxin reductase 1.
Figure 1. Complex mechanisms induced by glucose, glutamine, and lactate uptake encompass the metabolism of tumor cells. 3-PG: 3-phosphoglycerate, 3-PH: 3-phosphohydroxypyruvate, ASL: arginosuccinate lyase, ASNS: asparagine synthetase, ASS: arginosuccinate synthase, αKG: alpha-ketoglutarate, GDH: glutamine dehydrogenase, GS: glutamine synthetase, LDH: lactate dehydrogenase, MCT1/4: monocarboxylate transporter 1/4, NOS: nitric oxide synthase, PDH: pyruvate dehydrogenase, PHGDH: phosphoglycerate dehydrogenase, PSAT: phosphoserine aminotransferase, PSPH: phosphoserine phosphatase, ROS: reactive oxygen species, SHMT: serine hydroxymethyltransferase, TXNRD1: thioredoxin reductase 1.
Nutrients 14 01722 g001
Figure 2. Metabolism-related cancer–nerve crosstalk. (A) Vitamin C transporter SLC2A3 is shown via immunohistochemistry to be upregulated in colorectal cancer patients with perineural invasion (PNI) [206]. mRNA (B) and immunohistochemistry (C) analyses found that asparagine synthetase (ASNS) is upregulated in PNI-positive oral squamous-cell carcinoma patients. Dotted circles represent nerve trunks, and stars indicate the tumor region [28]. (D) Lactate importer MCT1 is colocalized with Sox2- and KLF-positive (cell-proliferation markers) in cases of PNI in pancreatic adenocarcinoma [205]. Scale is 50 μm. Figures are modified from Gao et al., Fu et al., and Sandforth et al., respectively. Figure rights for reuse are available via the Creative Commons Attribution (CC BY) License.
Figure 2. Metabolism-related cancer–nerve crosstalk. (A) Vitamin C transporter SLC2A3 is shown via immunohistochemistry to be upregulated in colorectal cancer patients with perineural invasion (PNI) [206]. mRNA (B) and immunohistochemistry (C) analyses found that asparagine synthetase (ASNS) is upregulated in PNI-positive oral squamous-cell carcinoma patients. Dotted circles represent nerve trunks, and stars indicate the tumor region [28]. (D) Lactate importer MCT1 is colocalized with Sox2- and KLF-positive (cell-proliferation markers) in cases of PNI in pancreatic adenocarcinoma [205]. Scale is 50 μm. Figures are modified from Gao et al., Fu et al., and Sandforth et al., respectively. Figure rights for reuse are available via the Creative Commons Attribution (CC BY) License.
Nutrients 14 01722 g002
Table 1. Metabolite contribution to tumor survival varies between cancer types.
Table 1. Metabolite contribution to tumor survival varies between cancer types.
MetaboliteCancer TypeRole in Tumor Progression
Vitamin ABreast4-HPR induces cell death [37]; vitamin A and retinol reduce risk [105]
Colon/Colorectal4-HPR induces cell death [37]
Head/Neck4-HPR induces cell death [37]
GastricInhibits polycyclic hydrocarbon-induced carcinomas [37,108]
LungBlood levels of α- and β-carotene, total carotenoids, and retinol are inversely associated with cancer risk [109]
Prostate4-HPR induces cell death [37]
Vitamin B1BreastIntermediate concentrations promote Ehrlich’s ascites proliferation in thiamine-deficient patients; high concentrations inhibit proliferation [110]; patients exhibit decreased expression of SLC9A3 transporter gene [111,112]
Head/NeckPatients exhibit decreased expression of SLC9A3 transporter gene [112]
Intake reduces risk of esophageal cancer [103]
Vitamin B3Head/NeckIntake reduces risk of esophageal cancer [103]
Vitamin B6Head/NeckIntake reduces risk of esophageal cancer [103]
Vitamin B9Head/NeckIntake reduces risk of esophageal cancer [103]
Vitamin B12Head/NeckIntake increases risk of esophageal cancer [103]
Leukemia/LymphomaElevated plasma levels associated with 1-year cancer risk [113]
LiverElevated plasma levels associated with 1-year cancer risk [113]
LungPositively associated with cancer risk in dose-dependent manner [114]
PancreaticElevated plasma levels associated with 1-year cancer risk [113]
Vitamin CBreastLow concentrations induce cell invasiveness; high doses restrict EMT [115]
SkinLow doses reduce cell viability and invasiveness; high doses promote cell migration [116]
Vitamin DBreastCalcitroil and D3 analogs suppress MMP-2 and -9 and VCAM-1; low serum D3 levels are associated with high incidence [117]
Colon/ColorectalLow serum D3 levels are associated with high incidence [117]; analog PRI-2191 enhances ability of 5-FU to restrict cell cycle [118]; serum levels of ≥33 ng/mL correlates with a 50% decreased risk [119]
GastricLow serum D3 levels associated with high incidence [117]
Head/NeckMART-10 induces cell-cycle arrest and suppresses p21 and p27 [120]
ProstateLower serum levels are associated with an increased risk; D3 and analogs inhibit invasiveness and expression of MMP-2 and -9 and VCAM-1 [117]
Vitamin EBreastTocotrienols exhibit chemotherapeutic and antitumor properties [41]; γ-tocotrienol induces cell-cycle arrest [121,122]
Colon/ColorectalTocotrienols exhibit antitumor properties [41]; γ-tocotrienol mediates apoptosis via apoptotic caspase-3 and NFκB suppression [123]
LiverTocotrienols exhibit chemotherapeutic properties [41]
LungTocotrienols exhibit chemotherapeutic properties [41]
PancreaticTocotrienols exhibit chemotherapeutic properties [41]
ProstateTocotrienols exhibit chemotherapeutic properties [41]
Vitamin KBreastK2 induces nonapoptotic cell death [42]
ArginineBreastLow plasma levels act as a prognostic biomarker [124]; arginine starvation is used to treat arginosuccinate synthase-deficient patients [101,124]
OvarianCancer cells are deficient in arginosuccinate synthase-1; ADI-PED-20 is used to degrade arginine [125]
ProstateLow plasma levels act as a prognostic biomarker [126]
SkinCells are deficient in arginosuccinate synthase-1; ADI-PEG20-resistant cancer exhibits c-MYC binding to the promoter of arginosuccinate synthase-1 [26]
AsparagineBreastMaintains health of glutamine-independent cells [96]
CervicalFacilitates mTOR activation in the absence of glutamine [96]
LiposarcomaMaintains health of glutamine-independent cells [96]
CysteineBreastInhibition of histone deacetylase-6 sensitizes TNBC cells to cysteine deprivation via cystine/glutamate antiporter-targeted therapies [98]
Colon/ColorectalStarvation induces a reduction in liver-metastatic cell proliferation [29]
PancreaticThe deletion of cystine transporter gene SLC7A11 inhibits autophagy and diminishes cysteine homeostasis [127]
GlycineColon/ColorectalMetabolism increases when starved of serine [128]; serine–glycine inhibition should be used in conjunction to decrease tumor size [102]
LungDe novo serine and glycine are allocated to glutathione synthesis [128]
LactateBreast10 mM L-lactate acts as chemoattractant and facilitates migration [129]; intermediate and high supplementation upregulates oncogene, proliferation gene, tumor suppressor, and transcription-factor expression [130];
CervicalWhen given glucose and lactate, oxidative cancer cells prefer lactate; cells thrive when given lactate supplementation; oxidative cells exhibit high expression of MCT1 versus MCT4; MCT1 inhibition induces necrosis in oxidative cells [25]; DLAD targets metabolism [131]
Colon/ColorectalGlycolytic cells fail to thrive upon glucose starvation with lactate supplementation [25]
Head/NeckDLAD targets metabolism [131]
LungNFκB signaling [132]; LDH-A inhibition sensitizes cells to radiation [133]
SkinDLAD targets metabolism [131]
SerineBreastCells prefer serine over glycine and exhibit a decrease in nucleic acid synthesis when starved of serine [99]; brain metastatic cells upregulate de novo serine when starved of exogenous alternative [134]
Colon/ColorectalCells prefer serine over glycine and exhibit a decrease in nucleic acid synthesis when starved of serine [99]; metabolism increases when starved of serine; starvation decreases YAP activation [135]; serine–glycine catabolism induced by stress promotes formate production [136]; serine–glycine inhibition should be used in conjunction to decrease tumor size; starvation induces metabolic stress and p53-activated glycolysis [102]
LungPromotes purine synthesis in cancer cells; de novo serine and glycine are allocated to glutathione synthesis [128]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Gu, I.; Gregory, E.; Atwood, C.; Lee, S.-O.; Song, Y.H. Exploring the Role of Metabolites in Cancer and the Associated Nerve Crosstalk. Nutrients 2022, 14, 1722. https://doi.org/10.3390/nu14091722

AMA Style

Gu I, Gregory E, Atwood C, Lee S-O, Song YH. Exploring the Role of Metabolites in Cancer and the Associated Nerve Crosstalk. Nutrients. 2022; 14(9):1722. https://doi.org/10.3390/nu14091722

Chicago/Turabian Style

Gu, Inah, Emory Gregory, Casey Atwood, Sun-Ok Lee, and Young Hye Song. 2022. "Exploring the Role of Metabolites in Cancer and the Associated Nerve Crosstalk" Nutrients 14, no. 9: 1722. https://doi.org/10.3390/nu14091722

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop